1
|
Sakkal M, Hajal AA. Machine learning predictions of tumor progression: How reliable are we? Comput Biol Med 2025; 191:110156. [PMID: 40245687 DOI: 10.1016/j.compbiomed.2025.110156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/06/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Cancer continues to pose significant challenges in healthcare due to the complex nature of tumor progression. In this digital era, artificial intelligence has emerged as a powerful tool that can potentially transform multiple aspects of cancer care. METHODS In the current study, we conducted a comprehensive literature search across databases such as PubMed, Scopus, and IEEE Xplore. Studies published between 2014 and 2024 were considered. The selection process involved a systematic screening based on predefined inclusion and exclusion criteria. Studies were included if they focused on applying machine learning techniques for tumor progression modeling, diagnosis, or prognosis, were published in peer-reviewed journals or conference proceedings, were available in English, and presented experimental results, simulations, or real-world applications. In total, 87 papers were included in this review, ensuring a diverse and representative analysis of the field. A workflow is included to illustrate the procedure followed to achieve this aim. RESULTS This review delves into the cutting-edge applications of machine learning (ML), including supervised learning methods like Support Vector Machines and Random Forests, as well as advanced deep learning (DL). It focuses on the integration of ML into oncological research, particularly its application in tumor progression through the tumor microenvironment, genetic data, histopathological data, and radiological data. This work provides a critical analysis of the challenges associated with the reliability and accuracy of ML models, which limit their clinical integration. CONCLUSION This review offers expert insights and strategies to address these challenges in order to improve the robustness and applicability of ML in real-world oncology settings. By emphasizing the potential for personalized cancer treatment and bridging gaps between technology and clinical needs, this review serves as a comprehensive resource for advancing the integration of ML models into clinical oncology.
Collapse
Affiliation(s)
- Molham Sakkal
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Abdallah Abou Hajal
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
2
|
Chen H, Wen Y, Wu W, Zhang Y, Pan X, Guan Y, Qin D. Prediction of Malignancy and Pathological Types of Solid Lung Nodules on CT Scans Using a Volumetric SWIN Transformer. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1509-1517. [PMID: 39402355 DOI: 10.1007/s10278-024-01090-1] [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: 10/24/2023] [Revised: 01/19/2024] [Accepted: 03/01/2024] [Indexed: 05/22/2025]
Abstract
Lung adenocarcinoma and squamous cell carcinoma are the two most common pathological lung cancer subtypes. Accurate diagnosis and pathological subtyping are crucial for lung cancer treatment. Solitary solid lung nodules with lobulation and spiculation signs are often indicative of lung cancer; however, in some cases, postoperative pathology finds benign solid lung nodules. It is critical to accurately identify solid lung nodules with lobulation and spiculation signs before surgery; however, traditional diagnostic imaging is prone to misdiagnosis, and studies on artificial intelligence-assisted diagnosis are few. Therefore, we introduce a volumetric SWIN Transformer-based method. It is a multi-scale, multi-task, and highly interpretable model for distinguishing between benign solid lung nodules with lobulation and spiculation signs, lung adenocarcinomas, and lung squamous cell carcinoma. The technique's effectiveness was improved by using 3-dimensional (3D) computed tomography (CT) images instead of conventional 2-dimensional (2D) images to combine as much information as possible. The model was trained using 352 of the 441 CT image sequences and validated using the rest. The experimental results showed that our model could accurately differentiate between benign lung nodules with lobulation and spiculation signs, lung adenocarcinoma, and squamous cell carcinoma. On the test set, our model achieves an accuracy of 0.9888, precision of 0.9892, recall of 0.9888, and an F1-score of 0.9888, along with a class activation mapping (CAM) visualization of the 3D model. Consequently, our method could be used as a preoperative tool to assist in diagnosing solitary solid lung nodules with lobulation and spiculation signs accurately and provide a theoretical basis for developing appropriate clinical diagnosis and treatment plans for the patients.
Collapse
Affiliation(s)
- Huicong Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510799, China
| | - Yanhua Wen
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Road, Guangzhou, Guangdong, 510700, China
| | - Wensheng Wu
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Road, Guangzhou, Guangdong, 510700, China
| | - Yingying Zhang
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Road, Guangzhou, Guangdong, 510700, China
| | - Xiaohuan Pan
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yubao Guan
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Road, Guangzhou, Guangdong, 510700, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510799, China.
