1
|
Liu D, Lu N, Zang F, Lu M, Zhang J, Zhao Y, Wan H, Wang M, Li QQ, Wang F, Luo S, Ma M, Shi F, Wu H, Tu J, Zhang Y. Magnetic Resonance Imaging-Based Radiogenomic Analysis Reveals Genomic Determinants for Nanoparticle Delivery into Tumors. ACS NANO 2024; 18:34615-34629. [PMID: 39663893 DOI: 10.1021/acsnano.4c09387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Even though the enhanced permeability and retention (EPR) effect is applicable for the passive targeting of solid tumors, many nanodrugs have failed to achieve meaningful clinical outcomes due to the heterogeneity of EPR effect. Therefore, understanding the mechanism of the EPR effect is crucial to overcome the obstacles nanomedicines face in clinical translation. The aim of this study was to establish a reliable method to increase awareness of the critical influencing factors of nanoparticle (NP) transport into tumors based on the EPR effect using a combined radiogenomics and clinical magnetic resonance imaging (MRI) technique and gene set pathway enrichment analysis. Employing poly(lactic-co-glycolic acid) (PLGA)-coated Fe3O4 NPs as the contrast agent, the monolayer and multilayer distribution of the NPs were observed and quantitatively analyzed by MRI, improving the accuracy of evaluating vascular permeability by MRI. By performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of genes and pathways, we identified a variety of genes affecting vascular permeability, such as Cldn1, Dlg2, Bves, Prkag3, Cldn10, and Cldn8, which are related to tight junctions and control the permeability of blood vessels in tumors. The method presented here provides an MRI-supported approach to increase the breadth of data collected from genetic screens, reveals genetic evidence of the presence of NPs in tumors and lays a foundation for clinical patient stratification and personalized treatment.
Collapse
Affiliation(s)
- Di Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Na Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fengchao Zang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Mingze Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jingyue Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Hao Wan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Mengjun Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Qian-Qian Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Shouhua Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ming Ma
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Haoan Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| |
Collapse
|
2
|
Ren L, Chen DB, Yan X, She S, Yang Y, Zhang X, Liao W, Chen H. Bridging the Gap Between Imaging and Molecular Characterization: Current Understanding of Radiomics and Radiogenomics in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:2359-2372. [PMID: 39619602 PMCID: PMC11608547 DOI: 10.2147/jhc.s423549] [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: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide and the third leading cause of cancer-related deaths. Imaging plays a crucial role in the screening, diagnosis, and monitoring of HCC; however, the potential mechanism regarding phenotypes or molecular subtyping remains underexplored. Radiomics significantly expands the selection of features available by extracting quantitative features from imaging data. Radiogenomics bridges the gap between imaging and genetic/transcriptomic information by associating imaging features with critical genes and pathways, thereby providing biological annotations to these features. Despite challenges in interpreting these connections, assessing their universality, and considering the diversity in HCC etiology and genetic information across different populations, radiomics and radiogenomics offer new perspectives for precision treatment in HCC. This article provides an up-to-date summary of the advancements in radiomics and radiogenomics throughout the HCC care continuum, focusing on the clinical applications, advantages, and limitations of current techniques and offering prospects. Future research should aim to overcome these challenges to improve the prognosis of HCC patients and leverage imaging information for patient benefit.
Collapse
Affiliation(s)
- Liying Ren
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Dong Bo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xuanzhi Yan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Shaoping She
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Yao Yang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xue Zhang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Hongsong Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| |
Collapse
|
3
|
Qi YJ, Su GH, You C, Zhang X, Xiao Y, Jiang YZ, Shao ZM. Radiomics in breast cancer: Current advances and future directions. Cell Rep Med 2024; 5:101719. [PMID: 39293402 PMCID: PMC11528234 DOI: 10.1016/j.xcrm.2024.101719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
Breast cancer is a common disease that causes great health concerns to women worldwide. During the diagnosis and treatment of breast cancer, medical imaging plays an essential role, but its interpretation relies on radiologists or clinical doctors. Radiomics can extract high-throughput quantitative imaging features from images of various modalities via traditional machine learning or deep learning methods following a series of standard processes. Hopefully, radiomic models may aid various processes in clinical practice. In this review, we summarize the current utilization of radiomics for predicting clinicopathological indices and clinical outcomes. We also focus on radio-multi-omics studies that bridge the gap between phenotypic and microscopic scale information. Acknowledging the deficiencies that currently hinder the clinical adoption of radiomic models, we discuss the underlying causes of this situation and propose future directions for advancing radiomics in breast cancer research.
Collapse
Affiliation(s)
- Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
You C, Su GH, Zhang X, Xiao Y, Zheng RC, Sun SY, Zhou JY, Lin LY, Wang ZZ, Wang H, Chen Y, Peng WJ, Jiang YZ, Shao ZM, Gu YJ. Multicenter radio-multiomic analysis for predicting breast cancer outcome and unravelling imaging-biological connection. NPJ Precis Oncol 2024; 8:193. [PMID: 39244594 PMCID: PMC11380684 DOI: 10.1038/s41698-024-00666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/24/2024] [Indexed: 09/09/2024] Open
Abstract
Radiomics offers a noninvasive avenue for predicting clinicopathological factors. However, thorough investigations into a robust breast cancer outcome-predicting model and its biological significance remain limited. This study develops a robust radiomic model for prognosis prediction, and further excavates its biological foundation and transferring prediction performance. We retrospectively collected preoperative dynamic contrast-enhanced MRI data from three distinct breast cancer patient cohorts. In FUSCC cohort (n = 466), Lasso was used to select features correlated with patient prognosis and multivariate Cox regression was utilized to integrate these features and build the radiomic risk model, while multiomic analysis was conducted to investigate the model's biological implications. DUKE cohort (n = 619) and I-SPY1 cohort (n = 128) were used to test the performance of the radiomic signature in outcome prediction. A thirteen-feature radiomic signature was identified in the FUSCC cohort training set and validated in the FUSCC cohort testing set, DUKE cohort and I-SPY1 cohort for predicting relapse-free survival (RFS) and overall survival (OS) (RFS: p = 0.013, p = 0.024 and p = 0.035; OS: p = 0.036, p = 0.005 and p = 0.027 in the three cohorts). Multiomic analysis uncovered metabolic dysregulation underlying the radiomic signature (ATP metabolic process: NES = 1.84, p-adjust = 0.02; cholesterol biosynthesis: NES = 1.79, p-adjust = 0.01). Regarding the therapeutic implications, the radiomic signature exhibited value when combining clinical factors for predicting the pathological complete response to neoadjuvant chemotherapy (DUKE cohort, AUC = 0.72; I-SPY1 cohort, AUC = 0.73). In conclusion, our study identified a breast cancer outcome-predicting radiomic signature in a multicenter radio-multiomic study, along with its correlations with multiomic features in prognostic risk assessment, laying the groundwork for future prospective clinical trials in personalized risk stratification and precision therapy.
Collapse
Affiliation(s)
- Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ren-Cheng Zheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Yun Sun
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Yin Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu-Yi Lin
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Zhou Wang
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Yan Chen
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Wei-Jun Peng
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Wang G, Ding F, Chen K, Liang Z, Han P, Wang L, Cui F, Zhu Q, Cheng Z, Chen X, Huang C, Cheng H, Wang X, Zhao X. CT-based radiomics nomogram to predict proliferative hepatocellular carcinoma and explore the tumor microenvironment. J Transl Med 2024; 22:683. [PMID: 39218938 PMCID: PMC11367757 DOI: 10.1186/s12967-024-05393-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Proliferative hepatocellular carcinomas (HCCs) is a class of aggressive tumors with poor prognosis. We aimed to construct a computed tomography (CT)-based radiomics nomogram to predict proliferative HCC, stratify clinical outcomes and explore the tumor microenvironment. METHODS Patients with pathologically diagnosed HCC following a hepatectomy were retrospectively collected from two medical centers. A CT-based radiomics nomogram incorporating radiomics model and clinicoradiological features to predict proliferative HCC was constructed using the training cohort (n = 184), and validated using an internal test cohort (n = 80) and an external test cohort (n = 89). The predictive performance of the nomogram for clinical outcomes was evaluated for HCC patients who underwent surgery (n = 201) or received transarterial chemoembolization (TACE, n = 104). RNA sequencing data and histological tissue slides from The Cancer Imaging Archive database were used to perform transcriptomics and pathomics analysis. RESULTS The areas under the receiver operating characteristic curve of the radiomics nomogram to predict proliferative HCC were 0.84, 0.87, and 0.85 in the training, internal test, and external test cohorts, respectively. The radiomics nomogram could stratify early recurrence-free survivals in the surgery outcome cohort (hazard ratio [HR] = 2.25; P < 0.001) and progression-free survivals in the TACE outcome cohort (HR = 2.21; P = 0.03). Transcriptomics and pathomics analysis indicated that the radiomics nomogram was associated with carbon metabolism, immune cells infiltration, TP53 mutation, and heterogeneity of tumor cells. CONCLUSION The CT-based radiomics nomogram could predict proliferative HCC, stratify clinical outcomes, and measure a pro-tumor microenvironment.
