101
|
Xu T, Liu X, Chen Y, Wang S, Jiang C, Gong J. CT-based deep learning radiomics biomarker for programmed cell death ligand 1 expression in non-small cell lung cancer. BMC Med Imaging 2024; 24:196. [PMID: 39085788 PMCID: PMC11292915 DOI: 10.1186/s12880-024-01380-8] [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: 08/05/2023] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Programmed cell death ligand 1 (PD-L1), as a reliable predictive biomarker, plays an important role in guiding immunotherapy of lung cancer. To investigate the value of CT-based deep learning radiomics signature to predict PD-L1 expression in non-small cell lung cancers(NSCLCs). METHODS 259 consecutive patients with pathological confirmed NSCLCs were retrospectively collected and divided into the training cohort and validation cohort according to the chronological order. The univariate and multivariate analyses were used to build the clinical model. Radiomics and deep learning features were extracted from preoperative non-contrast CT images. After feature selection, Radiomics score (Rad-score) and deep learning radiomics score (DLR-score) were calculated through a linear combination of the selected features and their coefficients. Predictive performance for PD-L1 expression was evaluated via the area under the curve (AUC) of receiver operating characteristic, the calibration curves, and the decision curve analysis. RESULTS The clinical model based on Cytokeratin 19 fragment and lobulated shape obtained an AUC of 0.767(95% CI: 0.673-0.860) in the training cohort and 0.604 (95% CI:0.477-0.731) in the validation cohort. 11 radiomics features and 15 deep learning features were selected by LASSO regression. AUCs of the Rad-score were 0.849 (95%CI: 0.783-0.914) and 0.717 (95%CI: 0.607-0.826) in the training cohort and validation cohort, respectively. AUCs of DLR-score were 0.938 (95%CI: 0.899-0.977) and 0.818(95%CI:0.727-0.910) in the training cohort and validation cohort, respectively. AUCs of the DLR-score were significantly higher than those of the Rad-score and the clinical model. CONCLUSION The CT-based deep learning radiomics signature could achieve clinically acceptable predictive performance for PD-L1 expression, which showed potential to be a surrogate imaging biomarker or a complement of immunohistochemistry assessment.
Collapse
Affiliation(s)
- Ting Xu
- The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaowen Liu
- The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Yaxi Chen
- The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Shuxing Wang
- The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Changsi Jiang
- Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), 1F, Building 4, No. 1017 Dongmen North Road, Shenzhen, 518020, China
| | - Jingshan Gong
- Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), 1F, Building 4, No. 1017 Dongmen North Road, Shenzhen, 518020, China.
| |
Collapse
|
102
|
Da-Ano R, Andrade-Miranda G, Tankyevych O, Visvikis D, Conze PH, Rest CCL. Automated PD-L1 status prediction in lung cancer with multi-modal PET/CT fusion. Sci Rep 2024; 14:16720. [PMID: 39030240 PMCID: PMC11271510 DOI: 10.1038/s41598-024-66487-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: 01/26/2024] [Accepted: 07/01/2024] [Indexed: 07/21/2024] Open
Abstract
Programmed death-ligand 1 (PD-L1) expressions play a crucial role in guiding therapeutic interventions such as the use of tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) in lung cancer. Conventional determination of PD-L1 status includes careful surgical or biopsied tumor specimens. These specimens are gathered through invasive procedures, representing a risk of difficulties and potential challenges in getting reliable and representative tissue samples. Using a single center cohort of 189 patients, our objective was to evaluate various fusion methods that used non-invasive computed tomography (CT) and18 F-FDG positron emission tomography (PET) images as inputs to various deep learning models to automatically predict PD-L1 in non-small cell lung cancer (NSCLC). We compared three different architectures (ResNet, DenseNet, and EfficientNet) and considered different input data (CT only, PET only, PET/CT early fusion, PET/CT late fusion without as well as with partially and fully shared weights to determine the best model performance. Models were assessed utilizing areas under the receiver operating characteristic curves (AUCs) considering their 95% confidence intervals (CI). The fusion of PET and CT images as input yielded better performance for PD-L1 classification. The different data fusion schemes systematically outperformed their individual counterparts when used as input of the various deep models. Furthermore, early fusion consistently outperformed late fusion, probably as a result of its capacity to capture more complicated patterns by merging PET and CT derived content at a lower level. When we looked more closely at the effects of weight sharing in late fusion architectures, we discovered that while it might boost model stability, it did not always result in better results. This suggests that although weight sharing could be beneficial when modality parameters are similar, the anatomical and metabolic information provided by CT and PET scans are too dissimilar to consistently lead to improved PD-L1 status predictions.
Collapse
Affiliation(s)
- Ronrick Da-Ano
- LaTIM, UMR 1101, Inserm, University of Brest, Brest, France
| | | | - Olena Tankyevych
- LaTIM, UMR 1101, Inserm, University of Brest, Brest, France
- Nuclear Medicine, University of Poitiers, Poitiers, France
| | | | - Pierre-Henri Conze
- LaTIM, UMR 1101, Inserm, University of Brest, Brest, France
- IMT Atlantique, Brest, France
| | - Catherine Cheze Le Rest
- LaTIM, UMR 1101, Inserm, University of Brest, Brest, France
- Nuclear Medicine, University of Poitiers, Poitiers, France
| |
Collapse
|
103
|
Hua Y, Sun Z, Xiao Y, Li H, Ma X, Luo X, Tan W, Xie Z, Zhang Z, Tang C, Zhuang H, Xu W, Zhu H, Chen Y, Shang C. Pretreatment CT-based machine learning radiomics model predicts response in unresectable hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors and interventional therapy. J Immunother Cancer 2024; 12:e008953. [PMID: 39029924 PMCID: PMC11261678 DOI: 10.1136/jitc-2024-008953] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Lenvatinib plus PD-1 inhibitors and interventional (LPI) therapy have demonstrated promising treatment effects in unresectable hepatocellular carcinoma (HCC). However, biomarkers for predicting the response to LPI therapy remain to be further explored. We aimed to develop a radiomics model to noninvasively predict the efficacy of LPI therapy. METHODS Clinical data of patients with HCC receiving LPI therapy were collected in our institution. The clinical model was built with clinical information. Nine machine learning classifiers were tested and the multilayer perceptron classifier with optimal performance was used as the radiomics model. The clinical-radiomics model was constructed by integrating clinical and radiomics scores through logistic regression analysis. RESULTS 151 patients were enrolled in this study (2:1 randomization, 101 and 50 in the training and validation cohorts), of which three achieved complete response, 69 showed partial response, 46 showed stable disease, and 33 showed progressive disease. The objective response rate, disease control rate, and conversion resection rates were 47.7, 78.1 and 23.2%. 14 features were selected from the initially extracted 1223 for radiomics model construction. The area under the curves of the radiomics model (0.900 for training and 0.893 for validation) were comparable to that of the clinical-radiomics model (0.912 for training and 0.892 for validation), and both were superior to the clinical model (0.669 for training and 0.585 for validation). Meanwhile, the radiomics model can categorize participants into high-risk and low-risk groups for progression-free survival (PFS) and overall survival (OS) in the training (HR 1.913, 95% CI 1.121 to 3.265, p=0.016 for PFS; HR 4.252, 95% CI 2.051 to 8.816, p=0.001 for OS) and validation sets (HR 2.347, 95% CI 1.095 to 5.031, p=0.012 for PFS; HR 2.592, 95% CI 1.050 to 6.394, p=0.019 for OS). CONCLUSION The promising machine learning radiomics model was developed and validated to predict the efficacy of LPI therapy for patients with HCC and perform risk stratification, with comparable performance to clinical-radiomics model.
Collapse
Affiliation(s)
- Yonglin Hua
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhixian Sun
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxin Xiao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Huilong Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaowu Ma
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xuan Luo
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenliang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Zhiqin Xie
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Ziyu Zhang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chenwei Tang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hongkai Zhuang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikai Xu
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haihong Zhu
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Yajin Chen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Changzhen Shang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
104
|
Han Y, Chang Y, Wang J, Li N, Yu Y, Yang Z, Lv W, Liu W, Yin J, Wu J. A study predicting long-term survival capacity in postoperative advanced gastric cancer patients based on MAOA and subcutaneous muscle fat characteristics. World J Surg Oncol 2024; 22:184. [PMID: 39010072 PMCID: PMC11251287 DOI: 10.1186/s12957-024-03466-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND The prognosis of advanced gastric cancer (AGC) is relatively poor, and long-term survival depends on timely intervention. Currently, predicting survival rates remains a hot topic. The application of radiomics and immunohistochemistry-related techniques in cancer research is increasingly widespread. However, their integration for predicting long-term survival in AGC patients has not been fully explored. METHODS We Collected 150 patients diagnosed with AGC at the Affiliated Zhongshan Hospital of Dalian University who underwent radical surgery between 2015 and 2019. Following strict inclusion and exclusion criteria, 90 patients were included in the analysis. We Collected postoperative pathological specimens from enrolled patients, analyzed the expression levels of MAOA using immunohistochemical techniques, and quantified these levels as the MAOAHScore. Obtained plain abdominal CT images from patients, delineated the region of interest at the L3 vertebral body level, and extracted radiomics features. Lasso Cox regression was used to select significant features to establish a radionics risk score, convert it into a categorical variable named risk, and use Cox regression to identify independent predictive factors for constructing a clinical prediction model. ROC, DCA, and calibration curves validated the model's performance. RESULTS The enrolled patients had an average age of 65.71 years, including 70 males and 20 females. Multivariate Cox regression analysis revealed that risk (P = 0.001, HR = 3.303), MAOAHScore (P = 0.043, HR = 2.055), and TNM stage (P = 0.047, HR = 2.273) emerged as independent prognostic risk factors for 3-year overall survival (OS) and The Similar results were found in the analysis of 3-year disease-specific survival (DSS). The nomogram developed could predict 3-year OS and DSS rates, with areas under the ROC curve (AUCs) of 0.81 and 0.797, respectively. Joint calibration and decision curve analyses (DCA) confirmed the nomogram's good predictive performance and clinical utility. CONCLUSION Integrating immunohistochemistry and muscle fat features provides a more accurate prediction of long-term survival in gastric cancer patients. This study offers new perspectives and methods for a deeper understanding of survival prediction in AGC.
Collapse
Grants
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
Collapse
Affiliation(s)
- Yubo Han
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yaoyuan Chang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiaqi Wang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Nanbo Li
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yang Yu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zhengbo Yang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Weipeng Lv
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wenfei Liu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| | - Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| |
Collapse
|
105
|
Wu A, Luo L, Zeng Q, Wu C, Shu X, Huang P, Wang Z, Hu T, Feng Z, Tu Y, Zhu Y, Cao Y, Li Z. Comparative assessment of the capability of machine learning-based radiomic models for predicting omental metastasis in locally advanced gastric cancer. Sci Rep 2024; 14:16208. [PMID: 39003337 PMCID: PMC11246510 DOI: 10.1038/s41598-024-66979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024] Open
Abstract
The study aims to investigate the predictive capability of machine learning algorithms for omental metastasis in locally advanced gastric cancer (LAGC) and to compare the performance metrics of various machine learning predictive models. A retrospective collection of 478 pathologically confirmed LAGC patients was undertaken, encompassing both clinical features and arterial phase computed tomography images. Radiomic features were extracted using 3D Slicer software. Clinical and radiomic features were further filtered through lasso regression. Selected clinical and radiomic features were used to construct omental metastasis predictive models using support vector machine (SVM), decision tree (DT), random forest (RF), K-nearest neighbors (KNN), and logistic regression (LR). The models' performance metrics included accuracy, area under the curve (AUC) of the receiver operating characteristic curve, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). In the training cohort, the RF predictive model surpassed LR, SVM, DT, and KNN in terms of accuracy, AUC, sensitivity, specificity, PPV, and NPV. Compared to the other four predictive models, the RF model significantly improved PPV. In the test cohort, all five machine learning predictive models exhibited lower PPVs. The DT model demonstrated the most significant variation in performance metrics relative to the other models, with a sensitivity of 0.231 and specificity of 0.990. The LR-based predictive model had the lowest PPV at 0.210, compared to the other four models. In the external validation cohort, the performance metrics of the predictive models were generally consistent with those in the test cohort. The LR-based model for predicting omental metastasis exhibited a lower PPV. Among the machine learning algorithms, the RF predictive model demonstrated higher accuracy and improved PPV relative to LR, SVM, KNN, and DT models.
Collapse
Affiliation(s)
- Ahao Wu
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lianghua Luo
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
- General Surgery Department of Jiangxi Provincial People's Hospital, Nanchang, 330006, Jiangxi Province, China
| | - Qingwen Zeng
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Changlei Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xufeng Shu
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Pang Huang
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zhonghao Wang
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Tengcheng Hu
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zongfeng Feng
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yanyan Zhu
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yi Cao
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Zhengrong Li
- Department of Digestive Surgery, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Department of Digestive Surgery, Digestive Disease Hospital, The Third Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
106
|
Wang YRJ, Wang P, Yan Z, Zhou Q, Gunturkun F, Li P, Hu Y, Wu WE, Zhao K, Zhang M, Lv H, Fu L, Jin J, Du Q, Wang H, Chen K, Qu L, Lin K, Iv M, Wang H, Sun X, Vogel H, Han S, Tian L, Wu F, Gong J. Advancing presurgical non-invasive molecular subgroup prediction in medulloblastoma using artificial intelligence and MRI signatures. Cancer Cell 2024; 42:1239-1257.e7. [PMID: 38942025 DOI: 10.1016/j.ccell.2024.06.002] [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: 08/18/2023] [Revised: 04/25/2024] [Accepted: 06/05/2024] [Indexed: 06/30/2024]
Abstract
Global investigation of medulloblastoma has been hindered by the widespread inaccessibility of molecular subgroup testing and paucity of data. To bridge this gap, we established an international molecularly characterized database encompassing 934 medulloblastoma patients from thirteen centers across China and the United States. We demonstrate how image-based machine learning strategies have the potential to create an alternative pathway for non-invasive, presurgical, and low-cost molecular subgroup prediction in the clinical management of medulloblastoma. Our robust validation strategies-including cross-validation, external validation, and consecutive validation-demonstrate the model's efficacy as a generalizable molecular diagnosis classifier. The detailed analysis of MRI characteristics replenishes the understanding of medulloblastoma through a nuanced radiographic lens. Additionally, comparisons between East Asia and North America subsets highlight critical management implications. We made this comprehensive dataset, which includes MRI signatures, clinicopathological features, treatment variables, and survival data, publicly available to advance global medulloblastoma research.