| |
Collapse
|
3
|
Peng M, Yang X, Wang Y, Zhou L, Ge F, Liu S, Liu W, Cheng L, Wang K. Clinical combined PET/CT radiomics model prediction of benefit from platinum-based chemotherapy and chemoradiotherapy in patients with small cell lung cancer. Nucl Med Commun 2025; 46:558-569. [PMID: 40084524 DOI: 10.1097/mnm.0000000000001971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
OBJECTIVE To develop and validate a clinical combined radiomics model for predicting the treatment response and long-term survival prognosis of small cell lung cancer (SCLC) patients receiving platinum-based chemotherapy, as well as survival outcomes following chemoradiotherapy. METHODS A total of 98 SCLC patients treated with platinum-based first-line chemotherapy were included in this study. Five prediction models for assessing the short-term efficacy of platinum-based first-line chemotherapy were developed using a logistic regression algorithm. The performance of the models was assessed by calculating the area under the curve of the receiver operating characteristic curves. For predicting progression-free survival (PFS) and overall survival in the platinum-based chemotherapy group and the chemoradiotherapy group, the optimal cutoff value was determined using X-tile software. Kaplan-Meier survival curves were plotted, and the log-rank test was used to compare survival outcomes. RESULTS Among the five models for predicting short-term efficacy, the clinical combined positron emission tomography/computed tomography (PET/CT) radiomics model performed the best, achieving areas under the curve of 0.832 and 0.833 for the training and test sets, respectively. The Kaplan-Meier survival analysis indicated that both the high-scoring Combine group and high-scoring PET/CT group were significantly associated with worse PFS and worse overall survival in the platinum-only chemotherapy group. Additionally, the high-scoring CT group was significantly associated with worse PFS in the chemoradiotherapy group. CONCLUSION The clinical combined PET/CT radiomics model can noninvasively and accurately predict the response to platinum-based treatments in SCLC as well as long-term survival prognosis, which can contribute to personalized treatment strategies and guide precision therapy for SCLC patients.
Collapse
Affiliation(s)
- Mengye Peng
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Xinyue Yang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Yanmei Wang
- Scientific Research Center Department, Beijing General Electric Company, Beijing
| | - Liangqin Zhou
- Imaging Center Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fan Ge
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Shijia Liu
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Wei Liu
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Liang Cheng
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| | - Kezheng Wang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, Harbin
| |
Collapse
|
4
|
Wang C, Wang C, Zhang J, Ding M, Ge Y, He X. Development and validation of a radiogenomics prognostic model integrating PET/CT radiomics and glucose metabolism-related gene signatures for non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07354-4. [PMID: 40423774 DOI: 10.1007/s00259-025-07354-4] [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: 03/27/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a highly heterogeneous malignancy characterized by altered glucose metabolism. Integration of PET/CT radiomics with glucose metabolism-related genomic signatures could provide a more comprehensive approach for prognosis and treatment guidance. METHODS Radiomics features were extracted from PET/CT images of 156 NSCLC patients from The Cancer Imaging Archive (TCIA) database, and glucose metabolism-related gene signatures were obtained from TCGA and GEO databases. We developed a multimodal radiogenomics prognostic model (RGC-score) using least absolute shrinkage and selection operator (LASSO) regression, combining PET/CT radiomics, glucose metabolism-related genes (GMR-genes). Functional enrichment analysis, immune infiltration assessment, and drug sensitivity analysis were performed to investigate the biological significance of glucose metabolism-related genes (GMR-genes). RESULTS The RGC-score model effectively stratified NSCLC patients into distinct high- and low-risk groups with significant differences in survival outcomes (P < 0.001), demonstrating excellent predictive performance (1-year AUC = 0.907, 5-year AUC = 0.968).GMR-genes are mainly involved in the process of metabolic remodeling of tumors, which is closely related to the immune microenvironment (especially CD8+ T cell infiltration) and immune checkpoint molecule expression. Additionally, significant differences in drug sensitivity were identified between glucose metabolism subtypes. CONCLUSION The RGC-score robustly predicts NSCLC prognosis and informs metabolic-immune interactions for personalized therapy. Limitations include the retrospective design and modest validation cohort size, necessitating prospective multicenter trials.
Collapse
Affiliation(s)
- Chunsheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Congjie Wang
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Jianguo Zhang
- Department of Pulmonary Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Mingjun Ding
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211116, Jiangsu, China.
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211116, Jiangsu, China.
| |
Collapse
|
5
|
Liu C, He Y, Luo J. The influence of image selection and segmentation on the extraction of lung cancer imaging radiomics features using 3D-Slicer software. BMC Cancer 2025; 25:728. [PMID: 40247266 PMCID: PMC12007235 DOI: 10.1186/s12885-025-14094-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Extracting image features can predict the prognosis and treatment effect of non-small cell lung cancer, which has been increasingly confirmed. However, the specific operation using 3D-Slicer still lacks standardization. For example, image segmentation is manually performed based on the lung window or automatically performed through the mediastinal window. The images used for feature extraction are either enhanced or plain scanned. It is questionable whether these influencing factors will affect the extraction results and which results will be affected. This article intends to preliminarily explore the above issues. METHODS This article downloaded images of 22 patients with lung cancer from The Cancer Imaging Archive (TCIA), including 11 cases of adenocarcinoma and 11 cases of squamous cell carcinoma. Perform tumor image segmentation on the lung window and mediastinal window of the plain scan image, and the lung window and mediastinal window of the enhanced image. Manual drawing is used on the lung window, and automatic drawing is used on the mediastinal window and make manual modifications. Extracting radiomics features using Python radiomics. Firstly, analyze the image features of the original sequence and perform the Shapiro test. If it follows a normal distribution, perform an analysis of variance. If it does not follow a normal distribution, perform the Friedman test. Compare the significantly different image features pairwise. Then, a preliminary analysis was conducted on the differences between squamous cell carcinoma and adenocarcinoma in each group. RESULTS A total of 88 sets of imaging features were extracted, with 107 features in each group. Among them, 33 features showed significant differences. Continuing with pairwise repeated testing, it was found that there were 2 significant differences between enhanced and plain lung windows. There were 12 significant differences between enhanced lung windows and plain mediastinal windows. There is one significant difference between plain scanning and enhancement mediastinal window. There are 14 significant differences between the plain lung window and the enhanced mediastinal window groups. There are 14 significant differences between the lung window and the mediastinal window in the plain scan. There are 13 significant differences between the enhanced lung window and the mediastinal window. According to pathological grouping testing, it was found that there 54 significant differences between squamous cell carcinoma and adenocarcinoma. CONCLUSION The enhancement of lung CT has a relatively small impact on extracting image features, while selecting lung or mediastinal windows for image segmentation has a significant impact on extracting image features. Therefore, choosing lung or mediastinal windows for feature extraction should be carefully considered, as the size of the image segmentation range has a significant impact on image features. The impact of lung squamous cell carcinoma and adenocarcinoma on imaging features is also significant, indicating a high possibility of distinguishing between squamous cell carcinoma and adenocarcinoma based on radiomics (Liu C, He Y, Luo J, The Influence of Image Selection and Segmentation on the Extraction of Lung Cancer Imaging Radiomics Features Using 3D-Slicer Software, 2024).