Collapse
Affiliation(s)
- Gongzheng Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Feier Ding
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Kaige Chen
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhuoshuai Liang
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Pengxi Han
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Linxiang Wang
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Fengyun Cui
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Qiang Zhu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhaoping Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Xingzhi Chen
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, 100080, People's Republic of China
| | - Chencui Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, 100080, People's Republic of China
| | - Hongxia Cheng
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| | - Xinya Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
6
|
Cheng M, Zhang H, Huang W, Li F, Gao J. Deep Learning Radiomics Analysis of CT Imaging for Differentiating Between Crohn's Disease and Intestinal Tuberculosis. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1516-1528. [PMID: 38424279 PMCID: PMC11300798 DOI: 10.1007/s10278-024-01059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
This study aimed to develop and evaluate a CT-based deep learning radiomics model for differentiating between Crohn's disease (CD) and intestinal tuberculosis (ITB). A total of 330 patients with pathologically confirmed as CD or ITB from the First Affiliated Hospital of Zhengzhou University were divided into the validation dataset one (CD: 167; ITB: 57) and validation dataset two (CD: 78; ITB: 28). Based on the validation dataset one, the synthetic minority oversampling technique (SMOTE) was adopted to create balanced dataset as training data for feature selection and model construction. The handcrafted and deep learning (DL) radiomics features were extracted from the arterial and venous phases images, respectively. The interobserver consistency analysis, Spearman's correlation, univariate analysis, and the least absolute shrinkage and selection operator (LASSO) regression were used to select features. Based on extracted multi-phase radiomics features, six logistic regression models were finally constructed. The diagnostic performances of different models were compared using ROC analysis and Delong test. The arterial-venous combined deep learning radiomics model for differentiating between CD and ITB showed a high prediction quality with AUCs of 0.885, 0.877, and 0.800 in SMOTE dataset, validation dataset one, and validation dataset two, respectively. Moreover, the deep learning radiomics model outperformed the handcrafted radiomics model in same phase images. In validation dataset one, the Delong test results indicated that there was a significant difference in the AUC of the arterial models (p = 0.037), while not in venous and arterial-venous combined models (p = 0.398 and p = 0.265) as comparing deep learning radiomics models and handcrafted radiomics models. In our study, the arterial-venous combined model based on deep learning radiomics analysis exhibited good performance in differentiating between CD and ITB.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Medical Information, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Hanyue Zhang
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Fei Li
- School of Cyber Science and Engineering, Wuhan University, Wuhan, 430072, China
| | - Jianbo Gao
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
7
|
Wang C, Wu F, Wang F, Chong HH, Sun H, Huang P, Xiao Y, Yang C, Zeng M. The Association Between Tumor Radiomic Analysis and Peritumor Habitat-Derived Radiomic Analysis on Gadoxetate Disodium-Enhanced MRI With Microvascular Invasion in Hepatocellular Carcinoma. J Magn Reson Imaging 2024. [PMID: 38997242 DOI: 10.1002/jmri.29523] [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: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has a poor prognosis, often characterized by microvascular invasion (MVI). Radiomics and habitat imaging offer potential for preoperative MVI assessment. PURPOSE To identify MVI in HCC by habitat imaging, tumor radiomic analysis, and peritumor habitat-derived radiomic analysis. STUDY TYPE Retrospective. SUBJECTS Three hundred eighteen patients (53 ± 11.42 years old; male = 276) with pathologically confirmed HCC (training:testing = 224:94). FIELD STRENGTH/SEQUENCE 1.5 T, T2WI (spin echo), and precontrast and dynamic T1WI using three-dimensional gradient echo sequence. ASSESSMENT Clinical model, habitat model, single sequence radiomic models, the peritumor habitat-derived radiomic model, and the combined models were constructed for evaluating MVI. Follow-up clinical data were obtained by a review of medical records or telephone interviews. STATISTICAL TESTS Univariable and multivariable logistic regression, receiver operating characteristic (ROC) curve, calibration, decision curve, Delong test, K-M curves, log rank test. A P-value less than 0.05 (two sides) was considered to indicate statistical significance. RESULTS Habitat imaging revealed a positive correlation between the number of subregions and MVI probability. The Radiomic-Pre model demonstrated AUCs of 0.815 (95% CI: 0.752-0.878) and 0.708 (95% CI: 0.599-0.817) for detecting MVI in the training and testing cohorts, respectively. Similarly, the AUCs for MVI detection using Radiomic-HBP were 0.790 (95% CI: 0.724-0.855) for the training cohort and 0.712 (95% CI: 0.604-0.820) for the test cohort. Combination models exhibited improved performance, with the Radiomics + Habitat + Dilation + Habitat 2 + Clinical Model (Model 7) achieving the higher AUC than Model 1-4 and 6 (0.825 vs. 0.688, 0.726, 0.785, 0.757, 0.804, P = 0.013, 0.048, 0.035, 0.041, 0.039, respectively) in the testing cohort. High-risk patients (cutoff value >0.11) identified by this model showed shorter recurrence-free survival. DATA CONCLUSION The combined model including tumor size, habitat imaging, radiomic analysis exhibited the best performance in predicting MVI, while also assessing prognostic risk. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Cheng Wang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei Wu
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang Wang
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Huan-Huan Chong
- Department of Radiology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Haitao Sun
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Huang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyao Xiao
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Yang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengsu Zeng
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Wang D, Liu S, Fu J, Zhang P, Zheng S, Qiu B, Liu H, Ye Y, Guo J, Zhou Y, Jiang H, Yin S, He H, Xie C, Liu H. Correlation of K trans derived from dynamic contrast-enhanced MRI with treatment response and survival in locally advanced NSCLC patients undergoing induction immunochemotherapy and concurrent chemoradiotherapy. J Immunother Cancer 2024; 12:e008574. [PMID: 38910009 PMCID: PMC11328668 DOI: 10.1136/jitc-2023-008574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
PURPOSE This study aimed to investigate the prognostic significance of pretreatment dynamic contrast-enhanced (DCE)-MRI parameters concerning tumor response following induction immunochemotherapy and survival outcomes in patients with locally advanced non-small cell lung cancer (NSCLC) who underwent immunotherapy-based multimodal treatments. MATERIAL AND METHODS Unresectable stage III NSCLC patients treated by induction immunochemotherapy, concurrent chemoradiotherapy (CCRT) with or without consolidative immunotherapy from two prospective clinical trials were screened. Using the two-compartment Extend Tofts model, the parameters including Ktrans, Kep, Ve, and Vp were calculated from DCE-MRI data. The apparent diffusion coefficient was calculated from diffusion-weighted-MRI data. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to assess the predictive performance of MRI parameters. The Cox regression model was used for univariate and multivariate analysis. RESULTS 111 unresectable stage III NSCLC patients were enrolled. Patients received two cycles of induction immunochemotherapy and CCRT, with or without consolidative immunotherapy. With the median follow-up of 22.3 months, the median progression-free survival (PFS) and overall survival (OS) were 16.3 and 23.8 months. The multivariate analysis suggested that Eastern Cooperative Oncology Group score, TNM stage and the response to induction immunochemotherapy were significantly related to both PFS and OS. After induction immunochemotherapy, 67 patients (59.8%) achieved complete response or partial response and 44 patients (40.2%) had stable disease or progressive disease. The Ktrans of primary lung tumor before induction immunochemotherapy yielded the best performance in predicting the treatment response, with an AUC of 0.800. Patients were categorized into two groups: high-Ktrans group (n=67, Ktrans>164.3×10-3/min) and low-Ktrans group (n=44, Ktrans≤164.3×10-3/min) based on the ROC analysis. The high-Ktrans group had a significantly higher objective response rate than the low-Ktrans group (85.1% (57/67) vs 22.7% (10/44), p<0.001). The high-Ktrans group also presented better PFS (median: 21.1 vs 11.3 months, p=0.002) and OS (median: 34.3 vs 15.6 months, p=0.035) than the low-Ktrans group. CONCLUSIONS Pretreatment Ktrans value emerged as a significant predictor of the early response to induction immunochemotherapy and survival outcomes in unresectable stage III NSCLC patients who underwent immunotherapy-based multimodal treatments. Elevated Ktrans values correlated positively with enhanced treatment response, leading to extended PFS and OS durations.
Collapse
Affiliation(s)
- DaQuan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - SongRan Liu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Jia Fu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - PengXin Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ShiYang Zheng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- United Imaging Healthcare, ShangHai, China
| | - YongQuan Ye
- United Imaging of Healthcare America, Houston, Texas, USA
| | - JinYu Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Yin Zhou
- SuZhou TongDiao Company, Suzhou, China
| | | | - ShaoHan Yin
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - HaoQiang He
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ChuanMiao Xie
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Liu K, Jiang T, Rao W, Chen B, Yin X, Xu P, Hu S. Peptidic heterodimer-based radiotracer targeting fibroblast activation protein and integrin α vβ 3. Eur J Nucl Med Mol Imaging 2024; 51:1544-1557. [PMID: 38276986 DOI: 10.1007/s00259-024-06623-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/20/2024] [Indexed: 01/27/2024]
Abstract
PURPOSE Several studies have demonstrated the advantages of heterodimers over their corresponding monomers due to the multivalency effect. This effect leads to an increased number of effective targeted receptors and, consequently, improved tumor uptake. Fibroblast activation protein (FAP) and integrin αvβ3 are found to be overexpressed in different components of the tumor microenvironment. In our pursuit of enhancing tumor uptake and retention, we designed and developed a novel peptidic heterodimer that synergistically targets both FAP and integrin αvβ3. METHODS FAP-RGD was synthesized from FAP-2286 and c(RGDfK) through a multi-step organic synthesis. The dual receptor binding property of 68Ga-FAP-RGD was investigated by cell uptake and competitive binding assays. Preclinical pharmacokinetics were determined in HT1080-FAP and U87MG tumor models using micro-positron emission tomography/computed tomography (micro-PET/CT) and biodistribution studies. The antitumor efficacy of 177Lu-FAP-RGD was assessed in U87MG tumor models. The radiation exposure and clinical diagnostic performance of 68 Ga-FAP-RGD were evaluated in healthy volunteers and cancer patients. RESULTS Bi-specific radiotracer 68Ga-FAP-RGD exhibited high binding affinity for both FAP and integrin αvβ3. In comparison to 68Ga-FAP-2286 and 68Ga-RGDfK, 68Ga-FAP-RGD displayed enhanced tumor uptake and longer tumor retention time in preclinical models. 177Lu-FAP-RGD could efficiently suppress the growth of U87MG tumor in vivo when applied at an activity of 18.5 and 29.6 MBq. The effective dose of 68Ga-FAP-RGD was 1.06 × 10-2 mSv/MBq. 68Ga-FAP-RGD demonstrated low background activity and stable accumulation in most neoplastic lesions up to 3 h. CONCLUSION Taking the advantages of multivalency effect, the bi-specific radiotracer 68Ga-FAP-RGD showed superior tumor uptake and retention compared to its corresponding monomers. Preclinical studies with 68Ga- or 177Lu-labeled FAP-RGD showed favorable image contrast and effective antitumor responses. Despite the excellent performance of 68Ga-FAP-RGD in clinical diagnosis, experimental efforts are currently underway to optimize the structure of FAP-RGD to increase its potential for clinical application in endoradiotherapy.