Collapse
Affiliation(s)
- Yan-Ran Joyce Wang
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Stanford University, Stanford, CA 94304, USA.
| | - Pengcheng Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Zihan Yan
- Department of Pediatric Neurosurgery, Beijing Tiantan Hospital, Capital Medicine University, Beijing Neurosurgical Institute, Beijing 100070, China
| | - Quan Zhou
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Neurosurgery, Stanford School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Fatma Gunturkun
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Peng Li
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Yanshen Hu
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Wei Emma Wu
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Radiology Oncology, Stanford University, Stanford, CA 94305, USA
| | - Kankan Zhao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Michael Zhang
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Neurosurgery, Stanford School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Haoyi Lv
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Lehao Fu
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Jiajie Jin
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Qing Du
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Haoyu Wang
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Kun Chen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Liangqiong Qu
- The Department of Statistics and Actuarial Science and the Institute of Data Science, The University of Hong Kong, Hong Kong 999077, China
| | - Keldon Lin
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ 85054, USA
| | - Michael Iv
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Neurosurgery, Stanford School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Hao Wang
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoyan Sun
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Hannes Vogel
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Summer Han
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Lu Tian
- School of Medicine, Stanford University, Stanford, CA 94304, USA; Department of Statistics, Stanford School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Feng Wu
- School of Engineering, University of Science and Technology of China, Hefei 230001, China
| | - Jian Gong
- Department of Pediatric Neurosurgery, Beijing Tiantan Hospital, Capital Medicine University, Beijing Neurosurgical Institute, Beijing 100070, China.
| |
Collapse
|
107
|
Masson-Grehaigne C, Lafon M, Palussière J, Leroy L, Bonhomme B, Jambon E, Italiano A, Cousin S, Crombé A. Enhancing Immunotherapy Response Prediction in Metastatic Lung Adenocarcinoma: Leveraging Shallow and Deep Learning with CT-Based Radiomics across Single and Multiple Tumor Sites. Cancers (Basel) 2024; 16:2491. [PMID: 39001553 PMCID: PMC11240700 DOI: 10.3390/cancers16132491] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
This study aimed to evaluate the potential of pre-treatment CT-based radiomics features (RFs) derived from single and multiple tumor sites, and state-of-the-art machine-learning survival algorithms, in predicting progression-free survival (PFS) for patients with metastatic lung adenocarcinoma (MLUAD) receiving first-line treatment including immune checkpoint inhibitors (CPIs). To do so, all adults with newly diagnosed MLUAD, pre-treatment contrast-enhanced CT scan, and performance status ≤ 2 who were treated at our cancer center with first-line CPI between November 2016 and November 2022 were included. RFs were extracted from all measurable lesions with a volume ≥ 1 cm3 on the CT scan. To capture intra- and inter-tumor heterogeneity, RFs from the largest tumor of each patient, as well as lowest, highest, and average RF values over all lesions per patient were collected. Intra-patient inter-tumor heterogeneity metrics were calculated to measure the similarity between each patient lesions. After filtering predictors with univariable Cox p < 0.100 and analyzing their correlations, five survival machine-learning algorithms (stepwise Cox regression [SCR], LASSO Cox regression, random survival forests, gradient boosted machine [GBM], and deep learning [Deepsurv]) were trained in 100-times repeated 5-fold cross-validation (rCV) to predict PFS on three inputs: (i) clinicopathological variables, (ii) all radiomics-based and clinicopathological (full input), and (iii) uncorrelated radiomics-based and clinicopathological variables (uncorrelated input). The Models' performances were evaluated using the concordance index (c-index). Overall, 140 patients were included (median age: 62.5 years, 36.4% women). In rCV, the highest c-index was reached with Deepsurv (c-index = 0.631, 95%CI = 0.625-0.647), followed by GBM (c-index = 0.603, 95%CI = 0.557-0.646), significantly outperforming standard SCR whatever its input (c-index range: 0.560-0.570, all p < 0.0001). Thus, single- and multi-site pre-treatment radiomics data provide valuable prognostic information for predicting PFS in MLUAD patients undergoing first-line CPI treatment when analyzed with advanced machine-learning survival algorithms.
Collapse
Affiliation(s)
- Cécile Masson-Grehaigne
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
| | - Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Jean Palussière
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
| | - Laura Leroy
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Benjamin Bonhomme
- Department of Biopathology, Institut Bergonié, F-33076 Bordeaux, France
| | - Eva Jambon
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
- SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, F-33076 Bordeaux, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Amandine Crombé
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
- SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, F-33076 Bordeaux, France
| |
Collapse
|
108
|
Szymaszek P, Tyszka-Czochara M, Ortyl J. Application of Photoactive Compounds in Cancer Theranostics: Review on Recent Trends from Photoactive Chemistry to Artificial Intelligence. Molecules 2024; 29:3164. [PMID: 38999115 PMCID: PMC11243723 DOI: 10.3390/molecules29133164] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
According to the World Health Organization (WHO) and the International Agency for Research on Cancer (IARC), the number of cancer cases and deaths worldwide is predicted to nearly double by 2030, reaching 21.7 million cases and 13 million fatalities. The increase in cancer mortality is due to limitations in the diagnosis and treatment options that are currently available. The close relationship between diagnostics and medicine has made it possible for cancer patients to receive precise diagnoses and individualized care. This article discusses newly developed compounds with potential for photodynamic therapy and diagnostic applications, as well as those already in use. In addition, it discusses the use of artificial intelligence in the analysis of diagnostic images obtained using, among other things, theranostic agents.
Collapse
Affiliation(s)
- Patryk Szymaszek
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Kraków, Poland
| | | | - Joanna Ortyl
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Kraków, Poland
- Photo HiTech Ltd., Bobrzyńskiego 14, 30-348 Kraków, Poland
- Photo4Chem Ltd., Juliusza Lea 114/416A-B, 31-133 Cracow, Poland
| |
Collapse
|
109
|
Yan Y, He X, Xu Y, Zhao F, Peng J, Shao Y. Pattern of heterogeneity in normal brain ageing: screening for mild cognitive impairment and its risk of progression with a radiomics model. Age Ageing 2024; 53:afae140. [PMID: 38984695 DOI: 10.1093/ageing/afae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/01/2024] [Indexed: 07/11/2024] Open
Abstract
PURPOSE This study aimed to develop a normal brain ageing model based on magnetic resonance imaging and radiomics, therefore identifying radscore, an imaging indicator representing white matter heterogeneity and exploring the significance of radscore in detecting people's cognitive changes. METHODS Three hundred sixty cognitively normal (CN) subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database and 105 CN subjects from the Parkinson's Progression Markers Initiative database were used to develop the model. In ADNI, 230 mild cognitive impairment (MCI) subjects were matched with 230 CN old-aged subjects to evaluate their heterogeneity difference. One hundred four MCI subjects with 48 months of follow-up were divided into low and high heterogeneity groups. Kaplan-Meier survival curve analysis was used to observe the importance of heterogeneity results for predicting MCI progression. RESULTS The area under the receiver operating characteristic curve of the model in the training, internal test and external test sets was 0.7503, 0.7512 and 0.7514, respectively. There was a significantly positive correlation between age and radscore of CN subjects (r = 0.501; P < .001). The radscore of MCI subjects was significantly higher than that of matched CN subjects (P < .001). The median radscore ratios of MCI to CN from four age groups (66-70y, 71-75y, 76-80y and 81-85y) were 1.611, 1.760, 1.340 and 1.266, respectively. The probability to progression of low and high heterogeneity groups had a significant difference (P = .002). CONCLUSION When radscore is significantly higher than that of normal ageing, it is necessary to alert the possibility of cognitive impairment and deterioration.
Collapse
Affiliation(s)
- Yuting Yan
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Xiaodong He
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Yuyun Xu
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Fanfan Zhao
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Jiaxuan Peng
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Yuan Shao
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| |
Collapse
|
110
|
Anurogo D, Luthfiana D, Anripa N, Fauziah AI, Soleha M, Rahmah L, Ratnawati H, Wargasetia TL, Pratiwi SE, Siregar RN, Sholichah RN, Maulana MS, Ikrar T, Chang YH, Qiu JT. The Art of Bioimmunogenomics (BIGs) 5.0 in CAR-T Cell Therapy for Lymphoma Management. Adv Pharm Bull 2024; 14:314-330. [PMID: 39206402 PMCID: PMC11347730 DOI: 10.34172/apb.2024.034] [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: 03/17/2023] [Revised: 01/13/2024] [Accepted: 03/03/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Lymphoma, the most predominant neoplastic disorder, is divided into Hodgkin and Non-Hodgkin Lymphoma classifications. Immunotherapeutic modalities have emerged as essential methodologies in combating lymphoid malignancies. Chimeric Antigen Receptor (CAR) T cells exhibit promising responses in chemotherapy-resistant B-cell non-Hodgkin lymphoma cases. Methods This comprehensive review delineates the advancement of CAR-T cell therapy as an immunotherapeutic instrument, the selection of lymphoma antigens for CAR-T cell targeting, and the conceptualization, synthesis, and deployment of CAR-T cells. Furthermore, it encompasses the advantages and disadvantages of CAR-T cell therapy and the prospective horizons of CAR-T cells from a computational research perspective. In order to improve the design and functionality of artificial CARs, there is a need for TCR recognition investigation, followed by the implementation of a quality surveillance methodology. Results Various lymphoma antigens are amenable to CAR-T cell targeting, such as CD19, CD20, CD22, CD30, the kappa light chain, and ROR1. A notable merit of CAR-T cell therapy is the augmentation of the immune system's capacity to generate tumoricidal activity in patients exhibiting chemotherapy-resistant lymphoma. Nevertheless, it also introduces manufacturing impediments that are laborious, technologically demanding, and financially burdensome. Physical, physicochemical, and physiological limitations further exacerbate the challenge of treating solid neoplasms with CAR-T cells. Conclusion While the efficacy and safety of CAR-T cell immunotherapy remain subjects of fervent investigation, the promise of this cutting-edge technology offers valuable insights for the future evolution of lymphoma treatment management approaches. Moreover, CAR-T cell therapies potentially benefit patients, motivating regulatory bodies to foster international collaboration.
Collapse
Affiliation(s)
- Dito Anurogo
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Faculty of Medicine and Health Sciences, Muhammadiyah University of Makassar, Makassar, South Sulawesi, 90221, Indonesia
| | - Dewi Luthfiana
- Bioinformatics Research Center, Indonesian Institute of Bioinformatics (INBIO), Malang, East Java, 65162, Indonesia
| | - Nuralfin Anripa
- Department of Environmental Science, Dumoga University, Kotamobagu, South Sulawesi, 95711, Indonesia
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Apriliani Ismi Fauziah
- MSc Program in Tropical Medicine, Kaohsiung Medical University, Kaohsiung City, 807378, Taiwan
| | - Maratu Soleha
- National Research and Innovation Agency (BRIN), Central Jakarta, 10340, Indonesia
- IKIFA College of Health Sciences, East Jakarta, Special Capital Region of Jakarta, 13470, Indonesia
| | - Laila Rahmah
- Department of Digital Health, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416634793, Iran
- Faculty of Medicine, Muhammadiyah University of Surabaya, Surabaya, East Java, 60113, Indonesia
| | - Hana Ratnawati
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia
| | | | - Sari Eka Pratiwi
- Department of Biology and Pathobiology, Faculty of Medicine, Tanjungpura University, Pontianak, West Kalimantan, 78115, Indonesia
| | - Riswal Nafi Siregar
- National Research and Innovation Agency (BRIN), Central Jakarta, 10340, Indonesia
| | - Ratis Nour Sholichah
- Department of Biotechnology, Postgraduate School of Gadjah Mada University, Yogyakarta, 55284, Indonesia
| | - Muhammad Sobri Maulana
- Community Health Center (Puskesmas) Temon 1, Kulon Progo, Special Region of Yogyakarta, 55654, Indonesia
| | - Taruna Ikrar
- Director of Members-at-Large, International Association of Medical Regulatory Authorities (IAMRA), Texas, 76039, USA
- Aivita Biomedical Inc., Irvine, California, 92612, USA
- Chairman of Medical Council, The Indonesian Medical Council (KKI), Central Jakarta, 10350, Indonesia
- Adjunct Professor, School of Military Medicine, The Republic of Indonesia Defense University (RIDU), Jakarta Pusat, 10440, Indonesia
- Department of Pharmacology, Faculty of Medicine, Malahayati University, Bandar Lampung, Lampung, 35152, Indonesia
| | - Yu Hsiang Chang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Locus Cell Co., LTD., Xizhi Dist., New Taipei City, 221, Taiwan
| | - Jiantai Timothy Qiu
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 110301, Taiwan
| |
Collapse
|
111
|
Xi Z, Ye Y, Yang Y, He Y, Song Z, Ma Q, Zeng H, Shao G. Radiomics analysis based on contrast-enhanced MRI for predicting short-term efficacy of drug-eluting beads transarterial chemoembolization in hepatocellular carcinoma. Abdom Radiol (NY) 2024; 49:2387-2400. [PMID: 39030402 DOI: 10.1007/s00261-024-04319-3] [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: 10/07/2023] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 07/21/2024]
Abstract
OBJECTIVE We developed and validated a clinical-radiomics model for preoperative prediction of the short-term efficacy of initial drug-eluting beads transarterial chemoembolization (D-TACE) treatment in patients with hepatocellular carcinoma (HCC). METHODS In this retrospective cohort study of 113 patients with intermediate and advanced HCC, 5343 features were extracted based on three sequences of the arterial phase (AP), diffusion-weighted imaging, and T2-weighted images based on contrast-enhanced magnetic resonance imaging, and minimum redundancy maximum correlation and least absolute shrinkage and selection operator (LASSO) regression were applied for feature selection and model construction. Multifactor logistic regression was used to build a clinical-imaging model based on clinical factors and a clinical-radiomics model. The area under the curve (AUC) and calibration curves were used to assess model performance, and the clinical value of the model was analyzed using decision curve analysis. The relationship between the actual and predicted short-term efficacy of the combined model and progression-free survival (PFS) was evaluated using Kaplan-Meier survival curves and log-rank tests. RESULTS A total of 34 radiomics features were selected by LASSO, and the clinical-radiomics model had the best predictive performance (AUC = 0.902 and AUC = 0.845 for the training and testing sets, respectively), and the model based on AP had the best predictive performance among the four radiomics models (AUC = 0.89 for the training set and AUC = 0.85 for the testing set); the multifactorial logistic regression results showed that microsphere type (p = 0.042) and AP Rad-score (p = 0.01) were associated with short-term efficacy. In addition, a difference in PFS was observed in patients with HCC with different short-term efficacies predicted by the combined model. Moreover, prognosis was better in the objective versus non-objective response group. CONCLUSIONS The combined clinical-radiomics model is an effective predictor of the short-term efficacy of initial D-TACE in patients with HCC, contributing to clinical and economic benefits for patients.