Collapse
Affiliation(s)
- Chunmei Liu
- Department of Radiation Oncology, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang, Hebei Province, China
| | - Yuzheng He
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang, Hebei Province, China
| | - Jianmin Luo
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
6
|
Liang Y, Xie M, Zang X, Zhang X, Xue X. Evaluation of ImmunoPET in the efficacy and prognosis of immunotherapy for lung cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189289. [PMID: 39999945 DOI: 10.1016/j.bbcan.2025.189289] [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: 11/28/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Advances in immune oncology have established immunotherapy as the first-line standard treatment for lung cancer; however, its efficacy remains limited to a subset of patients. Developing predictive biomarkers within the tumor microenvironment (TME) to assess the efficacy and prognosis of immunotherapy can enhance drug development and treatment strategies. Immuno-positron emission tomography (ImmunoPET) non-invasively visualizes the biological distribution of key targets in the TME using highly specific, radiolabeled tracers. PET imaging of the TME can serve as a reliable biomarker for predicting and monitoring responses to immune therapy, complementing existing immunohistochemical techniques. This review will focus on the development of ImmunoPET biomarkers, as well as the application of corresponding tracers and radionuclides in lung cancer. We will focus on available clinical tracers and those under development, outlining each TME target and its clinical validation for tumor immunotherapy efficacy and prognosis, while discussing the latest advances that may enhance ImmunoPET in future.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xuefeng Zang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong 261000, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
7
|
Xu H, Wang H, Jiang D, Wu Y, Xie S, Su Y, Guan Y, Xie F, Zhu W, Qin L. Comparison of 11C-Acetate and 18F-FDG PET/CT for Immune Infiltration and Prognosis in Hepatocellular Carcinoma. Cancer Sci 2025; 116:990-1003. [PMID: 39797622 PMCID: PMC11967256 DOI: 10.1111/cas.16449] [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: 11/03/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, making it a challenge to noninvasively monitor immune infiltration. Metabolic reprogramming in cancers, including hepatocellular carcinoma (HCC), is closely linked to immune status. In this study, we aimed to evaluate the ability of carbon-11 acetate (11C-acetate) and fluorine-18 fluorodeoxyglucose (18F-FDG) PET/CT findings in predicting overall survival (OS) and immune infiltration in HCC patients. Totally 32 patients who underwent preoperative 18F-FDG and 11C-acetate PET/CT, followed by liver resection for HCC, were prospectively enrolled at authors' institute between January 2019 and October 2021. Tracer uptake was qualified. Densities of CD3+, CD8+, and granzyme B+ CD8+ immune cells were assessed and the Immunoscore was defined by combining the densities of CD3+ and CD8+ in tumor interior (TI) and invasion margin (IM). Patients with avid HCCs in 11C-acetate PET/CT demonstrated a longer OS. Those with only 11C-acetate-avid HCCs exhibited a longer OS compared to those with only 18F-FDG uptake. In contrast to 18F-FDG uptake, 11C-acetate uptake was positively associated with CD3+, CD8+, and granzyme B+ CD8+ cell infiltration. Patients with a higher Immunoscore exhibited a longer OS and an increased uptake of 11C-acetate rather than 18F-FDG. The sensitivity of 11C-acetate PET/CT in the detection of patients with immune infiltration was superior to that of 18F-FDG PET/CT (88% [21 of 24] vs. 58% [14 of 24]). These data show that preoperative 11C-acetate PET/CT may be a promising approach for the evaluation of immune status and postoperative outcome of HCCs.