Collapse
Affiliation(s)
- Kehuang Liu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China
| | - Tao Jiang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China
| | - Wanqian Rao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China
| | - Pengfei Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha City, 410008, Hunan Province, China.
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Changsha City, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha City, 410008, Hunan Province, China.
| |
Collapse
|
10
|
Lei Y, Feng B, Wan M, Xu K, Cui J, Ma C, Sun J, Yao C, Gan S, Shi J, Cui E. Predicting microvascular invasion in hepatocellular carcinoma with a CT- and MRI-based multimodal deep learning model. Abdom Radiol (NY) 2024; 49:1397-1410. [PMID: 38433144 DOI: 10.1007/s00261-024-04202-1] [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: 10/29/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE To investigate the value of a multimodal deep learning (MDL) model based on computed tomography (CT) and magnetic resonance imaging (MRI) for predicting microvascular invasion (MVI) in hepatocellular carcinoma (HCC). METHODS A total of 287 patients with HCC from our institution and 58 patients from another individual institution were included. Among these, 119 patients with only CT data and 116 patients with only MRI data were selected for single-modality deep learning model development, after which select parameters were migrated for MDL model development with transfer learning (TL). In addition, 110 patients with simultaneous CT and MRI data were divided into a training cohort (n = 66) and a validation cohort (n = 44). We input the features extracted from DenseNet121 into an extreme learning machine (ELM) classifier to construct a classification model. RESULTS The area under the curve (AUC) of the MDL model was 0.844, which was superior to that of the single-phase CT (AUC = 0.706-0.776, P < 0.05), single-sequence MRI (AUC = 0.706-0.717, P < 0.05), single-modality DL model (AUCall-phase CT = 0.722, AUCall-sequence MRI = 0.731; P < 0.05), clinical (AUC = 0.648, P < 0.05), but not to that of the delay phase (DP) and in-phase (IP) MRI and portal venous phase (PVP) CT models. The MDL model achieved better performance than models described above (P < 0.05). When combined with clinical features, the AUC of the MDL model increased from 0.844 to 0.871. A nomogram, combining deep learning signatures (DLS) and clinical indicators for MDL models, demonstrated a greater overall net gain than the MDL models (P < 0.05). CONCLUSION The MDL model is a valuable noninvasive technique for preoperatively predicting MVI in HCC.
Collapse
Affiliation(s)
- Yan Lei
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
- Zunyi Medical University, 1 Xiaoyuan Road, Zunyi, People's Republic of China
| | - Bao Feng
- Laboratory of Intelligent Detection and Information Processing, School of Electronic Information and Automation, Guilin University of Aerospace Technology, 2 Jinji Road, Guilin, People's Republic of China
| | - Meiqi Wan
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
- Zunyi Medical University, 1 Xiaoyuan Road, Zunyi, People's Republic of China
| | - Kuncai Xu
- Laboratory of Intelligent Detection and Information Processing, School of Electronic Information and Automation, Guilin University of Aerospace Technology, 2 Jinji Road, Guilin, People's Republic of China
| | - Jin Cui
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
| | - Changyi Ma
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
| | - Junqi Sun
- Department of Radiology, Yuebei People's Hospital, 133 Huimin Street, Shaoguan, People's Republic of China
| | - Changyin Yao
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
- Guangdong Medical University, 2 Wenming East Road, Zhanjiang, People's Republic of China
| | - Shiman Gan
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China
- Guangdong Medical University, 2 Wenming East Road, Zhanjiang, People's Republic of China
| | - Jiangfeng Shi
- Laboratory of Intelligent Detection and Information Processing, School of Electronic Information and Automation, Guilin University of Aerospace Technology, 2 Jinji Road, Guilin, People's Republic of China
| | - Enming Cui
- Department of Radiology, Jiangmen Central Hospital, 23 Beijie Haibang Street, Jiangmen, People's Republic of China.
- Zunyi Medical University, 1 Xiaoyuan Road, Zunyi, People's Republic of China.
- Guangdong Medical University, 2 Wenming East Road, Zhanjiang, People's Republic of China.
- Jiangmen Key Laboratory of Artificial Intelligence in Medical Image Computation and Application, 23 Beijie Haibang Street, Jiangmen, People's Republic of China.
| |
Collapse
|
11
|
Sujit SJ, Aminu M, Karpinets TV, Chen P, Saad MB, Salehjahromi M, Boom JD, Qayati M, George JM, Allen H, Antonoff MB, Hong L, Hu X, Heeke S, Tran HT, Le X, Elamin YY, Altan M, Vokes NI, Sheshadri A, Lin J, Zhang J, Lu Y, Behrens C, Godoy MCB, Wu CC, Chang JY, Chung C, Jaffray DA, Wistuba II, Lee JJ, Vaporciyan AA, Gibbons DL, Heymach J, Zhang J, Cascone T, Wu J. Enhancing NSCLC recurrence prediction with PET/CT habitat imaging, ctDNA, and integrative radiogenomics-blood insights. Nat Commun 2024; 15:3152. [PMID: 38605064 PMCID: PMC11009351 DOI: 10.1038/s41467-024-47512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
While we recognize the prognostic importance of clinicopathological measures and circulating tumor DNA (ctDNA), the independent contribution of quantitative image markers to prognosis in non-small cell lung cancer (NSCLC) remains underexplored. In our multi-institutional study of 394 NSCLC patients, we utilize pre-treatment computed tomography (CT) and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) to establish a habitat imaging framework for assessing regional heterogeneity within individual tumors. This framework identifies three PET/CT subtypes, which maintain prognostic value after adjusting for clinicopathologic risk factors including tumor volume. Additionally, these subtypes complement ctDNA in predicting disease recurrence. Radiogenomics analysis unveil the molecular underpinnings of these imaging subtypes, highlighting downregulation in interferon alpha and gamma pathways in the high-risk subtype. In summary, our study demonstrates that these habitat imaging subtypes effectively stratify NSCLC patients based on their risk levels for disease recurrence after initial curative surgery or radiotherapy, providing valuable insights for personalized treatment approaches.
Collapse
Affiliation(s)
- Sheeba J Sujit
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tatiana V Karpinets
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maliazurina B Saad
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Morteza Salehjahromi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John D Boom
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mohamed Qayati
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M George
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Haley Allen
- Natural Sciences, Rice University, Houston, TX, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingzhi Hong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Hu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hai T Tran
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie Lin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Jaffray
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Genomics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Interception Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Zhang Y, Yang Y, Ma Y, Liu Y, Ye Z. Development and validation of an interpretable radiomic signature for preoperative estimation of tumor mutational burden in lung adenocarcinoma. Front Genet 2024; 15:1367434. [PMID: 38660677 PMCID: PMC11039798 DOI: 10.3389/fgene.2024.1367434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
Background Tumor mutational burden (TMB) is a promising biomarker for immunotherapy. The challenge of spatial and temporal heterogeneity and high costs weaken its power in clinical routine. The aim of this study is to estimate TMB preoperatively using a volumetric CT-based radiomic signature (rMB). Methods Seventy-one patients with resectable lung adenocarcinoma (LUAD) who underwent whole-exome sequencing (WXS) from 2011 to 2014 were enrolled from the institutional biobank of Tianjin Medical University Cancer Institute and Hospital (TMUCIH). Forty-nine LUAD patients with WXS from the Cancer Genome Atlas Program (TCGA) served as the external validation cohort. Computed tomography (CT) volumes were resampled to 1-mm isotropic, semi-automatically segmented, and manually adjusted by two radiologists. A total of 3,108 radiomic features were extracted via PyRadiomics and then harmonized across cohorts by ComBat. Features with inter-segmentation intra-class correlation coefficient (ICC) > 0.8, low collinearity, and significant univariate power were passed to the least absolute shrinkage and selection operator (LASSO)-logistic classifier to discriminate TMB-high/TMB-low at a threshold of 10 mut/Mb. The receiver operating characteristic (ROC) curve analysis and calibration curve were used to determine its efficiency. Shapley values (SHAP) attributed individual predictions to feature contributions. Clinical variables and circulating biomarkers were collected to find potential associations with TMB and rMB. Results The top frequently mutated genes significantly differed between the Chinese and TCGA cohorts, with a median TMB of 2.20 and 3.46 mut/Mb and 15 (21.12%) and 9 (18.37%) cases of TMB-high, respectively. After dimensionality reduction, rMB comprised 21 features, which reached an AUC of 0.895 (sensitivity = 0.867, specificity = 0.875, and accuracy = 0.873) in the discovery cohort and 0.878 (sensitivity = 1.0, specificity = 0.825, and accuracy = 0.857 in a consist cutoff) in the validation cohort. rMB of TMB-high patients was significantly higher than rMB of TMB-low patients in both cohorts (p < 0.01). rMB was well-calibrated in the discovery cohort and validation cohort (p = 0.27 and 0.74, respectively). The square-filtered gray-level concurrence matrix (GLCM) correlation was of significant importance in prediction. The proportion of circulating monocytes and the monocyte-to-lymphocyte ratio were associated with TMB, whereas the circulating neutrophils and lymphocyte percentage, original and derived neutrophil-to-lymphocyte ratio, and platelet-to-lymphocyte ratio were associated with rMB. Conclusion rMB, an intra-tumor radiomic signature, could predict lung adenocarcinoma patients with higher TMB. Insights from the Shapley values may enhance persuasiveness of the purposed signature for further clinical application. rMB could become a promising tool to triage patients who might benefit from a next-generation sequencing test.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Key Laboratory of Cancer Immunology and Biotherapy of Tianjin, Tianjin, China
| | - Yichen Yang
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, China
| | - Yue Ma
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Key Laboratory of Cancer Immunology and Biotherapy of Tianjin, Tianjin, China
| | - Ying Liu
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Key Laboratory of Cancer Immunology and Biotherapy of Tianjin, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Key Laboratory of Cancer Immunology and Biotherapy of Tianjin, Tianjin, China
| |
Collapse
|
13
|
Yu C, Ruan Y, Yu L, Wang X, Hu Z, Zhu G, Huang T. Predicting postoperative prognosis of pancreatic cancer using a computed tomography-based radio-clinical model: exploring biologic functions. J Gastrointest Surg 2024; 28:458-466. [PMID: 38583896 DOI: 10.1016/j.gassur.2024.02.005] [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: 10/31/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 04/09/2024]
Abstract
Computed tomography (CT) imaging has the potential to assist in predicting the prognosis and treatment strategies for pancreatic cancer (PC). This study aimed to develop and validate a radio-clinical model based on preoperative multiphase CT assessments to predict the overall survival (OS) of PC and identify differentially expressed genes associated with OS. METHODS Patients with PC who had undergone radical pancreatectomy (R0 resection) were divided into development and external validation sets. Independent predictors of OS were identified using Cox regression analyses and included in the nomogram, which was externally validated. The area under the curve was used to measure the model's accuracy in estimating OS probability. RNA sequencing data from The Cancer Genome Atlas were used for gene expression analysis. RESULTS In the development and external validation sets, survival was estimated respectively for 132 and 27 patients. Multivariate Cox regression analysis identified 5 independent OS predictors: age (P = .049), sex (P = .001), bilirubin level (P = .005), tumor size (P = .020), and venous invasion (P = .041). These variables were incorporated into the nomogram. Patients were divided into high- and low-risk groups for OS and survival curves showed that all patients in the low-risk group had better OS than that of those in the high-risk group (P < .001). Differentially expressed genes in patients with a poor prognosis were involved in neuroactive ligand-receptor interaction. CONCLUSION The radio-clinical model may be clinically useful for successfully predicting PC prognosis.