Collapse
Affiliation(s)
- Zihan Xi
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Yuanxin Ye
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yongbo Yang
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yiwei He
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Ziyang Song
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qian Ma
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, 325035, China
| | - Hui Zeng
- Department of Intervention, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Guoliang Shao
- Department of Intervention, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
112
|
Holder AM, Dedeilia A, Sierra-Davidson K, Cohen S, Liu D, Parikh A, Boland GM. Defining clinically useful biomarkers of immune checkpoint inhibitors in solid tumours. Nat Rev Cancer 2024; 24:498-512. [PMID: 38867074 DOI: 10.1038/s41568-024-00705-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Although more than a decade has passed since the approval of immune checkpoint inhibitors (ICIs) for the treatment of melanoma and non-small-cell lung, breast and gastrointestinal cancers, many patients still show limited response. US Food and Drug Administration (FDA)-approved biomarkers include programmed cell death 1 ligand 1 (PDL1) expression, microsatellite status (that is, microsatellite instability-high (MSI-H)) and tumour mutational burden (TMB), but these have limited utility and/or lack standardized testing approaches for pan-cancer applications. Tissue-based analytes (such as tumour gene signatures, tumour antigen presentation or tumour microenvironment profiles) show a correlation with immune response, but equally, these demonstrate limited efficacy, as they represent a single time point and a single spatial assessment. Patient heterogeneity as well as inter- and intra-tumoural differences across different tissue sites and time points represent substantial challenges for static biomarkers. However, dynamic biomarkers such as longitudinal biopsies or novel, less-invasive markers such as blood-based biomarkers, radiomics and the gut microbiome show increasing potential for the dynamic identification of ICI response, and patient-tailored predictors identified through neoadjuvant trials or novel ex vivo tumour models can help to personalize treatment. In this Perspective, we critically assess the multiple new static, dynamic and patient-specific biomarkers, highlight the newest consortia and trial efforts, and provide recommendations for future clinical trials to make meaningful steps forwards in the field.
Collapse
Affiliation(s)
- Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sonia Cohen
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - David Liu
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Aparna Parikh
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
113
|
Shetty S, Alvarado PC, Pettie D, Collier JH. Next-Generation Vaccine Development with Nanomaterials: Recent Advances, Possibilities, and Challenges. Annu Rev Biomed Eng 2024; 26:273-306. [PMID: 38959389 DOI: 10.1146/annurev-bioeng-110122-124359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Nanomaterials are becoming important tools for vaccine development owing to their tunable and adaptable nature. Unique properties of nanomaterials afford opportunities to modulate trafficking through various tissues, complement or augment adjuvant activities, and specify antigen valency and display. This versatility has enabled recent work designing nanomaterial vaccines for a broad range of diseases, including cancer, inflammatory diseases, and various infectious diseases. Recent successes of nanoparticle vaccines during the coronavirus disease 2019 (COVID-19) pandemic have fueled enthusiasm further. In this review, the most recent developments in nanovaccines for infectious disease, cancer, inflammatory diseases, allergic diseases, and nanoadjuvants are summarized. Additionally, challenges and opportunities for clinical translation of this unique class of materials are discussed.
Collapse
Affiliation(s)
- Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Pablo Cordero Alvarado
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Deleah Pettie
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| |
Collapse
|
114
|
Katifelis H, Gazouli M. RNA biomarkers in cancer therapeutics: The promise of personalized oncology. Adv Clin Chem 2024; 123:179-219. [PMID: 39181622 DOI: 10.1016/bs.acc.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cancer therapy is a rapidly evolving and constantly expanding field. Current approaches include surgery, conventional chemotherapy and novel biologic agents as in immunotherapy, that together compose a wide armamentarium. The plethora of choices can, however, be clinically challenging in prescribing the most suitable treatment for any given patient. Fortunately, biomarkers can greatly facilitate the most appropriate selection. In recent years, RNA-based biomarkers have proven most promising. These molecules that range from small noncoding RNAs to protein coding gene transcripts can be valuable in cancer management and especially in cancer therapeutics. Compared to their DNA counterparts which are stable throughout treatment, RNA-biomarkers are dynamic. This allows prediction of success prior to treatment start and can identify alterations in expression that could reflect response. Moreover, improved nucleic acid technology allows RNA to be extracted from practically every biofluid/matrix and evaluated with exceedingly high analytic sensitivity. In addition, samples are largely obtained by minimally invasive procedures and as such can be used serially to assess treatment response real-time. This chapter provides the reader insight on currently known RNA biomarkers, the latest research employing Artificial Intelligence in the identification of such molecules and in clinical decisions driving forward the era of personalized oncology.
Collapse
Affiliation(s)
- Hector Katifelis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
115
|
Jeon H, Gor R, D’Aiello A, Stiles B, Illei PB, Halmos B. Advancing neoadjuvant therapies in resectable non-small cell lung cancer: implications for novel treatment strategies and biomarker discovery. Pathol Oncol Res 2024; 30:1611817. [PMID: 38957347 PMCID: PMC11217184 DOI: 10.3389/pore.2024.1611817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024]
Abstract
The delivery of neoadjuvant and perioperative therapies for non-small cell lung cancer has been radically altered by significant advances and by the incorporation of targeted therapies as well as immune checkpoint inhibitors alone or alongside conventional chemotherapy. This evolution has been particularly notable in the incorporation of immunotherapy and targeted therapy into the treatment of resectable NSCLC, where recent FDA approvals of drugs such as nivolumab and pembrolizumab, in combination with platinum doublet chemotherapy, have led to considerable improvements in pathological complete response rates and the potential for enhanced long-term survival outcomes. This review emphasizes the growing importance of biomarkers in optimizing treatment selection and explores the impact of emerging studies that challenge existing treatment paradigms and investigate novel therapeutic combinations poised to redefine standard of care practices. Furthermore, the discussion extends to the unmet needs within perioperative treatment assessment and prognostication, highlighting the prospective value of biomarkers in evaluating treatment responses and prognosis.
Collapse
Affiliation(s)
- Hyein Jeon
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rajvi Gor
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States
| | - Angelica D’Aiello
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Brendon Stiles
- Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Peter B. Illei
- Department of Pathology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
116
|
Gao X, Chen H, Wang Y, Xu F, Zhang A, Yang Y, Gu Y. Automatic prediction of non-iodine-avid status in lung metastases for radioactive I 131 treatment in differentiated thyroid cancer patients. Front Endocrinol (Lausanne) 2024; 15:1429115. [PMID: 38933823 PMCID: PMC11201526 DOI: 10.3389/fendo.2024.1429115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Objectives The growing incidence of differentiated thyroid cancer (DTC) have been linked to insulin resistance and metabolic syndrome. The imperative need for developing effective diagnostic imaging tools to predict the non-iodine-avid status of lung metastasis (LMs) in differentiated thyroid cancer (DTC) patients is underscored to prevent unnecessary radioactive iodine treatment (RAI). Methods Primary cohort consisted 1962 pretreated LMs of 496 consecutive DTC patients with pretreated initially diagnosed LMs who underwent chest CT and subsequent post-treatment radioiodine SPECT. After automatic lesion segmentation by SE V-Net, SE Net deep learning was trained to predict non-iodine-avid status of LMs. External validation cohort contained 123 pretreated LMs of 24 consecutive patients from other two hospitals. Stepwise validation was further performed according to the nodule's largest diameter. Results The SE-Net deep learning network yielded area under the receiver operating characteristic curve (AUC) values of 0.879 (95% confidence interval: 0.852-0.906) and 0.713 (95% confidence interval: 0.613-0.813) for internal and external validation. With the LM diameter decreasing from ≥10mm to ≤4mm, the AUCs remained relatively stable, for smallest nodules (≤4mm), the model yielded an AUC of 0.783. Decision curve analysis showed that most patients benefited using deep learning to decide radioactive I131 treatment. Conclusion This study presents a noninvasive, less radioactive and fully automatic approach that can facilitate suitable DTC patient selection for RAI therapy of LMs. Further prospective multicenter studies with larger study cohorts and related metabolic factors should address the possibility of comprehensive clinical transformation.
Collapse
Affiliation(s)
- Xinyi Gao
- Shanghai Institute of Medical Imaging, Fenglin Road, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Dongan Road, Shanghai, China
- Department of Radiology, Zhejiang Cancer Hospital, Banshan East Road, Hangzhou, Zhejiang, China
| | - Haoyi Chen
- Hangzhou Dianzi University, Baiyang, Qiantang, Hangzhou, Zhejiang, China
| | - Yun Wang
- Department of Nuclear medicine, Zhejiang Cancer Hospital, Banshan East Road, Hangzhou, Zhejiang, China
| | - Feijia Xu
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Anni Zhang
- Department of Radiology, The First People’s Hospital of Fuyang, Beihuan Road, Hangzhou, Zhejiang, China
| | - Yong Yang
- Hangzhou Dianzi University, Baiyang, Qiantang, Hangzhou, Zhejiang, China
| | - Yajia Gu
- Shanghai Institute of Medical Imaging, Fenglin Road, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Dongan Road, Shanghai, China
| |
Collapse
|
117
|
Xu L, Wu Y, Shen X, Zhou L, Lu Y, Teng Z, Du J, Ding M, Han H, Niu T. Exploring the biological basis of CT imaging features in pancreatic neuroendocrine tumors: a two-center study. Phys Med Biol 2024; 69:125013. [PMID: 38810631 DOI: 10.1088/1361-6560/ad51c7] [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: 01/08/2024] [Accepted: 05/29/2024] [Indexed: 05/31/2024]
Abstract
Objective.Medical imaging offered a non-invasive window to visualize tumors, with radiomics transforming these images into quantitative data for tumor phenotyping. However, the intricate web linking imaging features, clinical endpoints, and tumor biology was mostly uncharted. This study aimed to unravel the connections between CT imaging features and clinical characteristics, including tumor histopathological grading, clinical stage, and endocrine symptoms, alongside immunohistochemical markers of tumor cell growth, such as the Ki-67 index and nuclear mitosis rate.Approach.We conducted a retrospective analysis of data from 137 patients with pancreatic neuroendocrine tumors who had undergone contrast-enhanced CT scans across two institutions. Our study focused on three clinical factors: pathological grade, clinical stage, and endocrine symptom status, in addition to two immunohistochemical markers: the Ki-67 index and the rate of nuclear mitosis. We computed both predefined (2D and 3D) and learning-based features (via sparse autoencoder, or SAE) from the scans. To unearth the relationships between imaging features, clinical factors, and immunohistochemical markers, we employed the Spearman rank correlation along with the Benjamini-Hochberg method. Furthermore, we developed and validated radiomics signatures to foresee these clinical factors.Main results.The 3D imaging features showed the strongest relationships with clinical factors and immunohistochemical markers. For the association with pathological grade, the mean absolute value of the correlation coefficient (CC) of 2D, SAE, and 3D features was 0.3318 ± 0.1196, 0.2149 ± 0.0361, and 0.4189 ± 0.0882, respectively. While for the association with Ki-67 index and rate of nuclear mitosis, the 3D features also showed higher correlations, with CC as 0.4053 ± 0.0786 and 0.4061 ± 0.0806. In addition, the 3D feature-based signatures showed optimal performance in clinical factor prediction.Significance.We found relationships between imaging features, clinical factors, and immunohistochemical markers. The 3D features showed higher relationships with clinical factors and immunohistochemical markers.