Collapse
Affiliation(s)
- Hao Xu
- Shanghai Institute of Infectious Diseases and Biosecurity, Huashan HospitalFudan UniversityShanghaiChina
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
| | - Hao Wang
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
- Cancer Metastasis InstituteFudan UniversityShanghaiChina
| | - Dong‐Lang Jiang
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Yan‐Fei Wu
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Sun‐Zhe Xie
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
- Cancer Metastasis InstituteFudan UniversityShanghaiChina
| | - Ying‐Han Su
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
- Cancer Metastasis InstituteFudan UniversityShanghaiChina
| | - Yi‐Hui Guan
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Wen‐Wei Zhu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
- Cancer Metastasis InstituteFudan UniversityShanghaiChina
| | - Lun‐Xiu Qin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan HospitalFudan UniversityShanghaiChina
- Cancer Metastasis InstituteFudan UniversityShanghaiChina
| |
Collapse
|
8
|
Lei Y, Fan W, Liu B, Liao Y, Liu C, Xue S, Zhou D, Wang H, Zhang Q. Integrated radiomics and immune infiltration analysis to decipher immunotherapy efficacy in lung adenocarcinoma. Quant Imaging Med Surg 2025; 15:3123-3147. [PMID: 40235745 PMCID: PMC11994542 DOI: 10.21037/qims-24-130] [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: 01/22/2024] [Accepted: 02/26/2025] [Indexed: 04/17/2025]
Abstract
Background Research in recent years has witnessed unprecedented improvements in immunotherapy, especially immune checkpoint blockade (ICB) for the treatment of lung adenocarcinoma (LUAD) patients. Nevertheless, due to the heterogeneity of immunotherapy response, reliable biomarkers are urgently needed to guide precision cancer therapy. In this study, we aimed to identify immune subtypes in LUAD and develop a radiogenomic model to improve immunotherapy predictive accuracy. Methods In this study, clinical data of LUAD patients were downloaded from The Cancer Genome Atlas (TCGA) databases, and immune subtypes were identified using the ConsensusClusterPlus package in R. Biological, genomic, and epigenomic distinctions were compared. The TCGA cohort and clinical cohort from the Third Xiangya Hospital were utilized to demonstrate no significant differences of survival probability between sexes. Feature extraction and definition were conducted from 103 computed tomography (CT) images from The Cancer Imaging Archive (TCIA) dataset via the "PyRadiomics" embedded in Python. A series of machine learning techniques were applied to build a radiogenomic model. Results Two LUAD subtypes with different molecular and immune characteristics were identified. Significant differences in biological, genomic, and epigenomic distinctions among the two subtypes were observed (P<0.05). The immune subtype A participated in pathways related to immune activation and displayed a higher tumor microenvironment (TME) score (P<0.001) with a better prognosis of LUAD [overall survival (OS), P=0.037; disease-specific survival (DSS), P=0.034]. Besides, the model appears to show better fit for females (P=0.015) than for males (P=0.641). Our constructed radiogenomic model incorporating 12 radiomics features displayed satisfactory potential to facilitate the predictive accuracy of immunotherapy in LUAD [test area under the curve (AUC) =0.89; train AUC =0.95]. Conclusions Our study presented a promising avenue to harness the rich radiomics data to identify the specific immune subtype and integrate it into the existing clinical decision-making system to facilitate the predictive accuracy of immunotherapy in LUAD.
Collapse
Affiliation(s)
- Yiyi Lei
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenjin Fan
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Beizhan Liu
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxuan Liao
- Xiangya School of Medicine, Central South University, Changsha, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical college, Beijing, China
| | - Chenxi Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shengjie Xue
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Dawei Zhou
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongyi Wang
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qiang Zhang
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Fan Y, Liu Y, Ouyang X, Su J, Zhou X, Jia Q, Chen W, Chen W, Liu X. Prediction of EGFR mutation status and its subtypes in non-small cell lung cancer based on 18 F-FDG PET/CT radiological features. Nucl Med Commun 2025; 46:326-336. [PMID: 39829249 DOI: 10.1097/mnm.0000000000001948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
PURPOSE Prediction of epidermal growth factor receptor (EGFR) mutation status and subtypes in patients with non-small cell lung cancer (NSCLC) based on 18 F-fluorodeoxyglucose ( 18 F-FDG) PET/computed tomography (CT) radiomics features. PATIENTS AND METHODS Retrospective analysis of 201 NSCLC patients with 18 F-FDG PET/CT and EGFR genetic testing was carried out. Radiomics features and clinical factors were used to construct a combined model for identifying EGFR mutation status. Mutation/wild-type models were trained in a training cohort ( n = 129) and validated in an internal validation cohort ( n = 41) vs an external validation cohort ( n = 50). A second model predicting the 19/21 mutation locus was also built and evaluated in a subset of EGFR mutations (training cohort, n = 55; validation cohort, n = 14). The predictive performance and net clinical benefit of the models were assessed by analysis of the area under curve (AUC) of the subjects, nomogram, calibration curve and decision curve. RESULTS The AUC of the combined model distinguishing EGFR mutation status was 0.864 in the training cohort and 0.806 and 0.791 in the internal vs external test sets respectively, and the AUC of the 19/21 mutation site model was 0.971 and 0.867 in the training cohort and internal validation cohort respectively. The calibration curves of the individual models showed better model predictions (Brier score <0.25). Decision curve analysis showed that the models had clinical application. CONCLUSION The combined model based on 18 F-FDG PET/CT radiomics features combined and clinical features can predict EGFR mutation status and subtypes in NSCLC patients, and guiding targeted therapy, and facilitate precision medicine development.