Collapse
Affiliation(s)
- Can Yu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lan Yu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinxin Wang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoshen Hu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanyu Zhu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tao Huang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
14
|
Salehjahromi M, Karpinets TV, Sujit SJ, Qayati M, Chen P, Aminu M, Saad MB, Bandyopadhyay R, Hong L, Sheshadri A, Lin J, Antonoff MB, Sepesi B, Ostrin EJ, Toumazis I, Huang P, Cheng C, Cascone T, Vokes NI, Behrens C, Siewerdsen JH, Hazle JD, Chang JY, Zhang J, Lu Y, Godoy MCB, Chung C, Jaffray D, Wistuba I, Lee JJ, Vaporciyan AA, Gibbons DL, Gladish G, Heymach JV, Wu CC, Zhang J, Wu J. Synthetic PET from CT improves diagnosis and prognosis for lung cancer: Proof of concept. Cell Rep Med 2024; 5:101463. [PMID: 38471502 PMCID: PMC10983039 DOI: 10.1016/j.xcrm.2024.101463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/07/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
[18F]Fluorodeoxyglucose positron emission tomography (FDG-PET) and computed tomography (CT) are indispensable components in modern medicine. Although PET can provide additional diagnostic value, it is costly and not universally accessible, particularly in low-income countries. To bridge this gap, we have developed a conditional generative adversarial network pipeline that can produce FDG-PET from diagnostic CT scans based on multi-center multi-modal lung cancer datasets (n = 1,478). Synthetic PET images are validated across imaging, biological, and clinical aspects. Radiologists confirm comparable imaging quality and tumor contrast between synthetic and actual PET scans. Radiogenomics analysis further proves that the dysregulated cancer hallmark pathways of synthetic PET are consistent with actual PET. We also demonstrate the clinical values of synthetic PET in improving lung cancer diagnosis, staging, risk prediction, and prognosis. Taken together, this proof-of-concept study testifies to the feasibility of applying deep learning to obtain high-fidelity PET translated from CT.
Collapse
Affiliation(s)
| | | | - Sheeba J Sujit
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed Qayati
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Maliazurina B Saad
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Lingzhi Hong
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX USA
| | - Julie Lin
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin J Ostrin
- Department of General Internal Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Iakovos Toumazis
- Department of Health Services Research, MD Anderson Cancer Center, Houston, TX, USA
| | - Peng Huang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey H Siewerdsen
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - John D Hazle
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - David Jaffray
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Gladish
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA; Lung Cancer Genomics Program, MD Anderson Cancer Center, Houston, TX, USA; Lung Cancer Interception Program, MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Cai L, Sidey-Gibbons C, Nees J, Riedel F, Schaefgen B, Togawa R, Killinger K, Heil J, Pfob A, Golatta M. Ultrasound Radiomics Features to Identify Patients With Triple-Negative Breast Cancer: A Retrospective, Single-Center Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:467-478. [PMID: 38069582 DOI: 10.1002/jum.16377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/04/2023] [Indexed: 02/08/2024]
Abstract
OBJECTIVES Patients with triple-negative breast cancer (TNBC) exhibit a fast tumor growth rate and poor survival outcomes. In this study, we aimed to develop and compare intelligent algorithms using ultrasound radiomics features in addition to clinical variables to identify patients with TNBC prior to histopathologic diagnosis. METHODS We used single-center, retrospective data of patients who underwent ultrasound before histopathologic verification and subsequent neoadjuvant systemic treatment (NAST). We developed a logistic regression with an elastic net penalty algorithm using pretreatment ultrasound radiomics features in addition to patient and tumor variables to identify patients with TNBC. Findings were compared to the histopathologic evaluation of the biopsy specimen. The main outcome measure was the area under the curve (AUC). RESULTS We included 1161 patients, 813 in the development set and 348 in the validation set. Median age was 50.1 years and 24.4% (283 of 1161) had TNBC. The integrative model using radiomics and clinical information showed significantly better performance in identifying TNBC compared to the radiomics model (AUC: 0.71, 95% confidence interval [CI]: 0.65-0.76 versus 0.64, 95% CI: 0.57-0.71, P = .004). The five most important variables were cN status, shape surface volume ratio (SA:V), gray level co-occurrence matrix (GLCM) correlation, gray level dependence matrix (GLDM) dependence nonuniformity normalized, and age. Patients with TNBC were more often categorized as BI-RADS 4 than BI-RADS 5 compared to non-TNBC patients (P = .002). CONCLUSION A machine learning algorithm showed promising potential to identify patients with TNBC using ultrasound radiomics features and clinical information prior to histopathologic evaluation.
Collapse
Affiliation(s)
- Lie Cai
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Chris Sidey-Gibbons
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juliane Nees
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fabian Riedel
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Schaefgen
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Riku Togawa
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristina Killinger
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Heil
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - André Pfob
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Golatta
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Occhipinti A, Verma S, Doan LMT, Angione C. Mechanism-aware and multimodal AI: beyond model-agnostic interpretation. Trends Cell Biol 2024; 34:85-89. [PMID: 38087709 DOI: 10.1016/j.tcb.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 02/04/2024]
Abstract
Artificial intelligence (AI) is widely used for exploiting multimodal biomedical data, with increasingly accurate predictions and model-agnostic interpretations, which are however also agnostic to biological mechanisms. Combining metabolic modelling, 'omics, and imaging data via multimodal AI can generate predictions that can be interpreted mechanistically and transparently, therefore with significantly higher therapeutic potential.
Collapse
Affiliation(s)
- Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesborough, UK; Centre for Digital Innovation, Teesside University, Middlesborough, UK; National Horizons Centre, Teesside University, Darlington, UK
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesborough, UK
| | - Le Minh Thao Doan
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesborough, UK
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesborough, UK; Centre for Digital Innovation, Teesside University, Middlesborough, UK; National Horizons Centre, Teesside University, Darlington, UK.
| |
Collapse
|
17
|
Ma XK, Liu TL, Ren YN, Ma XP, Yao Y, Hou XG, Ding J, Wang F, Huang HF, Zhu H, Yang Z. 124I-labeled anti-CD147 antibody for noninvasive detection of CD147-positive pan-cancers: construction and preclinical studies. Acta Pharmacol Sin 2024; 45:436-448. [PMID: 37749238 PMCID: PMC10789738 DOI: 10.1038/s41401-023-01162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
Extracellular matrix metalloproteinase inducer CD147 is a glycoprotein on the cell surface. There is minimal expression of CD147 in normal epithelial and fetal tissues, but it is highly expressed in a number of aggressive tumors. CD147 has been implicated in pan-cancer immunity and progression. With the development of CD147-targeting therapeutic strategy, accurate detection of CD147 expression in tumors and its changes during the therapy is necessary. In this study we constructed a novel radiotracer by labeling the anti-CD147 mAb with radionuclide 124/125I (124/125I-anti-CD147) for noninvasive detection of CD147 expression in pan-cancers, and characterized its physicochemical properties, affinity, metabolic characteristics, biodistribution and immunoPET imaging with 124I-IgG and 18F-FDG as controls. By examining the expression of CD147 in cancer cell lines, we found high CD147 expression in colon cancer cells LS174T, FADU human pharyngeal squamous cancer cells and 22RV1 human prostate cancer cells, and low expression of CD147 in human pancreatic cancer cells ASPC1 and human gastric cancer cells BGC823. 124/125I-anti-CD147 was prepared using N-bromine succinimide (NBS) as oxidant and purified by PD-10 column. Its radiochemical purity (RCP) was over 99% and maintained over 85% in saline or 5% human serum albumin (HSA) for more than 7 d; the RCP of 125I-anti-CD147 in blood was over 90% at 3 h post injection (p.i.) in healthy mice. The Kd value of 125I-anti-CD147 to CD147 protein was 6.344 nM, while that of 125I-IgG was over 100 nM. 125I-anti-CD147 showed much greater uptake in CD147 high-expression cancer cells compared to CD147 low-expression cancer cells. After intravenous injection in healthy mice, 125I-anti-CD147 showed high initial uptake in blood pool and liver, the uptake was decreased with time. The biological half-life of distribution and clearance phases in healthy mice were 0.63 h and 19.60 h, respectively. The effective dose of 124I-anti-CD147 was estimated as 0.104 mSv/MBq. We conducted immunoPET imaging in tumor-bearing mice, and demonstrated a significantly higher tumor-to-muscle ratio of 124I-anti-CD147 compared to that of 124I-IgG and 18F-FDG in CD147 (+) tumors. The expression levels of CD147 in cells and tumors were positively correlated with the maximum standardized uptake value (SUVmax) (P < 0.01). In conclusion, 124/125I-anti-CD147 displays high affinity to CD147, and represents potential for the imaging of CD147-positive tumors. The development of 124I-anti-CD147 may provide new insights into the regulation of tumor microenvironment and formulation of precision diagnosis and treatment programs for tumors.