Collapse
Affiliation(s)
- Lei Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, People's Republic of China
| | - Yan Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaoyong Shen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Luping Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yongkai Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Ze Teng
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jichen Du
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| | - Mingchao Ding
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| | - Hongbin Han
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Tianye Niu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, People's Republic of China
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| |
Collapse
|
118
|
Li M, Yuan Y, Zhou H, Feng F, Xu G. A multicenter study: predicting KRAS mutation and prognosis in colorectal cancer through a CT-based radiomics nomogram. Abdom Radiol (NY) 2024; 49:1816-1828. [PMID: 38393357 DOI: 10.1007/s00261-024-04218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE To establish a CT-based radiomics nomogram for preoperative prediction of KRAS mutation and prognostic stratification in colorectal cancer (CRC) patients. METHODS In a retrospective analysis, 408 patients with confirmed CRC were included, comprising 168 cases in the training set, 111 cases in the internal validation set, and 129 cases in the external validation set. Radiomics features extracted from the primary tumors were meticulously screened to identify those closely associated with KRAS mutation. Subsequently, a radiomics nomogram was constructed by integrating these radiomics features with clinically significant parameters. The diagnostic performance was assessed through the area under the receiver operating characteristic curve (AUC). Lastly, the prognostic significance of the nomogram was explored, and Kaplan-Meier analysis was employed to depict survival curves for the high-risk and low-risk groups. RESULTS A radiomics model was constructed using 19 radiomics features significantly associated with KRAS mutation. Furthermore, a nomogram was developed by integrating these radiomics features with two clinically significant parameters (age, tumor location). The nomogram achieved AUCs of 0.834, 0.813, and 0.811 in the training set, internal validation set, and external validation set, respectively. Additionally, the nomogram effectively stratified patients into high-risk (KRAS mutation) and low-risk (KRAS wild-type) groups, demonstrating a significant difference in overall survival (P < 0.001). Patients categorized in the high-risk group exhibited inferior overall survival in contrast to those classified in the low-risk group. CONCLUSIONS The CT-based radiomics nomogram demonstrates the capability to effectively predict KRAS mutation in CRC patients and stratify their prognosis preoperatively.
Collapse
Affiliation(s)
- Manman Li
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Yiwen Yuan
- Department of Translational Medical Center, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Hui Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Guodong Xu
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
119
|
Jian Y, Peng J, Wang W, Hu T, Wang J, Shi H, Li X, Chen J, Xu Y, Shao Y, Song Q, Shu Z. Prediction of cognitive decline in Parkinson's disease based on MRI radiomics and clinical features: A multicenter study. CNS Neurosci Ther 2024; 30:e14789. [PMID: 38923776 PMCID: PMC11196371 DOI: 10.1111/cns.14789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/25/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE To develop and validate a multimodal combinatorial model based on whole-brain magnetic resonance imaging (MRI) radiomic features for predicting cognitive decline in patients with Parkinson's disease (PD). METHODS This study included a total of 222 PD patients with normal baseline cognition, of whom 68 had cognitive impairment during a 4-year follow-up period. All patients underwent MRI scans, and radiomic features were extracted from the whole-brain MRI images of the training set, and dimensionality reduction was performed to construct a radiomics model. Subsequently, Screening predictive factors for cognitive decline from clinical features and then combining those with a radiomics model to construct a multimodal combinatorial model for predicting cognitive decline in PD patients. Evaluate the performance of the comprehensive model using the receiver-operating characteristic curve, confusion matrix, F1 score, and survival curve. In addition, the quantitative characteristics of diffusion tensor imaging (DTI) from corpus callosum were selected from 52 PD patients to further validate the clinical efficacy of the model. RESULTS The multimodal combinatorial model has good classification performance, with areas under the curve of 0.842, 0.829, and 0.860 in the training, test, and validation sets, respectively. Significant differences were observed in the number of cognitive decline PD patients and corpus callosum-related DTI parameters between the low-risk and high-risk groups distinguished by the model (p < 0.05). The survival curve analysis showed a statistically significant difference in the progression time of mild cognitive impairment between the low-risk and the high-risk groups. CONCLUSIONS The building of a multimodal combinatorial model based on radiomic features from MRI can predict cognitive decline in PD patients, thus providing adaptive strategies for clinical practice.
Collapse
Affiliation(s)
- Yongjie Jian
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College)HangzhouZhejiangChina
- Department of Radiology, Affiliated Hospital of Sichuan Nursing Vocational CollegeThe Third People's Hospital of Sichuan ProvinceChengduSichuanChina
| | - Jiaxuan Peng
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College)HangzhouZhejiangChina
| | - Wei Wang
- Department of RadiologyThe First Affiliated Hospital of Chongqing Medical and Pharmaceutical CollegeChongqingChina
| | - Tao Hu
- Department of Neurology, Affiliated Hospital of Sichuan Nursing Vocational CollegeThe Third People's Hospital of Sichuan ProvinceChengduSichuanChina
| | - Jing Wang
- Department of Medical TechnologySichuan Nursing Vocational CollegeChengduSichuanChina
| | - Hui Shi
- Department of Radiology, Affiliated Hospital of Sichuan Nursing Vocational CollegeThe Third People's Hospital of Sichuan ProvinceChengduSichuanChina
| | - Xiaoyong Li
- Department of Radiology, Affiliated Hospital of Sichuan Nursing Vocational CollegeThe Third People's Hospital of Sichuan ProvinceChengduSichuanChina
| | - Jingfang Chen
- Department of Radiology, Affiliated Hospital of Sichuan Nursing Vocational CollegeThe Third People's Hospital of Sichuan ProvinceChengduSichuanChina
| | - Yuyun Xu
- Center for Rehabilitation Medicine, Department of RadiologyZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Yuan Shao
- Center for Rehabilitation Medicine, Department of RadiologyZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Qiaowei Song
- Center for Rehabilitation Medicine, Department of RadiologyZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Zhenyu Shu
- Center for Rehabilitation Medicine, Department of RadiologyZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
120
|
Xu H, Li C, Zhang L, Ding Z, Lu T, Hu H. Immunotherapy efficacy prediction through a feature re-calibrated 2.5D neural network. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 249:108135. [PMID: 38569256 DOI: 10.1016/j.cmpb.2024.108135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND AND OBJECTIVE Lung cancer continues to be a leading cause of cancer-related mortality worldwide, with immunotherapy emerging as a promising therapeutic strategy for advanced non-small cell lung cancer (NSCLC). Despite its potential, not all patients experience benefits from immunotherapy, and the current biomarkers used for treatment selection possess inherent limitations. As a result, the implementation of imaging-based biomarkers to predict the efficacy of lung cancer treatments offers a promising avenue for improving therapeutic outcomes. METHODS This study presents an automatic system for immunotherapy efficacy prediction on the subjects with lung cancer, facilitating significant clinical implications. Our model employs an advanced 2.5D neural network that incorporates 2D intra-slice feature extraction and 3D inter-slice feature aggregation. We further present a lesion-focused prior to guide the re-calibration for intra-slice features, and a attention-based re-calibration for the inter-slice features. Finally, we design an accumulated back-propagation strategy to optimize network parameters in a memory-efficient fashion. RESULTS We demonstrate that the proposed method achieves impressive performance on an in-house clinical dataset, surpassing existing state-of-the-art models. Furthermore, the proposed model exhibits increased efficiency in inference for each subject on average. To further validate the effectiveness of our model and its components, we conducted comprehensive and in-depth ablation experiments and discussions. CONCLUSION The proposed model showcases the potential to enhance physicians' diagnostic performance due to its impressive performance in predicting immunotherapy efficacy, thereby offering significant clinical application value. Moreover, we conduct adequate comparison experiments of the proposed methods and existing advanced models. These findings contribute to our understanding of the proposed model's effectiveness and serve as motivation for future work in immunotherapy efficacy prediction.
Collapse
Affiliation(s)
- Haipeng Xu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350014, China.
| | - Chenxin Li
- Department of Electronic Engineering, The Chinese University of Hong Kong, Hong Kong 999077, SAR, China.
| | - Longfeng Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350014, China.
| | - Zhiyuan Ding
- School of Informatics, Xiamen University, Fujian 350014, China.
| | - Tao Lu
- Department of Radiology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fujian 350014, China.
| | - Huihua Hu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian 350014, China.
| |
Collapse
|
121
|
Kim CG, Hong MH, Kim D, Lee BH, Kim H, Ock CY, Kelly G, Bang YJ, Kim G, Lee JE, Kim C, Kim SH, Hong HJ, Park YM, Sim NS, Park H, Park JW, Lee CG, Kim KH, Park G, Jung I, Han D, Kim JH, Cha J, Lee I, Kang M, Song H, Oum C, Kim S, Kim S, Lim Y, Kim-Schulze S, Merad M, Yoon SO, Kim HJ, Koh YW, Kim HR. A Phase II Open-Label Randomized Clinical Trial of Preoperative Durvalumab or Durvalumab plus Tremelimumab in Resectable Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2024; 30:2097-2110. [PMID: 38457288 DOI: 10.1158/1078-0432.ccr-23-3249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/06/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024]
Abstract
PURPOSE Clinical implications of neoadjuvant immunotherapy in patients with locally advanced but resectable head and neck squamous cell carcinoma (HNSCC) remain largely unexplored. PATIENTS AND METHODS Patients with resectable HNSCC were randomized to receive a single dose of preoperative durvalumab (D) with or without tremelimumab (T) before resection, followed by postoperative (chemo)radiotherapy based on multidisciplinary discretion and 1-year D treatment. Artificial intelligence (AI)-powered spatial distribution analysis of tumor-infiltrating lymphocytes and high-dimensional profiling of circulating immune cells tracked dynamic intratumoral and systemic immune responses. RESULTS Of the 48 patients enrolled (D, 24 patients; D+T, 24 patients), 45 underwent surgical resection per protocol (D, 21 patients; D+T, 24 patients). D±T had a favorable safety profile and did not delay surgery. Distant recurrence-free survival (DRFS) was significantly better in patients treated with D+T than in those treated with D monotherapy. AI-powered whole-slide image analysis demonstrated that D+T significantly reshaped the tumor microenvironment toward immune-inflamed phenotypes, in contrast with the D monotherapy or cytotoxic chemotherapy. High-dimensional profiling of circulating immune cells revealed a significant expansion of T-cell subsets characterized by proliferation and activation in response to D+T therapy, which was rare following D monotherapy. Importantly, expansion of specific clusters in CD8+ T cells and non-regulatory CD4+ T cells with activation and exhaustion programs was associated with prolonged DRFS in patients treated with D+T. CONCLUSIONS Preoperative D±T is feasible and may benefit patients with resectable HNSCC. Distinct changes in the tumor microenvironment and circulating immune cells were induced by each treatment regimen, warranting further investigation.
Collapse
Affiliation(s)
- Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dahee Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Brian Hyohyoung Lee
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunwook Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Geoffrey Kelly
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yoon Ji Bang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gamin Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chaeyeon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Heon Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jun Hong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Min Park
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nam Suk Sim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heejung Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang Geol Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Goeun Park
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dawoon Han
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hoon Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junha Cha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | - Heon Song
- Lunit Inc., Seoul, Republic of Korea
| | | | | | | | | | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, New York
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Je Kim
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Republic of Korea
- Genome Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoon Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
122
|
Ren Z, Yang K, Zhu L, Yin D, Zhou Y. Regulatory T cells as crucial trigger and potential target for hyperprogressive disease subsequent to PD-1/PD-L1 blockade for cancer treatment. Int Immunopharmacol 2024; 132:111934. [PMID: 38574701 DOI: 10.1016/j.intimp.2024.111934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
PD-1/PD-L1 blockade therapy has brought great success to cancer treatment. Nevertheless, limited beneficiary populations and even hyperprogressive disease (HPD) greatly constrain the application of PD-1/PD-L1 inhibitors in clinical treatment. HPD is a special pattern of disease progression with rapid tumor growth and even serious consequences of patient death, which requires urgent attention. Among the many predisposing causes of HPD, regulatory T cells (Tregs) are suspected because they are amplified in cases of HPD. Tregs express PD-1 thus PD-1/PD-L1 blockade therapy may have an impact on Tregs which leads to HPD. Tregs are a subset of CD4+ T cells expressing FoxP3 and play critical roles in suppressing immunity. Tregs migrate toward tumors in the presence of chemokines to suppress antitumor immune responses, causing cancer cells to grow and proliferate. Studies have shown that deleting Tregs could enhance the efficacy of PD-1/PD-L1 blockade therapy and reduce the occurrence of HPD. This suggests that immunotherapy combined with Treg depletion may be an effective means of avoiding HPD. In this review, we summarized the immunosuppressive-related functions of Tregs in antitumor therapy and focused on advances in therapy combining Tregs depletion with PD-1/PD-L1 blockade in clinical studies. Moreover, we provided an outlook on Treg-targeted HPD early warning for PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Zhe Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Kaiqing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Detao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
123
|
Yu Y, Guo H, Zhang M, Hou F, Yang S, Huang C, Duan L, Wang H. Multi-institutional validation of a radiomics signature for identification of postoperative progression of soft tissue sarcoma. Cancer Imaging 2024; 24:59. [PMID: 38720384 PMCID: PMC11077743 DOI: 10.1186/s40644-024-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND To develop a magnetic resonance imaging (MRI)-based radiomics signature for evaluating the risk of soft tissue sarcoma (STS) disease progression. METHODS We retrospectively enrolled 335 patients with STS (training, validation, and The Cancer Imaging Archive sets, n = 168, n = 123, and n = 44, respectively) who underwent surgical resection. Regions of interest were manually delineated using two MRI sequences. Among 12 machine learning-predicted signatures, the best signature was selected, and its prediction score was inputted into Cox regression analysis to build the radiomics signature. A nomogram was created by combining the radiomics signature with a clinical model constructed using MRI and clinical features. Progression-free survival was analyzed in all patients. We assessed performance and clinical utility of the models with reference to the time-dependent receiver operating characteristic curve, area under the curve, concordance index, integrated Brier score, decision curve analysis. RESULTS For the combined features subset, the minimum redundancy maximum relevance-least absolute shrinkage and selection operator regression algorithm + decision tree classifier had the best prediction performance. The radiomics signature based on the optimal machine learning-predicted signature, and built using Cox regression analysis, had greater prognostic capability and lower error than the nomogram and clinical model (concordance index, 0.758 and 0.812; area under the curve, 0.724 and 0.757; integrated Brier score, 0.080 and 0.143, in the validation and The Cancer Imaging Archive sets, respectively). The optimal cutoff was - 0.03 and cumulative risk rates were calculated. DATA CONCLUSION To assess the risk of STS progression, the radiomics signature may have better prognostic power than a nomogram/clinical model.