Collapse
Affiliation(s)
- Yishuo Fan
- Department of Graduate School, Graduate School of Hebei North University, Zhangjiakou, Hebei,
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Yuang Liu
- Department of Graduate School, Graduate School of Hebei North University, Zhangjiakou, Hebei,
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Xiaohui Ouyang
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Jiagui Su
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Xiaohong Zhou
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Qichen Jia
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| | - Wenjing Chen
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd. and
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaofei Liu
- Department of Nuclear Medicine, The Eighth Medical Center of PLA General Hospital,
| |
Collapse
|
10
|
Lin X, Liu Z, Zhou K, Li Y, Huang G, Zhang H, Shu T, Huang Z, Wang Y, Zeng W, Liao Y, Bin J, Shi M, Liao W, Zhou W, Huang N. Intratumoral and peritumoral PET/CT-based radiomics for non-invasively and dynamically predicting immunotherapy response in NSCLC. Br J Cancer 2025; 132:558-568. [PMID: 39930148 PMCID: PMC11920075 DOI: 10.1038/s41416-025-02948-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND We aimed to develop a machine learning model based on intratumoral and peritumoral 18F-FDG PET/CT radiomics to non-invasively and dynamically predict the response to immunotherapy in non-small cell lung cancer (NSCLC). METHODS This retrospective study included 296 NSCLC patients, including a training cohort (N = 183), a testing cohort (N = 78), and a TCIA radiogenomic cohort (N = 35). The extreme gradient boosting algorithm was employed to develop the radiomic models. RESULTS The COMB-Radscore, which was developed by combining radiomic features from PET, CT, and PET/CT images, had the most satisfactory predictive performance with AUC (ROC) 0.894 and 0.819 in the training and testing cohorts, respectively. Survival analysis has demonstrated that COMB-Radscore is an independent prognostic factor for progression-free survival and overall survival. Moreover, COMB-Radscore demonstrates excellent dynamic predictive performance, with an AUC (ROC) of 0.857, enabling the earlier detection of potential disease progression in patients compared to radiological evaluation solely relying on tumor size. Further radiogenomic analysis showed that the COMB-Radscore was associated with infiltration abundance and functional status of CD8 + T cells. CONCLUSIONS The radiomic model holds promise as a precise, personalized, and dynamic decision support tool for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Xianwen Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zhiwei Liu
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kun Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuedan Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Shu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zeng
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Wenlan Zhou
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Lu D, Zhu X, Mu X, Huang X, Wei F, Qin L, Liu Q, Fu W, Deng Y. Development and validation of a nomogram for predicting bone marrow involvement in lymphoma patients based on 18F-FDG PET radiomics and clinical factors. Ann Nucl Med 2025:10.1007/s12149-025-02041-8. [PMID: 40158053 DOI: 10.1007/s12149-025-02041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/06/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE This study aimed to develop and validate a nomogram combining 18F-FDG PET radiomics and clinical factors to non-invasively predict bone marrow involvement (BMI) in patients with lymphoma. METHODS A radiomics nomogram was developed using monocentric data, randomly divided into a training set (70%) and a test set (30%). Bone marrow biopsy (BMB) served as the gold standard for BMI diagnosis. Independent clinical risk factors were identified through univariate and multivariate logistic regression analyses to construct a clinical model. Radiomics features were extracted from PET and CT images and selected using least absolute shrinkage and selection operator (LASSO) regression, yielding a radiomics score (Radscore) for each patient. Models based on clinical factors, CT Radscore, and PET Radscore were established and evaluated using eight machine learning algorithms to identify the optimal prediction model. A combined model was constructed and presented as a nomogram. Model performance was assessed using the area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis (DCA). RESULTS A total of 160 patients were included, of whom 70 had BMI based on BMB results. The training group comprised 112 patients (BMI: 56, without BMI: 56), while the test group included 48 patients (BMI: 14, without BMI: 34). Independent risk factors, including the number of extranodal involvements and B symptoms, were incorporated into the clinical model. In the clinical model, CT Radscore, and PET Radscore, the AUCs in the test set were 0.820 (95% CI: 0.705-0.935), 0.538 (95% CI: 0.351-0.723), and 0.836 (95% CI: 0.686-0.986). Due to the limited diagnostic performance of CT Radscore, the nomogram was constructed using PET Radscore and the clinical model. The radiomics nomogram achieved AUCs of 0.916 (95% CI: 0.865-0.967) in the training set and 0.863 (95% CI: 0.763-0.964) in the test set. Calibration curves and DCA confirmed the nomogram's discrimination, calibration, and clinical utility in both sets. CONCLUSION By integrating PET Radscore, the number of extranodal involvements, and B symptoms, this 18F-FDG PET radiomics-based nomogram offers a non-invasive method to predict bone marrow status in lymphoma patients, providing nuclear medicine physicians with valuable decision support for pre-treatment evaluation.