Collapse
Affiliation(s)
- Xiao-Kun Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Te-Li Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ya-Nan Ren
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Medical College, Guizhou University, Guiyang, 550025, China
| | - Xiao-Pan Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Medical College, Guizhou University, Guiyang, 550025, China
| | - Yuan Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xing-Guo Hou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jin Ding
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Feng Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Hai-Feng Huang
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
18
|
Yang J, Huang J, Han D, Ma X. Artificial Intelligence Applications in the Treatment of Colorectal Cancer: A Narrative Review. Clin Med Insights Oncol 2024; 18:11795549231220320. [PMID: 38187459 PMCID: PMC10771756 DOI: 10.1177/11795549231220320] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/26/2023] [Indexed: 01/09/2024] Open
Abstract
Colorectal cancer is the third most prevalent cancer worldwide, and its treatment has been a demanding clinical problem. Beyond traditional surgical therapy and chemotherapy, newly revealed molecular mechanisms diversify therapeutic approaches for colorectal cancer. However, the selection of personalized treatment among multiple treatment options has become another challenge in the era of precision medicine. Artificial intelligence has recently been increasingly investigated in the treatment of colorectal cancer. This narrative review mainly discusses the applications of artificial intelligence in the treatment of colorectal cancer patients. A comprehensive literature search was conducted in MEDLINE, EMBASE, and Web of Science to identify relevant papers, resulting in 49 articles being included. The results showed that, based on different categories of data, artificial intelligence can predict treatment outcomes and essential guidance information of traditional and novel therapies, thus enabling individualized treatment strategy selection for colorectal cancer patients. Some frequently implemented machine learning algorithms and deep learning frameworks have also been employed for long-term prognosis prediction in patients with colorectal cancer. Overall, artificial intelligence shows encouraging results in treatment strategy selection and prognosis evaluation for colorectal cancer patients.
Collapse
Affiliation(s)
- Jiaqing Yang
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Huang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Deqian Han
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Dhoundiyal S, Alam MA. Advancements in Biotechnology and Stem Cell Therapies for Breast Cancer Patients. Curr Stem Cell Res Ther 2024; 19:1072-1083. [PMID: 37815191 DOI: 10.2174/011574888x268109230924233850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
This comprehensive review article examines the integration of biotechnology and stem cell therapy in breast cancer diagnosis and treatment. It discusses the use of biotechnological tools such as liquid biopsies, genomic profiling, and imaging technologies for accurate diagnosis and monitoring of treatment response. Stem cell-based approaches, their role in modeling breast cancer progression, and their potential for breast reconstruction post-mastectomy are explored. The review highlights the importance of personalized treatment strategies that combine biotechnological tools and stem cell therapies. Ethical considerations, challenges in clinical translation, and regulatory frameworks are also addressed. The article concludes by emphasizing the potential of integrating biotechnology and stem cell therapy to improve breast cancer outcomes, highlighting the need for continued research and collaboration in this field.
Collapse
Affiliation(s)
- Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar
Pradesh, India
| |
Collapse
|
20
|
Chen B, Mao Y, Li J, Zhao Z, Chen Q, Yu Y, Yang Y, Dong Y, Lin G, Yao J, Lu M, Wu L, Bo Z, Chen G, Xie X. Predicting very early recurrence in intrahepatic cholangiocarcinoma after curative hepatectomy using machine learning radiomics based on CECT: A multi-institutional study. Comput Biol Med 2023; 167:107612. [PMID: 37939408 DOI: 10.1016/j.compbiomed.2023.107612] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/11/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Even after curative resection, the prognosis for patients with intrahepatic cholangiocarcinoma (iCCA) remains disappointing due to the extremely high incidence of postoperative recurrence. METHODS A total of 280 iCCA patients following curative hepatectomy from three independent institutions were recruited to establish the retrospective multicenter cohort study. The very early recurrence (VER) of iCCA was defined as the appearance of recurrence within 6 months. The 3D tumor region of interest (ROI) derived from contrast-enhanced CT (CECT) was used for radiomics analysis. The independent clinical predictors for VER were histological stage, AJCC stage, and CA199 levels. We implemented K-means clustering algorithm to investigate novel radiomics-based subtypes of iCCA. Six types of machine learning (ML) algorithms were performed for VER prediction, including logistic, random forest (RF), neural network, bayes, support vector machine (SVM), and eXtreme Gradient Boosting (XGBoost). Additionally, six clinical ML (CML) models and six radiomics-clinical ML (RCML) models were developed to predict VER. Predictive performance was internally validated by 10-fold cross-validation in the training cohort, and further evaluated in the external validation cohort. RESULTS Approximately 30 % of patients with iCCA experienced VER with extremely discouraging outcome (Hazard ratio (HR) = 5.77, 95 % Confidence Interval (CI) = 3.73-8.93, P < 0.001). Two distinct iCCA subtypes based on radiomics features were identified, and subtype 2 harbored a higher proportion of VER (47.62 % Vs 25.53 %) and significant shorter survival time than subtype 1. The average AUC values of the CML and RCML models were 0.744 ± 0.018, and 0.900 ± 0.014 in the training cohort, and 0.769 ± 0.065 and 0.929 ± 0.027 in the external validation cohort, respectively. CONCLUSION Two radiomics-based iCCA subtypes were identified, and six RCML models were developed to predict VER of iCCA, which can be used as valid tools to guide individualized management in clinical practice.
Collapse
Affiliation(s)
- Bo Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yicheng Mao
- Department of Optometry and Ophthalmology College, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiacheng Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhengxiao Zhao
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Qiwen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yaoyao Yu
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yulong Dong
- Department of Oncology, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai 200438, China
| | - Ganglian Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiangqiao Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengmeng Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lijun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhiyuan Bo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Xiaozai Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
21
|
Nguyen TM, Bertolus C, Giraud P, Burgun A, Saintigny P, Bibault JE, Foy JP. A Radiomics Approach to Identify Immunologically Active Tumor in Patients with Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2023; 15:5369. [PMID: 38001629 PMCID: PMC10670096 DOI: 10.3390/cancers15225369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND We recently developed a gene-expression-based HOT score to identify the hot/cold phenotype of head and neck squamous cell carcinomas (HNSCCs), which is associated with the response to immunotherapy. Our goal was to determine whether radiomic profiling from computed tomography (CT) scans can distinguish hot and cold HNSCC. METHOD We included 113 patients from The Cancer Genome Atlas (TCGA) and 20 patients from the Groupe Hospitalier Pitié-Salpêtrière (GHPS) with HNSCC, all with available pre-treatment CT scans. The hot/cold phenotype was computed for all patients using the HOT score. The IBEX software (version 4.11.9, accessed on 30 march 2020) was used to extract radiomic features from the delineated tumor region in both datasets, and the intraclass correlation coefficient (ICC) was computed to select robust features. Machine learning classifier models were trained and tested in the TCGA dataset and validated using the area under the receiver operator characteristic curve (AUC) in the GHPS cohort. RESULTS A total of 144 radiomic features with an ICC >0.9 was selected. An XGBoost model including these selected features showed the best performance prediction of the hot/cold phenotype with AUC = 0.86 in the GHPS validation dataset. CONCLUSIONS AND RELEVANCE We identified a relevant radiomic model to capture the overall hot/cold phenotype of HNSCC. This non-invasive approach could help with the identification of patients with HNSCC who may benefit from immunotherapy.
Collapse
Affiliation(s)
- Tan Mai Nguyen
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Chloé Bertolus
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
| | - Paul Giraud
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Sorbonne Université, Department of Radiation Oncology, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France
| | - Anita Burgun
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Jean-Emmanuel Bibault
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Department of Radiation Oncology, Hôpital Européen Georges-Pompidou, Université Paris Cité, 75015 Paris, France
| | - Jean-Philippe Foy
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Sorbonne Université, INSERM UMRS 938, Centre de Recherche de Saint Antoine, Team Cancer Biology and Therapeutics, 75011 Paris, France
| |
Collapse
|
22
|
Wei L, Niraula D, Gates EDH, Fu J, Luo Y, Nyflot MJ, Bowen SR, El Naqa IM, Cui S. Artificial intelligence (AI) and machine learning (ML) in precision oncology: a review on enhancing discoverability through multiomics integration. Br J Radiol 2023; 96:20230211. [PMID: 37660402 PMCID: PMC10546458 DOI: 10.1259/bjr.20230211] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 09/05/2023] Open
Abstract
Multiomics data including imaging radiomics and various types of molecular biomarkers have been increasingly investigated for better diagnosis and therapy in the era of precision oncology. Artificial intelligence (AI) including machine learning (ML) and deep learning (DL) techniques combined with the exponential growth of multiomics data may have great potential to revolutionize cancer subtyping, risk stratification, prognostication, prediction and clinical decision-making. In this article, we first present different categories of multiomics data and their roles in diagnosis and therapy. Second, AI-based data fusion methods and modeling methods as well as different validation schemes are illustrated. Third, the applications and examples of multiomics research in oncology are demonstrated. Finally, the challenges regarding the heterogeneity data set, availability of omics data, and validation of the research are discussed. The transition of multiomics research to real clinics still requires consistent efforts in standardizing omics data collection and analysis, building computational infrastructure for data sharing and storing, developing advanced methods to improve data fusion and interpretability, and ultimately, conducting large-scale prospective clinical trials to fill the gap between study findings and clinical benefits.