Collapse
Affiliation(s)
- Yuan Yu
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China
| | - Hongwei Guo
- Department of Operation Center, Women and Children's Hospital, Qingdao University, Shandong, China
| | - Meng Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shifeng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chencui Huang
- Department of Research Collaboration, Research and Development (R&D) center, Beijing Deepwise & League of Philosophy Doctor (PHD) Technology Co., Ltd, Beijing, China
| | - Lisha Duan
- Department of Radiology, The Third Hospital of Hebei Medical University, Hebei, China.
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China.
| |
Collapse
|
124
|
Tang S, Fan T, Wang X, Yu C, Zhang C, Zhou Y. Cancer Immunotherapy and Medical Imaging Research Trends from 2003 to 2023: A Bibliometric Analysis. J Multidiscip Healthc 2024; 17:2105-2120. [PMID: 38736544 PMCID: PMC11086400 DOI: 10.2147/jmdh.s457367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose With the rapid development of immunotherapy, cancer treatment has entered a new phase. Medical imaging, as a primary diagnostic method, is closely related to cancer immunotherapy. However, until now, there has been no systematic bibliometric analysis of the state of this field. Therefore, the main purpose of this article is to clarify the past research trajectory, summarize current research hotspots, reveal dynamic scientific developments, and explore future research directions. Patients and Methods A comprehensive search was conducted on the Web of Science Core Collection (WoSCC) database to identify publications related to immunotherapy specifically for the medical imaging of carcinoma. The search spanned the period from the year 2003 to 2023. Several analytical tools were employed. These included CiteSpace (6.2.4), and the Microsoft Office Excel (2016). Results By searching the database, a total of 704 English articles published between 2003 and 2023 were obtained. We have observed a rapid increase in the number of publications since 2018. The two most active countries are the United States (n=265) and China (n=170). Pittock, Sean J and Abu-sbeih, Hamzah are very concerned about the relationship between cancer immunotherapy and medical images and have published more academic papers (n = 5; n = 4). Among the top 10 co-cited authors, Topalian Sl (n=43) cited ranked first, followed by Graus F (n=40) cited. According to clustering, timeline, and burst word analysis, the results show that the current research focus is on "MRI", "deep learning", "tumor microenvironment" and so on. Conclusion Medical imaging and cancer immunotherapy are hot topics. The United States is the country with the most publications and the greatest influence in this field, followed by China. "MRI", "PET/PET-CT", "deep learning", "immune-related adverse events" and "tumor microenvironment" are currently hot research topics and potential targets.
Collapse
Affiliation(s)
- Shuli Tang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Tiantian Fan
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Xinxin Wang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Can Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Chunhui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| | - Yang Zhou
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150010, People’s Republic of China
| |
Collapse
|
125
|
Weishan H, Donglin Z, Guangmei D, Wenya L, Fasheng W, Jibing C. Immunoradiotherapy for NSCLC: mechanisms, clinical outcomes, and future directions. Clin Transl Oncol 2024; 26:1063-1076. [PMID: 37921958 PMCID: PMC11026276 DOI: 10.1007/s12094-023-03337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2023]
Abstract
Non-small-cell lung cancer (NSCLC) has an extremely low 5-year survival rate, with the only effective treatment being immunoradiotherapy (iRT). Here, we review the progress of clinical research on iRT for non-small-cell lung cancer (NSCLC) over 2018-2023, as well as the future directions. We first discuss the synergistic mechanisms of iRT, reflected in three aspects: immune regulation of RT, RT-activated immune-related pathways, and RT-related immune sensitization. iRT may include either external-beam or stereotactic-body RT combined with either immune checkpoint inhibitors (e.g., immunoglobulins against immune programmed cell death (PD) 1/PD ligand 1 or CD8+ T lymphocyte antigen 4) or traditional Chinese medicine drugs. Regarding clinical effectiveness and safety, iRT increases overall and progression-free survival and tumor control rate among patients with NSCLC but without a considerable increase in toxicity risk. We finally discuss iRT challenges and future directions reported over 2018-2023.
Collapse
Affiliation(s)
- He Weishan
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zheng Donglin
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Deng Guangmei
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Liu Wenya
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wu Fasheng
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Chen Jibing
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|
126
|
Bo Z, Song J, He Q, Chen B, Chen Z, Xie X, Shu D, Chen K, Wang Y, Chen G. Application of artificial intelligence radiomics in the diagnosis, treatment, and prognosis of hepatocellular carcinoma. Comput Biol Med 2024; 173:108337. [PMID: 38547656 DOI: 10.1016/j.compbiomed.2024.108337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/17/2024] [Indexed: 04/17/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, with an increasing incidence and poor prognosis. In the past decade, artificial intelligence (AI) technology has undergone rapid development in the field of clinical medicine, bringing the advantages of efficient data processing and accurate model construction. Promisingly, AI-based radiomics has played an increasingly important role in the clinical decision-making of HCC patients, providing new technical guarantees for prediction, diagnosis, and prognostication. In this review, we evaluated the current landscape of AI radiomics in the management of HCC, including its diagnosis, individual treatment, and survival prognosis. Furthermore, we discussed remaining challenges and future perspectives regarding the application of AI radiomics in HCC.
Collapse
Affiliation(s)
- Zhiyuan Bo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiatao Song
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qikuan He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bo Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ziyan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaozai Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danyang Shu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kaiyu Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
127
|
Cui Y, Zhao K, Meng X, Mao Y, Han C, Shi Z, Yang X, Tong T, Wu L, Liu Z. A computed tomography-based multitask deep learning model for predicting tumour stroma ratio and treatment outcomes in patients with colorectal cancer: a multicentre cohort study. Int J Surg 2024; 110:2845-2854. [PMID: 38348900 PMCID: PMC11093466 DOI: 10.1097/js9.0000000000001161] [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: 10/17/2023] [Accepted: 01/26/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Tumour-stroma interactions, as indicated by tumour-stroma ratio (TSR), offer valuable prognostic stratification information. Current histological assessment of TSR is limited by tissue accessibility and spatial heterogeneity. The authors aimed to develop a multitask deep learning (MDL) model to noninvasively predict TSR and prognosis in colorectal cancer (CRC). MATERIALS AND METHODS In this retrospective study including 2268 patients with resected CRC recruited from four centres, the authors developed an MDL model using preoperative computed tomography (CT) images for the simultaneous prediction of TSR and overall survival. Patients in the training cohort ( n =956) and internal validation cohort (IVC, n =240) were randomly selected from centre I. Patients in the external validation cohort 1 (EVC1, n =509), EVC2 ( n =203), and EVC3 ( n =360) were recruited from other three centres. Model performance was evaluated with respect to discrimination and calibration. Furthermore, the authors evaluated whether the model could predict the benefit from adjuvant chemotherapy. RESULTS The MDL model demonstrated strong TSR discrimination, yielding areas under the receiver operating curves (AUCs) of 0.855 (95% CI, 0.800-0.910), 0.838 (95% CI, 0.802-0.874), and 0.857 (95% CI, 0.804-0.909) in the three validation cohorts, respectively. The MDL model was also able to predict overall survival and disease-free survival across all cohorts. In multivariable Cox analysis, the MDL score (MDLS) remained an independent prognostic factor after adjusting for clinicopathological variables (all P <0.05). For stage II and stage III disease, patients with a high MDLS benefited from adjuvant chemotherapy [hazard ratio (HR) 0.391 (95% CI, 0.230-0.666), P =0.0003; HR=0.467 (95% CI, 0.331-0.659), P <0.0001, respectively], whereas those with a low MDLS did not. CONCLUSION The multitask DL model based on preoperative CT images effectively predicted TSR status and survival in CRC patients, offering valuable guidance for personalized treatment. Prospective studies are needed to confirm its potential to select patients who might benefit from chemotherapy.
Collapse
Affiliation(s)
- Yanfen Cui
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University; Taiyuan
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou
| | - Yun Mao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing
| | - Chu Han
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
| | - Zhenwei Shi
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
| | - Xiaotang Yang
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University; Taiyuan
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Wu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences
| |
Collapse
|
128
|
Zhang GY, Du XZ, Xu R, Chen T, Wu Y, Wu XJ, Liu S. Development and Validation of a Machine Learning-Based Model Using CT Radiomics for Predicting Immune Checkpoint Inhibitor-related Pneumonitis in Patients With NSCLC Receiving Anti-PD1 Immunotherapy: A Multicenter Retrospective CaseControl Study. Acad Radiol 2024; 31:2128-2143. [PMID: 37977890 DOI: 10.1016/j.acra.2023.10.039] [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: 08/24/2023] [Revised: 10/06/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
RATIONALE AND OBJECTIVES This study aimed to develop and evaluate a radiomics-based model combined with clinical and qualitative radiological (semantic feature [SF]) features to predict immune checkpoint inhibitor-related pneumonitis (CIP) in patients with non-small cell lung cancer (NSCLC) treated with programmed cell death protein 1 inhibitors. MATERIALS AND METHODS This was a multicenter retrospective casecontrol study conducted from January 1, 2018, to December 31, 2022, at three centers. Patients with NSCLC treated with anti-PD1 were enrolled and randomly divided into two groups (7:3): training (n = 95) and validation (n = 39). Logistic regression (LR) and support vector machine (SVM) algorithms were used to transform features into the models. RESULTS The study comprised 134 participants from three independent centers (male, 114/134, 85%; mean [±standard deviation] age, 63.92 [±7.9] years). The radiomics score (RS) models built based on the LR and SVM algorithms could accurately predict CIP (area under the receiver operating characteristics curve [AUC], 0.860 [0.780, 0.939] and 0.861 [0.781, 0.941], respectively). The AUCs for the RS-clinic-SF combined model were 0.903 (0.839, 0.967) and 0.826 (0.688, 0.964) in the training and validation cohorts, respectively. Decision curve analysis showed that the combined models achieved high clinical net benefit across the majority of the range of reasonable threshold probabilities. CONCLUSION This study demonstrated that the combined model constructed by the identified features of RS, clinical features, and SF has the potential to precisely predict CIP. The RS-clinic-SF combined model has the potential to be used more widely as a practical tool for the noninvasive prediction of CIP to support individualized treatment planning.
Collapse
Affiliation(s)
- Guo-Yue Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Xian-Zhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Ting Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (T.C.).
| | - Yue Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.).
| | - Xiao-Juan Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China (G.-y.Z., X.-z.D., R.X., Y.W., X.-j.W.); Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, 629000, Sichuan, P.R. China (X.-j.W.).
| | - Shui Liu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fengjie, Fengjie, Chongqing, 404600, P.R. China (S.L.).
| |
Collapse
|
129
|
Zhang W, Song LN, You YF, Qi FN, Cui XH, Yi MX, Zhu G, Chang RA, Zhang HJ. Application of artificial intelligence in the prediction of immunotherapy efficacy in hepatocellular carcinoma: Current status and prospects. Artif Intell Gastroenterol 2024; 5:90096. [DOI: 10.35712/aig.v5.i1.90096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/28/2024] [Accepted: 03/12/2024] [Indexed: 04/29/2024] Open
Abstract
Artificial Intelligence (AI) has increased as a potent tool in medicine, with promising oncology applications. The emergence of immunotherapy has transformed the treatment terrain for hepatocellular carcinoma (HCC), offering new hope to patients with this challenging malignancy. This article examines the role and future of AI in forecasting the effectiveness of immunotherapy in HCC. We highlight the potential of AI to revolutionize the prediction of therapy response, thus improving patient selection and clinical outcomes. The article further outlines the challenges and future research directions in this emerging field.
Collapse
Affiliation(s)
- Wei Zhang
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Li-Ning Song
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yun-Fei You
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Feng-Nan Qi
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao-Hong Cui
- Department of General Surgery, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Ming-Xun Yi
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ren-An Chang
- Research Center of Clinical Medicine and Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Hai-Jian Zhang
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Research Center of Clinical Medicine, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
130
|
Huang K, Huang X, Zeng C, Wang S, Zhan Y, Cai Q, Peng G, Yang Z, Zhou L, Chen J, Chen C. Radiomics signature for dynamic changes of tumor-infiltrating CD8+ T cells and macrophages in cervical cancer during chemoradiotherapy. Cancer Imaging 2024; 24:54. [PMID: 38654284 PMCID: PMC11036574 DOI: 10.1186/s40644-024-00680-0] [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: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Our previous study suggests that tumor CD8+ T cells and macrophages (defined as CD68+ cells) infiltration underwent dynamic and heterogeneous changes during concurrent chemoradiotherapy (CCRT) in cervical cancer patients, which correlated with their short-term tumor response. This study aims to develop a CT image-based radiomics signature for such dynamic changes. METHODS Thirty cervical squamous cell carcinoma patients, who were treated with CCRT followed by brachytherapy, were included in this study. Pre-therapeutic CT images were acquired. And tumor biopsies with immunohistochemistry at primary sites were performed at baseline (0 fraction (F)) and immediately after 10F. Radiomics features were extracted from the region of interest (ROI) of CT images using Matlab. The LASSO regression model with ten-fold cross-validation was utilized to select features and construct an immunomarker classifier and a radiomics signature. Their performance was evaluated by the area under the curve (AUC). RESULTS The changes of tumor-infiltrating CD8+T cells and macrophages after 10F radiotherapy as compared to those at baseline were used to generate the immunomarker classifier (AUC= 0.842, 95% CI:0.680-1.000). Additionally, a radiomics signature was developed using 4 key radiomics features to predict the immunomarker classifier (AUC=0.875, 95% CI:0.753-0.997). The patients stratified based on this signature exhibited significant differences in treatment response (p = 0.004). CONCLUSION The radiomics signature could be used as a potential predictor for the CCRT-induced dynamic alterations of CD8+ T cells and macrophages, which may provide a less invasive approach to appraise tumor immune status during CCRT in cervical cancer compared to tissue biopsy.