Collapse
Affiliation(s)
- Denglu Lu
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xinyu Zhu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xingyu Mu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiaoqi Huang
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Feng Wei
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Lilan Qin
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qixin Liu
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Wei Fu
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Yanyun Deng
- Department of Nuclear Medicine, Liuzhou Workers' Hospital, Liuzhou, 545000, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
12
|
Kotoulas SC, Spyratos D, Porpodis K, Domvri K, Boutou A, Kaimakamis E, Mouratidou C, Alevroudis I, Dourliou V, Tsakiri K, Sakkou A, Marneri A, Angeloudi E, Papagiouvanni I, Michailidou A, Malandris K, Mourelatos C, Tsantos A, Pataka A. A Thorough Review of the Clinical Applications of Artificial Intelligence in Lung Cancer. Cancers (Basel) 2025; 17:882. [PMID: 40075729 PMCID: PMC11898928 DOI: 10.3390/cancers17050882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 02/06/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
According to data from the World Health Organization (WHO), lung cancer is becoming a global epidemic. It is particularly high in the list of the leading causes of death not only in developed countries, but also worldwide; furthermore, it holds the leading place in terms of cancer-related mortality. Nevertheless, many breakthroughs have been made the last two decades regarding its management, with one of the most prominent being the implementation of artificial intelligence (AI) in various aspects of disease management. We included 473 papers in this thorough review, most of which have been published during the last 5-10 years, in order to describe these breakthroughs. In screening programs, AI is capable of not only detecting suspicious lung nodules in different imaging modalities-such as chest X-rays, computed tomography (CT), and positron emission tomography (PET) scans-but also discriminating between benign and malignant nodules as well, with success rates comparable to or even better than those of experienced radiologists. Furthermore, AI seems to be able to recognize biomarkers that appear in patients who may develop lung cancer, even years before this event. Moreover, it can also assist pathologists and cytologists in recognizing the type of lung tumor, as well as specific histologic or genetic markers that play a key role in treating the disease. Finally, in the treatment field, AI can guide in the development of personalized options for lung cancer patients, possibly improving their prognosis.
Collapse
Affiliation(s)
- Serafeim-Chrysovalantis Kotoulas
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Dionysios Spyratos
- Pulmonary Department, Unit of thoracic Malignancies Research, General Hospital of Thessaloniki “G. Papanikolaou”, Aristotle’s University of Thessaloniki, Leoforos Papanikolaou Municipality of Chortiatis, 57010 Thessaloniki, Greece; (D.S.); (K.P.); (K.D.)
| | - Konstantinos Porpodis
- Pulmonary Department, Unit of thoracic Malignancies Research, General Hospital of Thessaloniki “G. Papanikolaou”, Aristotle’s University of Thessaloniki, Leoforos Papanikolaou Municipality of Chortiatis, 57010 Thessaloniki, Greece; (D.S.); (K.P.); (K.D.)
| | - Kalliopi Domvri
- Pulmonary Department, Unit of thoracic Malignancies Research, General Hospital of Thessaloniki “G. Papanikolaou”, Aristotle’s University of Thessaloniki, Leoforos Papanikolaou Municipality of Chortiatis, 57010 Thessaloniki, Greece; (D.S.); (K.P.); (K.D.)
| | - Afroditi Boutou
- Pulmonary Department General, Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (A.B.); (A.T.)
| | - Evangelos Kaimakamis
- 1st ICU, Medical Informatics Laboratory, General Hospital of Thessaloniki “G. Papanikolaou”, Aristotle’s University of Thessaloniki, Leoforos Papanikolaou Municipality of Chortiatis, 57010 Thessaloniki, Greece;
| | - Christina Mouratidou
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Ioannis Alevroudis
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Vasiliki Dourliou
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Kalliopi Tsakiri
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Agni Sakkou
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Alexandra Marneri
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Elena Angeloudi
- Adult ICU, General Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (C.M.); (I.A.); (V.D.); (K.T.); (A.S.); (A.M.); (E.A.)
| | - Ioanna Papagiouvanni
- 4th Internal Medicine Department, General Hospital of Thessaloniki “Ippokrateio”, Aristotle’s University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece;
| | - Anastasia Michailidou
- 2nd Propaedeutic Internal Medicine Department, General Hospital of Thessaloniki “Ippokrateio”, Aristotle’s University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece;
| | - Konstantinos Malandris
- 2nd Internal Medicine Department, General Hospital of Thessaloniki “Ippokrateio”, Aristotle’s University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece;
| | - Constantinos Mourelatos
- Biology and Genetics Laboratory, Aristotle’s University of Thessaloniki, 54624 Thessaloniki, Greece;
| | - Alexandros Tsantos
- Pulmonary Department General, Hospital of Thessaloniki “Ippokrateio”, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (A.B.); (A.T.)
| | - Athanasia Pataka
- Respiratory Failure Clinic and Sleep Laboratory, General Hospital of Thessaloniki “G. Papanikolaou”, Aristotle’s University of Thessaloniki, Leoforos Papanikolaou Municipality of Chortiatis, 57010 Thessaloniki, Greece;
| |
Collapse
|
13
|
Nie F, Pei X, Du J, Shi W, Wang J, Feng L, Liu Y. Multiomics-Based Deep Learning Prediction of Prognosis and Therapeutic Response in Patients With Extensive-Stage Small Cell Lung Cancer Receiving Chemoimmunotherapy: A Retrospective Cohort Study. Int J Gen Med 2025; 18:981-996. [PMID: 40026810 PMCID: PMC11869764 DOI: 10.2147/ijgm.s506485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Objective This study aimed to develop a clinical early warning prediction model to evaluate the prognosis and response to chemoimmunotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC), thereby guiding clinical decision-making. Methods A retrospective analysis was conducted on the clinical data and radiomics parameters of 309 patients with ES-SCLC hospitalized at Baotou Cancer Hospital from February 2020 to September 2024. Patients were divided into reactive and non-reactive groups based on their response to chemoimmunotherapy.Machine learning algorithms (including random forests, decision trees, artificial neural networks, and generalized linear regression) were used to predict the combined treatment response. The model's predictive ability was evaluated using the receiver operating characteristic (ROC) curve and clinical decision curve analysis(DCA). The prognostic evaluation of patients receiving combination therapy was based on the COX regression model, with predictive performance assessed through nomogram visualization and calibration curves. Results Out of 309 patients with ES-SCLC, 248 (80.26%) responded to combination therapy. Logistic regression and Least absolute shrinkage and selection operator (LASSO) regression analyses identified Energy, sum of squares(SOS), mean sum(MES), sum variance(SUV), sum entropy(SUE), difference variance(DIV), and pathomics score as independent risk factors for treatment response. The area under the ROC curve for predicting treatment response using machine learning were 0.764 (95% confidence interval [CI]: 0.707~0.821) and 0.901 (95% CI: 0.846~0.956) in the training and validation sets. The C-index of the radiomics and pathomics prognostic nomogram model based on the COX prognostic model was 0.766 and 0.812 in those sets, respectively. Conclusion We developed prediction model based on multi-omics demonstrated satisfactory performance in predicting chemoimmunotherapy response in patients with ES-SCLC. The random forest prediction model, in particular, provides accurate response and prognostic risk assessments, thereby assisting clinical decision-making.