Collapse
Affiliation(s)
- Lise Wei
- Department of Radiation Oncology, University of Michigan, Michigan, United States
| | - Dipesh Niraula
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Evan D. H. Gates
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Jie Fu
- Department of Radiation Oncology, Stanford University, Stanford, California, United States
| | - Yi Luo
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Matthew J. Nyflot
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Stephen R. Bowen
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Issam M El Naqa
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Sunan Cui
- Department of Radiation Oncology, University of Washington, Washington, United States
| |
Collapse
|
23
|
Felfli M, Liu Y, Zerka F, Voyton C, Thinnes A, Jacques S, Iannessi A, Bodard S. Systematic Review, Meta-Analysis and Radiomics Quality Score Assessment of CT Radiomics-Based Models Predicting Tumor EGFR Mutation Status in Patients with Non-Small-Cell Lung Cancer. Int J Mol Sci 2023; 24:11433. [PMID: 37511192 PMCID: PMC10380456 DOI: 10.3390/ijms241411433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Assessment of the quality and current performance of computed tomography (CT) radiomics-based models in predicting epidermal growth factor receptor (EGFR) mutation status in patients with non-small-cell lung carcinoma (NSCLC). Two medical literature databases were systematically searched, and articles presenting original studies on CT radiomics-based models for predicting EGFR mutation status were retrieved. Forest plots and related statistical tests were performed to summarize the model performance and inter-study heterogeneity. The methodological quality of the selected studies was assessed via the Radiomics Quality Score (RQS). The performance of the models was evaluated using the area under the curve (ROC AUC). The range of the Risk RQS across the selected articles varied from 11 to 24, indicating a notable heterogeneity in the quality and methodology of the included studies. The average score was 15.25, which accounted for 42.34% of the maximum possible score. The pooled Area Under the Curve (AUC) value was 0.801, indicating the accuracy of CT radiomics-based models in predicting the EGFR mutation status. CT radiomics-based models show promising results as non-invasive alternatives for predicting EGFR mutation status in NSCLC patients. However, the quality of the studies using CT radiomics-based models varies widely, and further harmonization and prospective validation are needed before the generalization of these models.
Collapse
Affiliation(s)
- Mehdi Felfli
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Yan Liu
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Fadila Zerka
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Charles Voyton
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Alexandre Thinnes
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Sebastien Jacques
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Antoine Iannessi
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
- Centre Antoine Lacassagne, F-06100 Nice, France
| | - Sylvain Bodard
- AP-HP, Service d’Imagerie Adulte, Hôpital Necker Enfants Malades, Université de Paris Cité, F-75015 Paris, France
- CNRS UMR 7371, INSERM U 1146, Laboratoire d’Imagerie Biomédicale, Sorbonne Université, F-75006 Paris, France
| |
Collapse
|
24
|
Saad MB, Hong L, Aminu M, Vokes NI, Chen P, Salehjahromi M, Qin K, Sujit SJ, Lu X, Young E, Al-Tashi Q, Qureshi R, Wu CC, Carter BW, Lin SH, Lee PP, Gandhi S, Chang JY, Li R, Gensheimer MF, Wakelee HA, Neal JW, Lee HS, Cheng C, Velcheti V, Lou Y, Petranovic M, Rinsurongkawong W, Le X, Rinsurongkawong V, Spelman A, Elamin YY, Negrao MV, Skoulidis F, Gay CM, Cascone T, Antonoff MB, Sepesi B, Lewis J, Wistuba II, Hazle JD, Chung C, Jaffray D, Gibbons DL, Vaporciyan A, Lee JJ, Heymach JV, Zhang J, Wu J. Predicting benefit from immune checkpoint inhibitors in patients with non-small-cell lung cancer by CT-based ensemble deep learning: a retrospective study. Lancet Digit Health 2023; 5:e404-e420. [PMID: 37268451 PMCID: PMC10330920 DOI: 10.1016/s2589-7500(23)00082-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Only around 20-30% of patients with non-small-cell lung cancer (NCSLC) have durable benefit from immune-checkpoint inhibitors. Although tissue-based biomarkers (eg, PD-L1) are limited by suboptimal performance, tissue availability, and tumour heterogeneity, radiographic images might holistically capture the underlying cancer biology. We aimed to investigate the application of deep learning on chest CT scans to derive an imaging signature of response to immune checkpoint inhibitors and evaluate its added value in the clinical context. METHODS In this retrospective modelling study, 976 patients with metastatic, EGFR/ALK negative NSCLC treated with immune checkpoint inhibitors at MD Anderson and Stanford were enrolled from Jan 1, 2014, to Feb 29, 2020. We built and tested an ensemble deep learning model on pretreatment CTs (Deep-CT) to predict overall survival and progression-free survival after treatment with immune checkpoint inhibitors. We also evaluated the added predictive value of the Deep-CT model in the context of existing clinicopathological and radiological metrics. FINDINGS Our Deep-CT model demonstrated robust stratification of patient survival of the MD Anderson testing set, which was validated in the external Stanford set. The performance of the Deep-CT model remained significant on subgroup analyses stratified by PD-L1, histology, age, sex, and race. In univariate analysis, Deep-CT outperformed the conventional risk factors, including histology, smoking status, and PD-L1 expression, and remained an independent predictor after multivariate adjustment. Integrating the Deep-CT model with conventional risk factors demonstrated significantly improved prediction performance, with overall survival C-index increases from 0·70 (clinical model) to 0·75 (composite model) during testing. On the other hand, the deep learning risk scores correlated with some radiomics features, but radiomics alone could not reach the performance level of deep learning, indicating that the deep learning model effectively captured additional imaging patterns beyond known radiomics features. INTERPRETATION This proof-of-concept study shows that automated profiling of radiographic scans through deep learning can provide orthogonal information independent of existing clinicopathological biomarkers, bringing the goal of precision immunotherapy for patients with NSCLC closer. FUNDING National Institutes of Health, Mark Foundation Damon Runyon Foundation Physician Scientist Award, MD Anderson Strategic Initiative Development Program, MD Anderson Lung Moon Shot Program, Andrea Mugnaini, and Edward L C Smith.
Collapse
Affiliation(s)
- Maliazurina B Saad
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingzhi Hong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Morteza Salehjahromi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kang Qin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheeba J Sujit
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuetao Lu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elliana Young
- Department of Enterprise Data Engineering and Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qasem Al-Tashi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rizwan Qureshi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brett W Carter
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Percy P Lee
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Saumil Gandhi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael F Gensheimer
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Heather A Wakelee
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Joel W Neal
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J Sugarbaker Division of Thoracic Surgery, Michael E DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, New York University Langone Health, New York, NY, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Milena Petranovic
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Waree Rinsurongkawong
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vadeerat Rinsurongkawong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy Spelman
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcelo V Negrao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ferdinandos Skoulidis
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeff Lewis
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John D Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Jaffray
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
25
|
Xia TY, Zhou ZH, Meng XP, Zha JH, Yu Q, Wang WL, Song Y, Wang YC, Tang TY, Xu J, Zhang T, Long XY, Liang Y, Xiao WB, Ju SH. Predicting Microvascular Invasion in Hepatocellular Carcinoma Using CT-based Radiomics Model. Radiology 2023; 307:e222729. [PMID: 37097141 DOI: 10.1148/radiol.222729] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Background Prediction of microvascular invasion (MVI) may help determine treatment strategies for hepatocellular carcinoma (HCC). Purpose To develop a radiomics approach for predicting MVI status based on preoperative multiphase CT images and to identify MVI-associated differentially expressed genes. Materials and Methods Patients with pathologically proven HCC from May 2012 to September 2020 were retrospectively included from four medical centers. Radiomics features were extracted from tumors and peritumor regions on preoperative registration or subtraction CT images. In the training set, these features were used to build five radiomics models via logistic regression after feature reduction. The models were tested using internal and external test sets against a pathologic reference standard to calculate area under the receiver operating characteristic curve (AUC). The optimal AUC radiomics model and clinical-radiologic characteristics were combined to build the hybrid model. The log-rank test was used in the outcome cohort (Kunming center) to analyze early recurrence-free survival and overall survival based on high versus low model-derived score. RNA sequencing data from The Cancer Image Archive were used for gene expression analysis. Results A total of 773 patients (median age, 59 years; IQR, 49-64 years; 633 men) were divided into the training set (n = 334), internal test set (n = 142), external test set (n = 141), outcome cohort (n = 121), and RNA sequencing analysis set (n = 35). The AUCs from the radiomics and hybrid models, respectively, were 0.76 and 0.86 for the internal test set and 0.72 and 0.84 for the external test set. Early recurrence-free survival (P < .01) and overall survival (P < .007) can be categorized using the hybrid model. Differentially expressed genes in patients with findings positive for MVI were involved in glucose metabolism. Conclusion The hybrid model showed the best performance in prediction of MVI. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Summers in this issue.
Collapse
Affiliation(s)
- Tian-Yi Xia
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Zheng-Hao Zhou
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Xiang-Pan Meng
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Jun-Hao Zha
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Qian Yu
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Wei-Lang Wang
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Yang Song
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Yuan-Cheng Wang
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Tian-Yu Tang
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Jun Xu
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Tao Zhang
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Xue-Ying Long
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Yun Liang
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Wen-Bo Xiao
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| | - Sheng-Hong Ju
- From the Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao Road, Nanjing, China 210009 (T.Y.X., X.P.M., J.H.Z., Q.Y., W.L.W., Y.C.W., T.Y.T., S.H.J.); Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China (Z.H.Z., J.X.); MR Scientific Marketing, Siemens Healthineers, Shanghai, China (Y.S.); Department of Radiology, The Third Affiliated Hospital of Nantong University, Nantong, China (T.Z.); Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China (X.Y.L.); Department of Radiology, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); and Department of Radiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (W.B.X.)