Collapse
Affiliation(s)
- Kang Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
- Department of Radiation Oncology, Zhongshan City People's Hospital, Zhongshan, P.R. China
| | - Xuehan Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
- Shantou University Medical College, Shantou, P.R. China
| | - Chengbing Zeng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Siyan Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
- Shantou University Medical College, Shantou, P.R. China
| | - Yizhou Zhan
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Qingxin Cai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Guobo Peng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Zhining Yang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Li Zhou
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jianzhou Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China.
- Gustave Roussy Cancer Campus, Villejuif Cedex, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P.R. China.
| |
Collapse
|
131
|
Meng Z, Chen Y, Li H, Zhang Y, Yao X, Meng Y, Shi W, Liang Y, Hu Y, Liu D, Xie M, Yan B, Luo J. Machine learning and optical coherence tomography-derived radiomics analysis to predict persistent diabetic macular edema in patients undergoing anti-VEGF intravitreal therapy. J Transl Med 2024; 22:358. [PMID: 38627718 PMCID: PMC11022368 DOI: 10.1186/s12967-024-05141-7] [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: 10/08/2023] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Diabetic macular edema (DME) is a leading cause of vision loss in patients with diabetes. This study aimed to develop and evaluate an OCT-omics prediction model for assessing anti-vascular endothelial growth factor (VEGF) treatment response in patients with DME. METHODS A retrospective analysis of 113 eyes from 82 patients with DME was conducted. Comprehensive feature engineering was applied to clinical and optical coherence tomography (OCT) data. Logistic regression, support vector machine (SVM), and backpropagation neural network (BPNN) classifiers were trained using a training set of 79 eyes, and evaluated on a test set of 34 eyes. Clinical implications of the OCT-omics prediction model were assessed by decision curve analysis. Performance metrics (sensitivity, specificity, F1 score, and AUC) were calculated. RESULTS The logistic, SVM, and BPNN classifiers demonstrated robust discriminative abilities in both the training and test sets. In the training set, the logistic classifier achieved a sensitivity of 0.904, specificity of 0.741, F1 score of 0.887, and AUC of 0.910. The SVM classifier showed a sensitivity of 0.923, specificity of 0.667, F1 score of 0.881, and AUC of 0.897. The BPNN classifier exhibited a sensitivity of 0.962, specificity of 0.926, F1 score of 0.962, and AUC of 0.982. Similar discriminative capabilities were maintained in the test set. The OCT-omics scores were significantly higher in the non-persistent DME group than in the persistent DME group (p < 0.001). OCT-omics scores were also positively correlated with the rate of decline in central subfield thickness after treatment (Pearson's R = 0.44, p < 0.001). CONCLUSION The developed OCT-omics model accurately assesses anti-VEGF treatment response in DME patients. The model's robust performance and clinical implications highlight its utility as a non-invasive tool for personalized treatment prediction and retinal pathology assessment.
Collapse
Affiliation(s)
- Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Yanzhu Chen
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haoyu Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Yue Zhang
- Faculty of Infrastructure Engineering, Dalian University of Technology, Dalian, China
| | | | - Yongan Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Youling Liang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Yuqian Hu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Dan Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Manyun Xie
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China
| | - Bin Yan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China.
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, China.
| |
Collapse
|
132
|
Gao Y, Wu R, Pei Z, Ke C, Zeng D, Li X, Zhang Y. Cell cycle associated protein 1 associates with immune infiltration and ferroptosis in gastrointestinal cancer. Heliyon 2024; 10:e28794. [PMID: 38586390 PMCID: PMC10998105 DOI: 10.1016/j.heliyon.2024.e28794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Background Cell Cycle-Associated Protein 1 (CAPRIN1) play an important role in cell proliferation, oxidative stress, and inflammatory response. Nonetheless, its role in tumor immunity and ferroptosis is largely unknown in gastrointestinal cancer patients. Methods Through comprehensive bioinformatics, we investigate CAPRIN1 expression patterns and its role in diagnosis, functional signaling pathways, tumor immune infiltration and ferroptosis of different gastrointestinal cancer subtypes. Besides, immunohistochemistry (IHC) and immune blot were used to validate our esophagus cancer clinical data. The ferroptotic features of CAPRIN1 in vitro were assessed through knockdown assays in esophagus cancer cells. Results CAPRIN1 expression was significantly upregulated, correlated with poor prognosis, and served as an independent risk factor for most gastrointestinal cancer. Moreover, CAPRIN1 overexpression positively correlated with gene markers of most infiltrating immune cells, and immune checkpoints. CAPRIN1 knockdown significantly decreased the protein level of major histocompatibility complex class I molecules. We also identified a link between CAPRIN1 and ferroptosis-related genes in gastrointestinal cancer. Knockdown of CAPRIN1 significantly increased the production of lipid reactive oxygen species and malondialdehyde. Inhibition of CAPRIN1 expression promoted ferroptotic cell death induced by RAS-selective lethal 3 and erastin in human esophagus cancer cells. Conclusion Collectively, our results demonstrate that CAPRIN1 is aberrantly expressed in gastrointestinal cancer, is associated with poor prognosis, and could potentially influence immune infiltration and ferroptosis.
Collapse
Affiliation(s)
- Yan Gao
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Ruimin Wu
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhijun Pei
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Changbin Ke
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Daobing Zeng
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaohui Li
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
133
|
Chen C, Li Y, Liu H, Liao M, Yang J, Liu J. FAT1 upregulation is correlated with an immunosuppressive tumor microenvironment and predicts unfavorable outcome of immune checkpoint therapy in non-small cell lung cancer. Heliyon 2024; 10:e28356. [PMID: 38560204 PMCID: PMC10979093 DOI: 10.1016/j.heliyon.2024.e28356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 03/17/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Background Previous studies found that FAT1 was recurrently mutated and aberrantly expressed in multiple cancers, and the loss function of FAT1 promoted the formation of cancer-initiating cells in several cancers. However, in some types of cancer, FAT1 upregulation could lead to epithelial-mesenchymal transition (EMT). The role of FAT1 in cancer progression, which appears to be cancer-type-specific, is largely unknown. Methods QRT-PCR and immunochemistry were used to verify the expression of FAT1 in non-small cell lung cancer (NSCLC). QRT-PCR and Western blot were used to detect the influence of siFAT1 knockdown on the expression of potential targets of FAT1 in NSCLC cell lines. GEPIA, KM-plotter, CAMOIP, and ROC-Plotter were used to evaluate the association between FAT1 and clinical outcomes based on expression and clinical data from TCGA and immune checkpoint inhibitors (ICI) treated cohorts. Results We found that FAT1 upregulation was associated with the activation of TGF-β and EMT signaling pathways in NSCLC. Patients with a high FAT1 expression level tend to have a poor prognosis and hard to benefit from ICI therapy. Genes involved in TGF-β/EMT signaling pathways (SERPINE1, TGFB1/2, and POSTN) were downregulated upon knockdown of FAT1. Genomic and immunologic analysis showed that high cancer-associated fibroblast (CAF) abundance, decreased CD8+ T cells infiltration, and low TMB/TNB were correlated with the upregulation of FAT1, thus promoting an immunosuppressive tumor microenvironment (TME) which influence the effect of ICI-therapy. Conclusion Our findings revealed the pattern of FAT1 upregulation in the TME of patients with NSCLC, and demonstrated its utility as a biomarker for unfavorable clinical outcomes, thereby providing a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Chao Chen
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China
| | - Yanling Li
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Haozhen Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China
| | - Mengying Liao
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, 518035, China
| | - Jianyi Yang
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China
| |
Collapse
|
134
|
Xu J, Wang P, Li Y, Shi X, Yin T, Yu J, Teng F. Development and validation of an MRI-Based nomogram to predict the effectiveness of immunotherapy for brain metastasis in patients with non-small cell lung cancer. Front Immunol 2024; 15:1373330. [PMID: 38686383 PMCID: PMC11057328 DOI: 10.3389/fimmu.2024.1373330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction The variability and unpredictability of immune checkpoint inhibitors (ICIs) in treating brain metastases (BMs) in patients with advanced non-small cell lung cancer (NSCLC) is the main concern. We assessed the utility of novel imaging biomarkers (radiomics) for discerning patients with NSCLC and BMs who would derive advantages from ICIs treatment. Methods Data clinical outcomes and pretreatment magnetic resonance images (MRI) were collected on patients with NSCLC with BMs treated with ICIs between June 2019 and June 2022 and divided into training and test sets. Metastatic brain lesions were contoured using ITK-SNAP software, and 3748 radiomic features capturing both intra- and peritumoral texture patterns were extracted. A clinical radiomic nomogram (CRN) was built to evaluate intracranial progression-free survival, progression-free survival, and overall survival. The prognostic value of the CRN was assessed by Kaplan-Meier survival analysis and log-rank tests. Results In the study, a total of 174 patients were included, and 122 and 52 were allocated to the training and validation sets correspondingly. The intratumoral radiomic signature, peritumoral radiomic signature, clinical signature, and CRN predicted intracranial objective response rate. Kaplan-Meier analyses showed a significantly longer intracranial progression-free survival in the low-CRN group than in the high-CRN group (p < 0.001). The CRN was also significantly associated with progression-free survival (p < 0.001) but not overall survival. Discussion Radiomics biomarkers from pretreatment MRI images were predictive of intracranial response. Pretreatment radiomics may allow the early prediction of benefits.
Collapse
Affiliation(s)
- Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaonan Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
135
|
Fu M, Zhang X, Shen F, Ma J, Li Z. Prognostic value of peripheral blood neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, pan-immune-inflammation value and systemic immune-inflammation index for the efficacy of immunotherapy in patients with advanced gastric cancer. Immunotherapy 2024. [PMID: 38578121 DOI: 10.2217/imt-2024-0031] [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] [Indexed: 04/06/2024] Open
Abstract
Aim: The study aimed to assess the value of pretreatment peripheral blood neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), pan-immune-inflammation value (PIV) and systemic immune-inflammation index (SII) for predicting immunotherapy prognosis and efficacy in advanced gastric cancer (GC). Methods: A total of 84 advanced GC patients received immunotherapy were retrospectively collected. The optimal cut-off values were determined by receiver operating characteristic curves. The univariate and multivariate analysis investigated the effects of NLR, PLR, PIV and SII on patients prognosis. Results: NLR, PLR, PIV and SII had predictive value of efficacy. NLR ≥3.65 was an independent risk factor for worse outcomes. Conclusion: NLR, PLR, PIV and SII have predictive value of efficacy and NLR ≥3.65 suggests a poor prognosis following immunotherapy in advanced GC.
Collapse
Affiliation(s)
- Maodong Fu
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Xiuping Zhang
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Feng Shen
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Jun Ma
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Zhiyong Li
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| |
Collapse
|
136
|
Bartolomeo V, Cortiula F, Hendriks LEL, De Ruysscher D, Filippi AR. A Glimpse Into the Future for Unresectable Stage III Non-Small Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2024; 118:1455-1460. [PMID: 38159097 DOI: 10.1016/j.ijrobp.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Affiliation(s)
- Valentina Bartolomeo
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Department of Radiation Oncology (Maastro Clinic), Maastricht University Medical Center, GROW School for Oncology and Reproduction, Maastricht, The Netherlands
| | - Francesco Cortiula
- Department of Radiation Oncology (Maastro Clinic), Maastricht University Medical Center, GROW School for Oncology and Reproduction, Maastricht, The Netherlands; Department of Medical Oncology, Udine University Hospital, Udine, Italy
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro Clinic), Maastricht University Medical Center, GROW School for Oncology and Reproduction, Maastricht, The Netherlands
| | - Andrea R Filippi
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| |
Collapse
|
137
|
Zhan PC, Yang S, Liu X, Zhang YY, Wang R, Wang JX, Qiu QY, Gao Y, Lv DB, Li LM, Luo CL, Hu ZW, Li Z, Lyu PJ, Liang P, Gao JB. A radiomics signature derived from CT imaging to predict MSI status and immunotherapy outcomes in gastric cancer: a multi-cohort study. BMC Cancer 2024; 24:404. [PMID: 38561648 PMCID: PMC10985890 DOI: 10.1186/s12885-024-12174-0] [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: 09/16/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Accurate microsatellite instability (MSI) testing is essential for identifying gastric cancer (GC) patients eligible for immunotherapy. We aimed to develop and validate a CT-based radiomics signature to predict MSI and immunotherapy outcomes in GC. METHODS This retrospective multicohort study included a total of 457 GC patients from two independent medical centers in China and The Cancer Imaging Archive (TCIA) databases. The primary cohort (n = 201, center 1, 2017-2022), was used for signature development via Least Absolute Shrinkage and Selection Operator (LASSO) and logistic regression analysis. Two independent immunotherapy cohorts, one from center 1 (n = 184, 2018-2021) and another from center 2 (n = 43, 2020-2021), were utilized to assess the signature's association with immunotherapy response and survival. Diagnostic efficiency was evaluated using the area under the receiver operating characteristic curve (AUC), and survival outcomes were analyzed via the Kaplan-Meier method. The TCIA cohort (n = 29) was included to evaluate the immune infiltration landscape of the radiomics signature subgroups using both CT images and mRNA sequencing data. RESULTS Nine radiomics features were identified for signature development, exhibiting excellent discriminative performance in both the training (AUC: 0.851, 95%CI: 0.782, 0.919) and validation cohorts (AUC: 0.816, 95%CI: 0.706, 0.926). The radscore, calculated using the signature, demonstrated strong predictive abilities for objective response in immunotherapy cohorts (AUC: 0.734, 95%CI: 0.662, 0.806; AUC: 0.724, 95%CI: 0.572, 0.877). Additionally, the radscore showed a significant association with PFS and OS, with GC patients with a low radscore experiencing a significant survival benefit from immunotherapy. Immune infiltration analysis revealed significantly higher levels of CD8 + T cells, activated CD4 + B cells, and TNFRSF18 expression in the low radscore group, while the high radscore group exhibited higher levels of T cells regulatory and HHLA2 expression. CONCLUSION This study developed a robust radiomics signature with the potential to serve as a non-invasive biomarker for GC's MSI status and immunotherapy response, demonstrating notable links to post-immunotherapy PFS and OS. Additionally, distinct immune profiles were observed between low and high radscore groups, highlighting their potential clinical implications.