Collapse
Affiliation(s)
- Fang Nie
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Xiufeng Pei
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Jiale Du
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Wanting Shi
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Jianying Wang
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Lu Feng
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| | - Yonggang Liu
- Department of Thoracic Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, 014000, People’s Republic of China
| |
Collapse
|
14
|
Dong N, Wei S, Zheng L, Huang D, Zhang G, Li Y, Zhang H, Wang A, Huang R, Zhao X, Liang P. Nomogram integrating clinical-radiological and radiomics features for differentiating invasive from non-invasive pulmonary adenocarcinomas presenting as ground-glass nodules. Am J Cancer Res 2025; 15:797-810. [PMID: 40084360 PMCID: PMC11897637 DOI: 10.62347/aoan9966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE To construct a nomogram incorporating clinical-radiological and radiomics features from computed tomography (CT) for distinguishing invasive adenocarcinoma (IAC) from adenocarcinoma in situ (AIS) and minimally invasive adenocarcinoma (MIA) in ground-glass nodules (GGNs). METHODS This retrospective study included 473 GGN patients with postoperative pathological confirmation of AIS, MIA, or IAC. The training set comprised 257 patients from Yantaishan Hospital, while the test set, used for external validation, included 216 patients from the Affiliated Hospital of Binzhou Medical College. Radiomics features were selected, and a radiomics model was constructed using least absolute shrinkage and selection operator (LASSO) and minimum redundancy maximum relevance (mRMR) methods. A clinical-radiological model was developed using univariate and multivariate logistic regression. The nomogram was generated by combining the two models. Its performance was evaluated via receiver operating characteristic (ROC) curve analysis, calibration curve analysis, and decision curve analysis (DCA). RESULTS The radiomics model included 11 features, while the clinical-radiological model incorporated lobulation, age, and long diameter. The nomogram outperformed both individual models in terms of accuracy and area under the curve (AUC) in both the training and test sets. Calibration curve analysis confirmed good consistency between actual and predicted outcomes, and DCA indicated the nomogram's clinical utility. CONCLUSION The nomogram is a non-invasive, accurate tool for preoperative differentiation of GGN types, providing valuable guidance for clinicians in treatment planning.
Collapse
Affiliation(s)
- Ning Dong
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Sirong Wei
- Department of Vascular Intervention, Yantai Qishan HospitalYantai 264001, Shandong, China
| | - Lei Zheng
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Delong Huang
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Guowei Zhang
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Yunxin Li
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Hu Zhang
- Department of Radiology, Affiliated Hospital of Binzhou Medical CollegeBinzhou 256603, Shandong, China
| | - Aijie Wang
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Ranran Huang
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Xinyao Zhao
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| | - Peng Liang
- Department of Radiology, Yantaishan HospitalYantai 264003, Shandong, China
| |
Collapse
|
15
|
Lv C, Zhang G, Xu B, Huang M, Zhang Y, Kou M. Predictive value of CT radiomics and inflammatory markers for pulmonary adenocarcinoma spread through air spaces. Am J Cancer Res 2025; 15:587-600. [PMID: 40084350 PMCID: PMC11897614 DOI: 10.62347/ubdr6353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES To evaluate the predictive value of combining CT radiomics features and inflammatory markers for the preoperative prediction of spread through air spaces (STAS) in pulmonary adenocarcinoma. METHODS In this retrospective study, we analyzed data from 256 patients diagnosed with pulmonary adenocarcinoma between 2021 and 2023. Patients were categorized into two groups based on the presence (n = 115) or absence (n = 141) of STAS, as confirmed by histopathological examination. CT imaging data and routine blood test results, including inflammatory markers, were collected. A validation cohort of 233 patients was included for external validation. Statistical analyses, including univariate and multivariate logistic regression, were performed to identify independent predictors of STAS. Model performance was assessed using Receiver Operating Characteristic curve analysis. RESULTS Key CT radiomics features, such as density, satellite lesions, irregular shape, spiculation, vascular convergence, and the vacuole sign, were significantly associated with STAS. Among inflammatory markers, a lower lymphocyte-to-monocyte ratio (LMR) and higher neutrophil-to-lymphocyte (NLR) and platelet-to-lymphocyte ratios (PLR) were predictive of STAS. The combined predictive model, integrating CT radiomics and inflammatory markers, demonstrated a high discriminatory ability, achieving an area under the curve of 0.915, which was externally validated with an AUC of 0.847. CONCLUSIONS The combination of CT radiomics and inflammatory markers provides an effective, non-invasive preoperative tool for predicting STAS in pulmonary adenocarcinoma, aiding in early prognostication and treatment planning.