| |
Collapse
|
26
|
Volpe S, Gaeta A, Colombo F, Zaffaroni M, Mastroleo F, Vincini MG, Pepa M, Isaksson LJ, Turturici I, Marvaso G, Ferrari A, Cammarata G, Santamaria R, Franzetti J, Raimondi S, Botta F, Ansarin M, Gandini S, Cremonesi M, Orecchia R, Alterio D, Jereczek-Fossa BA. Blood- and Imaging-Derived Biomarkers for Oncological Outcome Modelling in Oropharyngeal Cancer: Exploring the Low-Hanging Fruit. Cancers (Basel) 2023; 15:cancers15072022. [PMID: 37046683 PMCID: PMC10093133 DOI: 10.3390/cancers15072022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023] Open
Abstract
Aims: To assess whether CT-based radiomics and blood-derived biomarkers could improve the prediction of overall survival (OS) and locoregional progression-free survival (LRPFS) in patients with oropharyngeal cancer (OPC) treated with curative-intent RT. Methods: Consecutive OPC patients with primary tumors treated between 2005 and 2021 were included. Analyzed clinical variables included gender, age, smoking history, staging, subsite, HPV status, and blood parameters (baseline hemoglobin levels, neutrophils, monocytes, and platelets, and derived measurements). Radiomic features were extracted from the gross tumor volumes (GTVs) of the primary tumor using pyradiomics. Outcomes of interest were LRPFS and OS. Following feature selection, a radiomic score (RS) was calculated for each patient. Significant variables, along with age and gender, were included in multivariable analysis, and models were retained if statistically significant. The models’ performance was compared by the C-index. Results: One hundred and five patients, predominately male (71%), were included in the analysis. The median age was 59 (IQR: 52–66) years, and stage IVA was the most represented (70%). HPV status was positive in 63 patients, negative in 7, and missing in 35 patients. The median OS follow-up was 6.3 (IQR: 5.5–7.9) years. A statistically significant association between low Hb levels and poorer LRPFS in the HPV-positive subgroup (p = 0.038) was identified. The calculation of the RS successfully stratified patients according to both OS (log-rank p < 0.0001) and LRPFS (log-rank p = 0.0002). The C-index of the clinical and radiomic model resulted in 0.82 [CI: 0.80–0.84] for OS and 0.77 [CI: 0.75–0.79] for LRPFS. Conclusions: Our results show that radiomics could provide clinically significant informative content in this scenario. The best performances were obtained by combining clinical and quantitative imaging variables, thus suggesting the potential of integrative modeling for outcome predictions in this setting of patients.
Collapse
|
27
|
Lv C, He N, Yang JJ, Xiao JJ, Zhang Y, Du J, Zuo S, Li HY, Gu H. Prediction of 3-year recurrence rate of hepatocellular carcinoma after resection based on contrast-enhanced CT: a single-centre study. Br J Radiol 2023; 96:20220702. [PMID: 36745047 PMCID: PMC10161905 DOI: 10.1259/bjr.20220702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE We present a new artificial intelligence-powered method to predict 3-year hepatocellular carcinoma (HCC) recurrence by analysing the radiomic profile of contrast-enhanced CT (CECT) images that was validated in patient cohorts. METHODS This retrospective cohort study of 224 HCC patients with follow-up for at least 3 years was performed at a single centre from 2012 to 2019. Two groups of radiomic signatures were extracted from the arterial and portal venous phases of pre-operative CECT. Then, the radiological model (RM), deep learning-based radiomics model (DLRM), and clinical & deep learning-based radiomics model (CDLRM) were established and validated in the area under curve (AUC), calibration curve, and clinical decision curve. RESULTS Comparison of the clinical baseline variables between the non-recurrence (n = 109) and recurrence group (n = 115), three clinical independent factors (Barcelona Clinic Liver Cancer staging, microvascular invasion, and α-fetoprotein) were incorporated into DLRM for the CDLRM construction. Among the 30 radiomic features most crucial to the 3 year recurrence rate, the selection from deep learning-based radiomics (DLR) features depends on CECT. through the Gini index. In most cases, CDLRM has shown superior accuracy and distinguished performance than DLRM and RM, with the 0.98 AUC in the training cohorts and 0.83 in the testing. CONCLUSION This study proposed that DLR-based CDLRM construction would be allowed for the predictive utility of 3-year recurrence outcomes of HCCs, providing high-risk patients with an effective and non-invasive method to possess extra clinical intervention. ADVANCES IN KNOWLEDGE This study has highlighted the predictive value of DLR in the 3-year recurrence rate of HCC.
Collapse
Affiliation(s)
- Chao Lv
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Nan He
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jie Jie Yang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jing Jing Xiao
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Yan Zhang
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jun Du
- Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shi Zuo
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hai Yang Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Huajian Gu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China.,Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
28
|
McCague C, Ramlee S, Reinius M, Selby I, Hulse D, Piyatissa P, Bura V, Crispin-Ortuzar M, Sala E, Woitek R. Introduction to radiomics for a clinical audience. Clin Radiol 2023; 78:83-98. [PMID: 36639175 DOI: 10.1016/j.crad.2022.08.149] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Radiomics is a rapidly developing field of research focused on the extraction of quantitative features from medical images, thus converting these digital images into minable, high-dimensional data, which offer unique biological information that can enhance our understanding of disease processes and provide clinical decision support. To date, most radiomics research has been focused on oncological applications; however, it is increasingly being used in a raft of other diseases. This review gives an overview of radiomics for a clinical audience, including the radiomics pipeline and the common pitfalls associated with each stage. Key studies in oncology are presented with a focus on both those that use radiomics analysis alone and those that integrate its use with other multimodal data streams. Importantly, clinical applications outside oncology are also presented. Finally, we conclude by offering a vision for radiomics research in the future, including how it might impact our practice as radiologists.
Collapse
Affiliation(s)
- C McCague
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - S Ramlee
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - M Reinius
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - I Selby
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - D Hulse
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Piyatissa
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - V Bura
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Radiology and Medical Imaging, County Clinical Emergency Hospital, Cluj-Napoca, Romania
| | - M Crispin-Ortuzar
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK
| | - E Sala
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - R Woitek
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Research Centre for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
29
|
Jia Y, Zhu Y, Li T, Song X, Duan Y, Yang D, Nie F. Evaluating Tumor-Infiltrating Lymphocytes in Breast Cancer: The Role of Conventional Ultrasound and Contrast-Enhanced Ultrasound. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:623-634. [PMID: 35866231 DOI: 10.1002/jum.16058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/21/2022] [Accepted: 06/25/2022] [Indexed: 05/27/2023]
Abstract
OBJECTIVES Tumor-infiltrating lymphocytes (TILs) have emerged as an efficient biomarker predicting treatment response and prognosis of breast cancer (BC). This study aimed to evaluate the association between conventional ultrasound and contrast-enhanced ultrasound (CEUS) imaging features with TIL levels in invasive BC patients. METHODS We retrospectively included 267 women with invasive BC who had undergone conventional ultrasound and CEUS. Patients were divided into low (≤10%) and high (>10%) TIL groups. Conventional ultrasound and CEUS features were analyzed by two sonographers. The associations between the TIL levels and imaging features were evaluated. RESULTS Of the 267 patients, 122 with high TILs and 145 with low TIL levels. High TIL tumors were more likely to have a circumscribed margin, oval or round shape, and enhanced posterior echoes on ultrasonography (p < 0.05). In contrast, low TIL tumors were more likely to have an irregular shape, un-circumscribed, indistinct and spiculated margin (p < 0.05). In CEUS, high TIL tumors showed a more regular shape, clearer margin, more homogeneous enhancement and higher peak intensity (PI) value (p < 0.05). Logistic analysis indicated that shape, posterior features, PI, and enhanced homogeneity were independent predictors for high TIL tumors. The model combined the four independent predictors have a moderate performance in predicting high TIL tumors with AUC 0.79, sensitivity 0.72, and specificity 0.78. CONCLUSIONS Conventional ultrasound and CEUS features were associated with TIL levels in invasive BC. Consequently, the results suggested that preoperative conventional ultrasound and CEUS may be a useful noninvasive imaging biomarker for individualized treatment decisions.
Collapse
Affiliation(s)
- Yingying Jia
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
- Department of Ultrasound, People's Hospital of Ningxia Hui Nationality Autonomous Region, Yinchuan, Ningxia, China
- Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, China
- Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou, China
| | - Yangyang Zhu
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, China
- Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou, China
| | - Ting Li
- Department of Ultrasound, People's Hospital of Ningxia Hui Nationality Autonomous Region, Yinchuan, Ningxia, China
| | - XueWen Song
- Pathology Department, Lanzhou University Second Hospital, Lanzhou, China
| | - Ying Duan
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, China
- Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou, China
| | - Dan Yang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, China
- Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou, China
| | - Fang Nie
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, China
- Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou, China
| |
Collapse
|
30
|
Lin G, Wang X, Ye H, Cao W. Radiomic Models Predict Tumor Microenvironment Using Artificial Intelligence-the Novel Biomarkers in Breast Cancer Immune Microenvironment. Technol Cancer Res Treat 2023; 22:15330338231218227. [PMID: 38111330 PMCID: PMC10734346 DOI: 10.1177/15330338231218227] [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: 09/12/2023] [Revised: 10/22/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer is the most common malignancy in women, and some subtypes are associated with a poor prognosis with a lack of efficacious therapy. Moreover, immunotherapy and the use of other novel antibody‒drug conjugates have been rapidly incorporated into the standard management of advanced breast cancer. To extract more benefit from these therapies, clarifying and monitoring the tumor microenvironment (TME) status is critical, but this is difficult to accomplish based on conventional approaches. Radiomics is a method wherein radiological image features are comprehensively collected and assessed to build connections with disease diagnosis, prognosis, therapy efficacy, the TME, etc In recent years, studies focused on predicting the TME using radiomics have increasingly emerged, most of which demonstrate meaningful results and show better capability than conventional methods in some aspects. Beyond predicting tumor-infiltrating lymphocytes, immunophenotypes, cytokines, infiltrating inflammatory factors, and other stromal components, radiomic models have the potential to provide a completely new approach to deciphering the TME and facilitating tumor management by physicians.
Collapse
Affiliation(s)
- Guang Lin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hunan Ye
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
31
|
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: 29] [Impact Index Per Article: 9.7] [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
|
32
|
Lesion-specific exposure parameters for breast cancer diagnosis on digital breast tomosynthesis and full-field digital mammography. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2022.103752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
33
|
Chen P, Zhang J, Wu J. Artificial Intelligence in Digital Pathology to Advance Cancer Immunotherapy. 21ST CENTURY PATHOLOGY 2022; 2:120. [PMID: 36282981 PMCID: PMC9578679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionized the treatment of many malignancies. For instance, in lung cancer, however, only 20~30% of patients can achieve durable clinical benefits from ICI monotherapy. Histopathologic and molecular features such as histological type, PD-L1 expression, and tumor mutation burden (TMB), play a paramount role in selecting appropriate regimens for cancer treatment in the era of immunotherapy. Unfortunately, none of the existing features are exclusive predictive biomarkers. Thus, there is an imperative need to pinpoint more effective biomarkers to identify patients who may achieve the most benefit from ICIs. The adoption of digital pathology in clinical flow, as being powered by artificial intelligence (AI) especially deep learning, has catalyzed the automated analysis of tissue slides. With the breakthrough of multiplex bioimaging technology, researchers can comprehensively characterize the tumor microenvironment, including the different immune cells' distribution, function, and interaction. Here, we briefly summarize recent AI studies in digital pathology and share our perspective on emerging paradigms and directions to advance the development of immunotherapy biomarkers.