Collapse
Affiliation(s)
- Peng-Chao Zhan
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Shuo Yang
- Department of Radiology, The Second Hospital, Cheello College of Medicine, Shandong University, 250033, Jinan, PR China
| | - Xing Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Yu-Yuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, PR China
| | - Rui Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Jia-Xing Wang
- Department of Interventional Medicine, The Second Hospital, Cheello College of Medicine, Shandong University, 250033, Jinan, Shandong, PR China
| | - Qing-Ya Qiu
- Zhengzhou University Medical College, 450052, Zhengzhou, Henan, PR China
| | - Yu Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Dong-Bo Lv
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Li-Ming Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Cheng-Long Luo
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Zhi-Wei Hu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, PR China
| | - Pei-Jie Lyu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Pan Liang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China
| | - Jian-Bo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, 450052, Zhengzhou, Henan, PR China.
| |
Collapse
|
138
|
Wei S, Gou X, Zhang Y, Cui J, Liu X, Hong N, Sheng W, Cheng J, Wang Y. Prediction of transformation in the histopathological growth pattern of colorectal liver metastases after chemotherapy using CT-based radiomics. Clin Exp Metastasis 2024; 41:143-154. [PMID: 38416301 DOI: 10.1007/s10585-024-10275-5] [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: 10/10/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Chemotherapy alters the prognostic biomarker histopathological growth pattern (HGP) phenotype in colorectal liver metastases (CRLMs) patients. We aimed to develop a CT-based radiomics model to predict the transformation of the HGP phenotype after chemotherapy. This study included 181 patients with 298 CRLMs who underwent preoperative contrast-enhanced CT followed by partial hepatectomy between January 2007 and July 2022 at two institutions. HGPs were categorized as pure desmoplastic HGP (pdHGP) or non-pdHGP. The samples were allocated to training, internal validation, and external validation cohorts comprising 153, 65, and 29 CRLMs, respectively. Radiomics analysis was performed on pre-enhanced, arterial phase, portal venous phase (PVP), and fused images. The model was used to predict prechemotherapy HGPs in 112 CRLMs, and HGP transformation was analysed by comparing these findings with postchemotherapy HGPs determined pathologically. The prevalence of pdHGP was 19.8% (23/116) and 45.8% (70/153) in chemonaïve and postchemotherapy patients, respectively (P < 0.001). The PVP radiomics signature showed good performance in distinguishing pdHGP from non-pdHGPs (AUCs of 0.906, 0.877, and 0.805 in the training, internal validation, and external validation cohorts, respectively). The prevalence of prechemotherapy pdHGP predicted by the radiomics model was 33.0% (37/112), and the prevalence of postchemotherapy pdHGP according to the pathological analysis was 47.3% (53/112; P = 0.029). The transformation of HGP was bidirectional, with 15.2% (17/112) of CRLMs transforming from prechemotherapy pdHGP to postchemotherapy non-pdHGP and 30.4% (34/112) transforming from prechemotherapy non-pdHGP to postchemotherapy pdHGP (P = 0.005). CT-based radiomics method can be used to effectively predict the HGP transformation in chemotherapy-treated CRLM patients, thereby providing a basis for treatment decisions.
Collapse
Affiliation(s)
- Shengcai Wei
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Xinyi Gou
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Jingjing Cui
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd, Yongteng North Road, Haidian District, Beijing, 100094, China
| | - Xiaoming Liu
- Department of Research and Development, Beijing United Imaging Research Institute of Intelligent Imaging, Yongteng North Road, Haidian District, Beijing, 100089, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China.
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen South St, Beijing, 100044, China.
| |
Collapse
|
139
|
Mei T, Wang T, Zhou Q. Multi-omics and artificial intelligence predict clinical outcomes of immunotherapy in non-small cell lung cancer patients. Clin Exp Med 2024; 24:60. [PMID: 38554212 PMCID: PMC10981593 DOI: 10.1007/s10238-024-01324-0] [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: 12/21/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024]
Abstract
In recent years, various types of immunotherapy, particularly the use of immune checkpoint inhibitors targeting programmed cell death 1 or programmed death ligand 1 (PD-L1), have revolutionized the management and prognosis of non-small cell lung cancer. PD-L1 is frequently used as a biomarker for predicting the likely benefit of immunotherapy for patients. However, some patients receiving immunotherapy have high response rates despite having low levels of PD-L1. Therefore, the identification of this group of patients is extremely important to improve prognosis. The tumor microenvironment contains tumor, stromal, and infiltrating immune cells with its composition differing significantly within tumors, between tumors, and between individuals. The omics approach aims to provide a comprehensive assessment of each patient through high-throughput extracted features, promising a more comprehensive characterization of this complex ecosystem. However, features identified by high-throughput methods are complex and present analytical challenges to clinicians and data scientists. It is thus feasible that artificial intelligence could assist in the identification of features that are beyond human discernment as well as in the performance of repetitive tasks. In this paper, we review the prediction of immunotherapy efficacy by different biomarkers (genomic, transcriptomic, proteomic, microbiomic, and radiomic), together with the use of artificial intelligence and the challenges and future directions of these fields.
Collapse
Affiliation(s)
- Ting Mei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ting Wang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Qinghua Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
140
|
Li X, Wu M, Wu M, Liu J, Song L, Wang J, Zhou J, Li S, Yang H, Zhang J, Cui X, Liu Z, Zeng F. A radiomics and genomics-derived model for predicting metastasis and prognosis in colorectal cancer. Carcinogenesis 2024; 45:170-180. [PMID: 38195111 DOI: 10.1093/carcin/bgad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
Approximately 50% of colorectal cancer (CRC) patients would develop metastasis with poor prognosis, therefore, it is necessary to effectively predict metastasis in clinical treatment. In this study, we aimed to establish a machine-learning model for predicting metastasis in CRC patients by considering radiomics and transcriptomics simultaneously. Here, 1023 patients with CRC from three centers were collected and divided into five queues (Dazhou Central Hospital n = 517, Nanchong Central Hospital n = 120 and the Cancer Genome Atlas (TCGA) n = 386). A total of 854 radiomics features were extracted from tumor lesions on CT images, and 217 differentially expressed genes were obtained from non-metastasis and metastasis tumor tissues using RNA sequencing. Based on radiotranscriptomic (RT) analysis, a novel RT model was developed and verified through genetic algorithms (GA). Interleukin (IL)-26, a biomarker in RT model, was verified for its biological function in CRC metastasis. Furthermore, 15 radiomics variables were screened through stepwise regression, which was highly correlated with the IL26 expression level. Finally, a radiomics model (RA) was established by combining GA and stepwise regression analysis with radiomics features. The RA model exhibited favorable discriminatory ability and accuracy for metastasis prediction in two independent verification cohorts. We designed multicenter, multi-scale cohorts to construct and verify novel combined radiomics and genomics models for predicting metastasis in CRC. Overall, RT model and RA model might help clinicians in directing personalized diagnosis and therapeutic regimen selection for patients with CRC.
Collapse
Affiliation(s)
- Xue Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Min Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Li Song
- Department of Clinical laboratory, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Jiasi Wang
- Department of Clinical laboratory, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Jun Zhou
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Shilin Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Hang Yang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Jun Zhang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100080, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan 635000, China
| |
Collapse
|
141
|
Qin Y, Huo M, Liu X, Li SC. Biomarkers and computational models for predicting efficacy to tumor ICI immunotherapy. Front Immunol 2024; 15:1368749. [PMID: 38524135 PMCID: PMC10957591 DOI: 10.3389/fimmu.2024.1368749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Numerous studies have shown that immune checkpoint inhibitor (ICI) immunotherapy has great potential as a cancer treatment, leading to significant clinical improvements in numerous cases. However, it benefits a minority of patients, underscoring the importance of discovering reliable biomarkers that can be used to screen for potential beneficiaries and ultimately reduce the risk of overtreatment. Our comprehensive review focuses on the latest advancements in predictive biomarkers for ICI therapy, particularly emphasizing those that enhance the efficacy of programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors immunotherapies. We explore biomarkers derived from various sources, including tumor cells, the tumor immune microenvironment (TIME), body fluids, gut microbes, and metabolites. Among them, tumor cells-derived biomarkers include tumor mutational burden (TMB) biomarker, tumor neoantigen burden (TNB) biomarker, microsatellite instability (MSI) biomarker, PD-L1 expression biomarker, mutated gene biomarkers in pathways, and epigenetic biomarkers. TIME-derived biomarkers include immune landscape of TIME biomarkers, inhibitory checkpoints biomarkers, and immune repertoire biomarkers. We also discuss various techniques used to detect and assess these biomarkers, detailing their respective datasets, strengths, weaknesses, and evaluative metrics. Furthermore, we present a comprehensive review of computer models for predicting the response to ICI therapy. The computer models include knowledge-based mechanistic models and data-based machine learning (ML) models. Among the knowledge-based mechanistic models are pharmacokinetic/pharmacodynamic (PK/PD) models, partial differential equation (PDE) models, signal networks-based models, quantitative systems pharmacology (QSP) models, and agent-based models (ABMs). ML models include linear regression models, logistic regression models, support vector machine (SVM)/random forest/extra trees/k-nearest neighbors (KNN) models, artificial neural network (ANN) and deep learning models. Additionally, there are hybrid models of systems biology and ML. We summarized the details of these models, outlining the datasets they utilize, their evaluation methods/metrics, and their respective strengths and limitations. By summarizing the major advances in the research on predictive biomarkers and computer models for the therapeutic effect and clinical utility of tumor ICI, we aim to assist researchers in choosing appropriate biomarkers or computer models for research exploration and help clinicians conduct precision medicine by selecting the best biomarkers.
Collapse
Affiliation(s)
- Yurong Qin
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Miaozhe Huo
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Xingwu Liu
- School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
142
|
Ma J, Chen K, Li S, Zhu L, Yu Y, Li J, Ma J, Ouyang J, Wu Z, Tan Y, He Z, Liu H, Pan Z, Li H, Liu Q, Song E. MRI-based radiomic models to predict surgical margin status and infer tumor immune microenvironment in breast cancer patients with breast-conserving surgery: a multicenter validation study. Eur Radiol 2024; 34:1774-1789. [PMID: 37658888 DOI: 10.1007/s00330-023-10144-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2023] [Accepted: 07/08/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVES Accurate preoperative estimation of the risk of breast-conserving surgery (BCS) resection margin positivity would be beneficial to surgical planning. In this multicenter validation study, we developed an MRI-based radiomic model to predict the surgical margin status. METHODS We retrospectively collected preoperative breast MRI of patients undergoing BCS from three hospitals (SYMH, n = 296; SYSUCC, n = 131; TSPH, n = 143). Radiomic-based model for risk prediction of the margin positivity was trained on the SYMH patients (7:3 ratio split for the training and testing cohorts), and externally validated in the SYSUCC and TSPH cohorts. The model was able to stratify patients into different subgroups with varied risk of margin positivity. Moreover, we used the immune-radiomic models and epithelial-mesenchymal transition (EMT) signature to infer the distribution patterns of immune cells and tumor cell EMT status under different marginal status. RESULTS The AUCs of the radiomic-based model were 0.78 (0.66-0.90), 0.88 (0.79-0.96), and 0.76 (0.68-0.84) in the testing cohort and two external validation cohorts, respectively. The actual margin positivity rates ranged between 0-10% and 27.3-87.2% in low-risk and high-risk subgroups, respectively. Positive surgical margin was associated with higher levels of EMT and B cell infiltration in the tumor area, as well as the enrichment of B cells, immature dendritic cells, and neutrophil infiltration in the peritumoral area. CONCLUSIONS This MRI-based predictive model can be used as a reliable tool to predict the risk of margin positivity of BCS. Tumor immune-microenvironment alteration was associated with surgical margin status. CLINICAL RELEVANCE STATEMENT This study can assist the pre-operative planning of BCS. Further research on the tumor immune microenvironment of different resection margin states is expected to develop new margin evaluation indicators and decipher the internal mechanism. KEY POINTS • The MRI-based radiomic prediction model (CSS model) incorporating features extracted from multiple sequences and segments could estimate the margin positivity risk of breast-conserving surgery. • The radiomic score of the CSS model allows risk stratification of patients undergoing breast-conserving surgery, which could assist in surgical planning. • With the help of MRI-based radiomics to estimate the components of the immune microenvironment, for the first time, it is found that the margin status of breast-conserving surgery is associated with the infiltration of immune cells in the microenvironment and the EMT status of breast tumor cells.
Collapse
Affiliation(s)
- Jiafan Ma
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Kai Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Artificial Intelligence Lab, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Shunrong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Liling Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yunfang Yu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Jingwu Li
- Department of Breast Surgery, Tangshan People's Hospital, Tangshan, 063001, Hebei, China
| | - Jie Ma
- Department of Breast Surgery, Tangshan People's Hospital, Tangshan, 063001, Hebei, China
| | - Jie Ouyang
- Department of Breast Surgery, Tungwah Hospital, Sun Yat-sen University, Dongguan, 523413, China
| | - Zhuo Wu
- Artificial Intelligence Lab, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yujie Tan
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zifan He
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Haiqing Liu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhilong Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Haojiang Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, China.
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Yat-sen Breast Tumor Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
143
|
Wang P, Wang X, Zhang M, Li G, Zhao N, Qiao Q. Combining the radiomics signature and HPV status for the risk stratification of patients with OPC. Oral Dis 2024; 30:272-280. [PMID: 36135344 DOI: 10.1111/odi.14386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/08/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The objective was to perform risk stratification of oropharyngeal cancer (OPC) for treatment de-escalation based on the radiomics analysis and human papillomavirus (HPV) status. METHODS A total of 265 patients with OPC who underwent baseline contrast-enhanced computed tomography were analyzed, and the patients were grouped into the training (n = 133) and test (n = 132) cohorts at a ratio of 1:1. Intratumoral and peritumoral radiomics features were extracted, and the radiomics signature (Rscore) was calculated using least absolute shrinkage and selection operator regression (LASSO) and Cox regression analyses. RESULTS Twelve features were selected to establish the radiomics signature (Rscore) of intratumoral regions +10-mm peritumoral regions, which yielded maximum AUCs of 0.835, 0.798, and 0.784 in the training, test, and validation cohorts, respectively. Patients with OPC were divided into the high-risk group, intermediate-risk group, and low-risk group based on the Rscore and HPV status and had different prognoses. Patients in the low-risk group benefit from radiotherapy alone, and patients in the intermediate-risk group only benefitted from chemoradiotherapy. CONCLUSION The radiomics signature appears to improve the predictive performance of clinical characteristics for oropharyngeal cancer. The combined stratification of the radiomics signature and HPV status might be preferred to select patients for de-escalated treatment.
Collapse
Affiliation(s)
- Ping Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xuan Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Miao Zhang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Guang Li
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Ning Zhao
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Qiao Qiao
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
144
|
Li H, Chai L, Pu H, Yin LL, Li M, Zhang X, Liu YS, Pang MH, Lu T. T2WI-based MRI radiomics for the prediction of preoperative extranodal extension and prognosis in resectable rectal cancer. Insights Imaging 2024; 15:57. [PMID: 38411722 PMCID: PMC10899552 DOI: 10.1186/s13244-024-01625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE To investigate whether T2-weighted imaging (T2WI)-based intratumoral and peritumoral radiomics can predict extranodal extension (ENE) and prognosis in patients with resectable rectal cancer. METHODS One hundred sixty-seven patients with resectable rectal cancer including T3T4N + cases were prospectively included. Radiomics features were extracted from intratumoral, peritumoral 3 mm, and peritumoral-mesorectal fat on T2WI images. Least absolute shrinkage and selection operator regression were used for feature selection. A radiomics signature score (Radscore) was built with logistic regression analysis. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of each Radscore. A clinical-radiomics nomogram was constructed by the most predictive radiomics signature and clinical risk factors. A prognostic model was constructed by Cox regression analysis to identify 3-year recurrence-free survival (RFS). RESULTS Age, cT stage, and lymph node-irregular border and/or adjacent fat invasion were identified as independent clinical risk factors to construct a clinical model. The nomogram incorporating intratumoral and peritumoral 3 mm Radscore and independent clinical risk factors achieved a better AUC than the clinical model in the training (0.799 vs. 0.736) and validation cohorts (0.723 vs. 0.667). Nomogram-based ENE (hazard ratio [HR] = 2.625, 95% CI = 1.233-5.586, p = 0.012) and extramural vascular invasion (EMVI) (HR = 2.523, 95% CI = 1.247-5.106, p = 0.010) were independent risk factors for predicting 3-year RFS. The prognostic model constructed by these two indicators showed good performance for predicting 3-year RFS in the training (AUC = 0.761) and validation cohorts (AUC = 0.710). CONCLUSION The nomogram incorporating intratumoral and peritumoral 3 mm Radscore and clinical risk factors could predict preoperative ENE. Combining nomogram-based ENE and MRI-reported EMVI may be useful in predicting 3-year RFS. CRITICAL RELEVANCE STATEMENT A clinical-radiomics nomogram could help preoperative predict ENE, and a prognostic model constructed by the nomogram-based ENE and MRI-reported EMVI could predict 3-year RFS in patients with resectable rectal cancer. KEY POINTS • Intratumoral and peritumoral 3 mm Radscore showed the most capability for predicting ENE. • Clinical-radiomics nomogram achieved the best predictive performance for predicting ENE. • Combining clinical-radiomics based-ENE and EMVI showed good performance for 3-year RFS.
Collapse
Affiliation(s)
- Hang Li
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Li Chai
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Pu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Long-Lin Yin
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
- Institute of Radiation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mou Li
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Xin Zhang
- Pharmaceutical Diagnostic Team, GE Healthcare, Beijing, 100176, China
| | - Yi-Sha Liu
- Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Ming-Hui Pang
- Department of Geriatric Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Tao Lu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China.
| |
Collapse
|
145
|
Suryani L, Lee HPY, Teo WK, Chin ZK, Loh KS, Tay JK. Precision Medicine for Nasopharyngeal Cancer-A Review of Current Prognostic Strategies. Cancers (Basel) 2024; 16:918. [PMID: 38473280 DOI: 10.3390/cancers16050918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV) driven malignancy arising from the nasopharyngeal epithelium. Current treatment strategies depend on the clinical stage of the disease, including the extent of the primary tumour, the extent of nodal disease, and the presence of distant metastasis. With the close association of EBV infection with NPC development, EBV biomarkers have shown promise in predicting treatment outcomes. Among the omic technologies, RNA and miRNA signatures have been widely studied, showing promising results in the research setting to predict treatment response. The transformation of radiology images into measurable features has facilitated the use of radiomics to generate predictive models for better prognostication and treatment selection. Nonetheless, much of this work remains in the research realm, and challenges remain in clinical implementation.
Collapse
Affiliation(s)
- Luvita Suryani
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hazel P Y Lee
- Department of Otolaryngology-Head & Neck Surgery, National University Hospital, Singapore 119228, Singapore
| | - Wei Keat Teo
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Zhi Kang Chin
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Kwok Seng Loh
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joshua K Tay
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
146
|
Ligero M, Gielen B, Navarro V, Cresta Morgado P, Prior O, Dienstmann R, Nuciforo P, Trebeschi S, Beets-Tan R, Sala E, Garralda E, Perez-Lopez R. A whirl of radiomics-based biomarkers in cancer immunotherapy, why is large scale validation still lacking? NPJ Precis Oncol 2024; 8:42. [PMID: 38383736 PMCID: PMC10881558 DOI: 10.1038/s41698-024-00534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
The search for understanding immunotherapy response has sparked interest in diverse areas of oncology, with artificial intelligence (AI) and radiomics emerging as promising tools, capable of gathering large amounts of information to identify suitable patients for treatment. The application of AI in radiology has grown, driven by the hypothesis that radiology images capture tumor phenotypes and thus could provide valuable insights into immunotherapy response likelihood. However, despite the rapid growth of studies, no algorithms in the field have reached clinical implementation, mainly due to the lack of standardized methods, hampering study comparisons and reproducibility across different datasets. In this review, we performed a comprehensive assessment of published data to identify sources of variability in radiomics study design that hinder the comparison of the different model performance and, therefore, clinical implementation. Subsequently, we conducted a use-case meta-analysis using homogenous studies to assess the overall performance of radiomics in estimating programmed death-ligand 1 (PD-L1) expression. Our findings indicate that, despite numerous attempts to predict immunotherapy response, only a limited number of studies share comparable methodologies and report sufficient data about cohorts and methods to be suitable for meta-analysis. Nevertheless, although only a few studies meet these criteria, their promising results underscore the importance of ongoing standardization and benchmarking efforts. This review highlights the importance of uniformity in study design and reporting. Such standardization is crucial to enable meaningful comparisons and demonstrate the validity of biomarkers across diverse populations, facilitating their implementation into the immunotherapy patient selection process.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bente Gielen
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Victor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
- Prostate Cancer Translational Research Group, Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Olivia Prior
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Regina Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Evis Sala
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
147
|
Fiste O, Gkiozos I, Charpidou A, Syrigos NK. Artificial Intelligence-Based Treatment Decisions: A New Era for NSCLC. Cancers (Basel) 2024; 16:831. [PMID: 38398222 PMCID: PMC10887017 DOI: 10.3390/cancers16040831] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality among women and men, in developed countries, despite the public health interventions including tobacco-free campaigns, screening and early detection methods, recent therapeutic advances, and ongoing intense research on novel antineoplastic modalities. Targeting oncogenic driver mutations and immune checkpoint inhibition has indeed revolutionized NSCLC treatment, yet there still remains the unmet need for robust and standardized predictive biomarkers to accurately inform clinical decisions. Artificial intelligence (AI) represents the computer-based science concerned with large datasets for complex problem-solving. Its concept has brought a paradigm shift in oncology considering its immense potential for improved diagnosis, treatment guidance, and prognosis. In this review, we present the current state of AI-driven applications on NSCLC management, with a particular focus on radiomics and pathomics, and critically discuss both the existing limitations and future directions in this field. The thoracic oncology community should not be discouraged by the likely long road of AI implementation into daily clinical practice, as its transformative impact on personalized treatment approaches is undeniable.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (A.C.); (N.K.S.)
| | | | | | | |
Collapse
|
148
|
Li Y, Wang P, Xu J, Shi X, Yin T, Teng F. Noninvasive radiomic biomarkers for predicting pseudoprogression and hyperprogression in patients with non-small cell lung cancer treated with immune checkpoint inhibition. Oncoimmunology 2024; 13:2312628. [PMID: 38343749 PMCID: PMC10857548 DOI: 10.1080/2162402x.2024.2312628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
This study aimed to develop a computed tomography (CT)-based radiomics model capable of precisely predicting hyperprogression and pseudoprogression (PP) in patients with non-small cell lung cancer (NSCLC) treated with immunotherapy. We retrospectively analyzed 105 patients with NSCLC, from three institutions, treated with immune checkpoint inhibitors (ICIs) and categorized them into training and independent testing set. Subsequently, we processed CT scans with a series of image-preprocessing techniques, and 6008 radiomic features capturing intra- and peritumoral texture patterns were extracted. We used the least absolute shrinkage and selection operator logistic regression model to select radiomic features and construct machine learning models. To further differentiate between progressive disease (PD) and hyperprogressive disease (HPD), we developed a new radiomics model. The logistic regression (LR) model showed optimal performance in distinguishing PP from HPD, with areas under the receiver operating characteristic curve (AUC) of 0.95 (95% confidence interval [CI]: 0.91-0.99) and 0.88 (95% CI: 0.66-1) in the training and testing sets, respectively. Additionally, the support vector machine model showed optimal performance in distinguishing PD from HPD, with AUC of 0.97 (95% CI: 0.93-1) and 0.87 (95% CI: 0.72-1) in the training and testing sets, respectively. Kaplan‒Meier survival curves showed clear stratification between PP predicted by the radiomics model and true progression (HPD and PD) (hazard ratio = 0.337, 95% CI: 0.200-0.568, p < 0.01) in overall survival. Our study demonstrates that radiomic features extracted from baseline CT scans are effective in predicting PP and HPD in patients with NSCLC treated with ICIs.
Collapse
Affiliation(s)
- Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Xiaonan Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
149
|
Qian H, Ren X, Xu M, Fang Z, Zhang R, Bu Y, Zhou C. Magnetic resonance imaging-based radiomics was used to evaluate the level of prognosis-related immune cell infiltration in breast cancer tumor microenvironment. BMC Med Imaging 2024; 24:31. [PMID: 38308230 PMCID: PMC10835863 DOI: 10.1186/s12880-024-01212-9] [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: 02/23/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
PURPOSE The tumor immune microenvironment is a valuable source of information for predicting prognosis in breast cancer (BRCA) patients. To identify immune cells associated with BRCA patient prognosis from the Cancer Genetic Atlas (TCGA), we established an MRI-based radiomics model for evaluating the degree of immune cell infiltration in breast cancer patients. METHODS CIBERSORT was utilized to evaluate the degree of infiltration of 22 immune cell types in breast cancer patients from the TCGA database, and both univariate and multivariate Cox regressions were employed to determine the prognostic significance of immune cell infiltration levels in BRCA patients. We identified independent prognostic factors for BRCA patients. Additionally, we obtained imaging features from the Cancer Imaging Archive (TCIA) database for 73 patients who underwent preoperative MRI procedures, and used the Least Absolute Shrinkage and Selection Operator (LASSO) to select the best imaging features for constructing an MRI-based radiomics model for evaluating immune cell infiltration levels in breast cancer patients. RESULTS According to the results of Cox regression analysis, M2 macrophages were identified as an independent prognostic factor for BRCA patients (HR = 32.288, 95% CI: 3.100-357.478). A total of nine significant features were selected to calculate the radiomics-based score. We established an intratumoral model with AUCs (95% CI) of 0.662 (0.495-0.802) and 0.678 (0.438-0.901) in the training and testing cohorts, respectively. Additionally, a peritumoral model was created with AUCs (95% CI) of 0.826 (0.710-0.924) and 0.752 (0.525-0.957), and a combined model was established with AUCs (95% CI) of 0.843 (0.723-0.938) and 0.744 (0.491-0.965). The peritumoral model demonstrated the highest diagnostic efficacy, with an accuracy, sensitivity, and specificity of 0.773, 0.727, and 0.818, respectively, in its testing cohort. CONCLUSION The MRI-based radiomics model has the potential to evaluate the degree of immune cell infiltration in breast cancer patients, offering a non-invasive imaging biomarker for assessing the tumor microenvironment in this disease.
Collapse
Affiliation(s)
- Hua Qian
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaojing Ren
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Fang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruixin Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyang Bu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China.
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
150
|
Chen M, Aboagye E. In Response. J Thorac Oncol 2024; 19:345. [PMID: 38325981 DOI: 10.1016/j.jtho.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 02/09/2024]
Affiliation(s)
- Mitchell Chen
- Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Eric Aboagye
- Surgery and Cancer, Imperial College London, London, United Kingdom.
| |
Collapse
|