Collapse
Affiliation(s)
- Pingjun Chen
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
34
|
Jimenez JE, Dai D, Xu G, Zhao R, Li T, Pan T, Wang L, Lin Y, Wang Z, Jaffray D, Hazle JD, Macapinlac HA, Wu J, Lu Y. Lesion-Based Radiomics Signature in Pretherapy 18F-FDG PET Predicts Treatment Response to Ibrutinib in Lymphoma. Clin Nucl Med 2022; 47:209-218. [PMID: 35020640 PMCID: PMC8851692 DOI: 10.1097/rlu.0000000000004060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE The aim of this study was to develop a pretherapy PET/CT-based prediction model for treatment response to ibrutinib in lymphoma patients. PATIENTS AND METHODS One hundred sixty-nine lymphoma patients with 2441 lesions were studied retrospectively. All eligible lymphomas on pretherapy 18F-FDG PET images were contoured and segmented for radiomic analysis. Lesion- and patient-based responsiveness to ibrutinib was determined retrospectively using the Lugano classification. PET radiomic features were extracted. A radiomic model was built to predict ibrutinib response. The prognostic significance of the radiomic model was evaluated independently in a test cohort and compared with conventional PET metrics: SUVmax, metabolic tumor volume, and total lesion glycolysis. RESULTS The radiomic model had an area under the receiver operating characteristic curve (ROC AUC) of 0.860 (sensitivity, 92.9%, specificity, 81.4%; P < 0.001) for predicting response to ibrutinib, outperforming the SUVmax (ROC AUC, 0.519; P = 0.823), metabolic tumor volume (ROC AUC, 0.579; P = 0.412), total lesion glycolysis (ROC AUC, 0.576; P = 0.199), and a composite model built using all 3 (ROC AUC, 0.562; P = 0.046). The radiomic model increased the probability of accurately predicting ibrutinib-responsive lesions from 84.8% (pretest) to 96.5% (posttest). At the patient level, the model's performance (ROC AUC = 0.811; P = 0.007) was superior to that of conventional PET metrics. Furthermore, the radiomic model showed robustness when validated in treatment subgroups: first (ROC AUC, 0.916; P < 0.001) versus second or greater (ROC AUC, 0.842; P < 0.001) line of defense and single treatment (ROC AUC, 0.931; P < 0.001) versus multiple treatments (ROC AUC, 0.824; P < 0.001). CONCLUSIONS We developed and validated a pretherapy PET-based radiomic model to predict response to treatment with ibrutinib in a diverse cohort of lymphoma patients.
Collapse
Affiliation(s)
- Jorge E Jimenez
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dong Dai
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guofan Xu
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ruiyang Zhao
- Department of Electrical and Computer Engineering, Rice University, Houston, TX
| | - Tengfei Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tinsu Pan
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yingyan Lin
- Department of Electrical and Computer Engineering, Rice University, Houston, TX
| | - Zhangyang Wang
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX
| | - David Jaffray
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John D. Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Homer A. Macapinlac
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yang Lu
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
35
|
Tundo GR, Sbardella D, Oddone F, Kudriaeva AA, Lacal PM, Belogurov AA, Graziani G, Marini S. At the Cutting Edge against Cancer: A Perspective on Immunoproteasome and Immune Checkpoints Modulation as a Potential Therapeutic Intervention. Cancers (Basel) 2021; 13:4852. [PMID: 34638337 PMCID: PMC8507813 DOI: 10.3390/cancers13194852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/22/2023] Open
Abstract
Immunoproteasome is a noncanonical form of proteasome with enzymological properties optimized for the generation of antigenic peptides presented in complex with class I MHC molecules. This enzymatic property makes the modulation of its activity a promising area of research. Nevertheless, immunotherapy has emerged as a front-line treatment of advanced/metastatic tumors providing outstanding improvement of life expectancy, even though not all patients achieve a long-lasting clinical benefit. To enhance the efficacy of the currently available immunotherapies and enable the development of new strategies, a broader knowledge of the dynamics of antigen repertoire processing by cancer cells is needed. Therefore, a better understanding of the role of immunoproteasome in antigen processing and of the therapeutic implication of its modulation is mandatory. Studies on the potential crosstalk between proteasome modulators and immune checkpoint inhibitors could provide novel perspectives and an unexplored treatment option for a variety of cancers.
Collapse
Affiliation(s)
| | | | | | - Anna A. Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (A.A.K.)
| | - Pedro M. Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, 00167 Rome, Italy;
| | - Alexey A. Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (A.A.K.)
- Lomonosov Moscow State University, Leninskie Gory, 119991 Moscow, Russia
| | - Grazia Graziani
- Laboratory of Molecular Oncology, IDI-IRCCS, 00167 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| |
Collapse
|
36
|
Characterizing intra-tumor regions on quantitative ultrasound parametric images to predict breast cancer response to chemotherapy at pre-treatment. Sci Rep 2021; 11:14865. [PMID: 34290259 PMCID: PMC8295369 DOI: 10.1038/s41598-021-94004-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023] Open
Abstract
The efficacy of quantitative ultrasound (QUS) multi-parametric imaging in conjunction with unsupervised classification algorithms was investigated for the first time in characterizing intra-tumor regions to predict breast tumor response to chemotherapy before the start of treatment. QUS multi-parametric images of breast tumors were generated using the ultrasound radiofrequency data acquired from 181 patients diagnosed with locally advanced breast cancer and planned for neo-adjuvant chemotherapy followed by surgery. A hidden Markov random field (HMRF) expectation maximization (EM) algorithm was applied to identify distinct intra-tumor regions on QUS multi-parametric images. Several features were extracted from the segmented intra-tumor regions and tumor margin on different parametric images. A multi-step feature selection procedure was applied to construct a QUS biomarker consisting of four features for response prediction. Evaluation results on an independent test set indicated that the developed biomarker coupled with a decision tree model with adaptive boosting (AdaBoost) as the classifier could predict the treatment response of patient at pre-treatment with an accuracy of 85.4% and an area under the receiver operating characteristic (ROC) curve (AUC) of 0.89. In comparison, the biomarkers consisted of the features derived from the entire tumor core (without consideration of the intra-tumor regions), and the entire tumor core and the tumor margin could predict the treatment response of patients with an accuracy of 74.5% and 76.4%, and an AUC of 0.79 and 0.76, respectively. Standard clinical features could predict the therapy response with an accuracy of 69.1% and an AUC of 0.6. Long-term survival analyses indicated that the patients predicted by the developed model as responders had a significantly better survival compared to the non-responders. Similar findings were observed for the two response cohorts identified at post-treatment based on standard clinical and pathological criteria. The results obtained in this study demonstrated the potential of QUS multi-parametric imaging integrated with unsupervised learning methods in identifying distinct intra-tumor regions in breast cancer to characterize its responsiveness to chemotherapy prior to the start of treatment.
Collapse
|
37
|
Liu D, Chen J, Hu X, Yang K, Liu Y, Hu G, Ge H, Zhang W, Liu H. Imaging-Genomics in Glioblastoma: Combining Molecular and Imaging Signatures. Front Oncol 2021; 11:699265. [PMID: 34295824 PMCID: PMC8290166 DOI: 10.3389/fonc.2021.699265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Based on artificial intelligence (AI), computer-assisted medical diagnosis can scientifically and efficiently deal with a large quantity of medical imaging data. AI technologies including deep learning have shown remarkable progress across medical image recognition and genome analysis. Imaging-genomics attempts to explore the associations between potential gene expression patterns and specific imaging phenotypes. These associations provide potential cellular pathophysiology information, allowing sampling of the lesion habitat with high spatial resolution. Glioblastoma (GB) poses spatial and temporal heterogeneous characteristics, challenging to current precise diagnosis and treatments for the disease. Imaging-genomics provides a powerful tool for non-invasive global assessment of GB and its response to treatment. Imaging-genomics also has the potential to advance our understanding of underlying cancer biology, gene alterations, and corresponding biological processes. This article reviews the recent progress in the utilization of the imaging-genomics analysis in GB patients, focusing on its implications and prospects in individualized diagnosis and management.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Xinhua Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Guanjie Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Honglin Ge
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Hijacked Immune Cells in the Tumor Microenvironment: Molecular Mechanisms of Immunosuppression and Cues to Improve T Cell-Based Immunotherapy of Solid Tumors. Int J Mol Sci 2021; 22:ijms22115736. [PMID: 34072260 PMCID: PMC8199456 DOI: 10.3390/ijms22115736] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/13/2022] Open
Abstract
The understanding of the tumor microenvironment (TME) has been expanding in recent years in the context of interactions among different cell types, through direct cell–cell communication as well as through soluble factors. It has become evident that the development of a successful antitumor response depends on several TME factors. In this context, the number, type, and subsets of immune cells, as well as the functionality, memory, and exhaustion state of leukocytes are key factors of the TME. Both the presence and functionality of immune cells, in particular T cells, are regulated by cellular and soluble factors of the TME. In this regard, one fundamental reason for failure of antitumor responses is hijacked immune cells, which contribute to the immunosuppressive TME in multiple ways. Specifically, reactive oxygen species (ROS), metabolites, and anti-inflammatory cytokines have central roles in generating an immunosuppressive TME. In this review, we focused on recent developments in the immune cell constituents of the TME, and the micromilieu control of antitumor responses. Furthermore, we highlighted the current challenges of T cell-based immunotherapies and potential future strategies to consider for strengthening their effectiveness.
Collapse
|