Collapse
Affiliation(s)
- Changlei Lv
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| | - Guoping Zhang
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| | - Bingqiang Xu
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| | - Minggang Huang
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| | - Yan Zhang
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| | - Mingqing Kou
- Department of Radiology, Shaanxi Provincial People's Hospital Xi'an 710068, Shaanxi, China
| |
Collapse
|
16
|
Wang H, Qiu J, Lu W, Xie J, Ma J. Radiomics based on multiple machine learning methods for diagnosing early bone metastases not visible on CT images. Skeletal Radiol 2025; 54:335-343. [PMID: 39028463 DOI: 10.1007/s00256-024-04752-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study utilizes [99mTc]-methylene diphosphate (MDP) single photon emission computed tomography (SPECT) images as a reference standard to evaluate whether the integration of radiomics features from computed tomography (CT) and machine learning algorithms can identify microscopic early bone metastases. Additionally, we also determine the optimal machine learning approach. MATERIALS AND METHODS We retrospectively studied 63 patients with early bone metastasis from July 2020 to March 2023. The ITK-SNAP software was used to delineate early bone metastases and normal bone tissue in SPECT images of each patient, which were then registered onto CT images to outline the volume of interest (VOI). The VOI includes 63 early bone metastasis volumes and 63 normal bone tissue volumes. 126 VOIs were randomly distributed in a 7:3 ratio between the training and testing groups, and 944 radiomics features were extracted from every VOI. We established 20 machine learning models using 5 feature selection algorithms and 4 classification methods. Evaluate the performance of the model using the area under the receiver operating characteristic curve (AUC). RESULTS Most machine learning models demonstrated outstanding discriminative capacity, with AUCs higher than 0.70. Notably, the K-Nearest Neighbors (KNN) classifier exhibited significant performance improvement compared to the other four classifiers. Specifically, the model constructed utilizing eXtreme Gradient Boosting (XGBoost) feature selection method integrated with KNN classifier achieved the maximum AUC, which is 0.989 in the training set and 0.975 in the testing set. CONCLUSIONS Radiomics features integrated with machine learning methods can identify early bone metastases that are not visible on CT images. In our analysis, KNN is considered the optimal classification method.
Collapse
Affiliation(s)
- Huili Wang
- College of Preventive Medicine & Institute of Radiation Medicine, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250012, China
| | - Jianfeng Qiu
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China
| | - Weizhao Lu
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China
| | - Jindong Xie
- College of Preventive Medicine & Institute of Radiation Medicine, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250012, China.
| | - Junchi Ma
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China.
| |
Collapse
|
17
|
Tsui WHA, Ding SC, Jiang P, Lo YMD. Artificial intelligence and machine learning in cell-free-DNA-based diagnostics. Genome Res 2025; 35:1-19. [PMID: 39843210 PMCID: PMC11789496 DOI: 10.1101/gr.278413.123] [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] [Indexed: 01/24/2025]
Abstract
The discovery of circulating fetal and tumor cell-free DNA (cfDNA) molecules in plasma has opened up tremendous opportunities in noninvasive diagnostics such as the detection of fetal chromosomal aneuploidies and cancers and in posttransplantation monitoring. The advent of high-throughput sequencing technologies makes it possible to scrutinize the characteristics of cfDNA molecules, opening up the fields of cfDNA genetics, epigenetics, transcriptomics, and fragmentomics, providing a plethora of biomarkers. Machine learning (ML) and/or artificial intelligence (AI) technologies that are known for their ability to integrate high-dimensional features have recently been applied to the field of liquid biopsy. In this review, we highlight various AI and ML approaches in cfDNA-based diagnostics. We first introduce the biology of cell-free DNA and basic concepts of ML and AI technologies. We then discuss selected examples of ML- or AI-based applications in noninvasive prenatal testing and cancer liquid biopsy. These applications include the deduction of fetal DNA fraction, plasma DNA tissue mapping, and cancer detection and localization. Finally, we offer perspectives on the future direction of using ML and AI technologies to leverage cfDNA fragmentation patterns in terms of methylomic and transcriptional investigations.
Collapse
Affiliation(s)
- W H Adrian Tsui
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Spencer C Ding
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Peiyong Jiang
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Y M Dennis Lo
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
18
|
Qiao T, Cheng Z, Duan Y. Innovative applications and future trends of multiparametric PET in the assessment of immunotherapy efficacy. Front Oncol 2025; 14:1530507. [PMID: 39902124 PMCID: PMC11788151 DOI: 10.3389/fonc.2024.1530507] [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: 11/19/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Background The integration of multiparametric PET (Positron Emission Tomography.) imaging and multi-omics data has demonstrated significant clinical potential in predicting the efficacy of cancer immunotherapies. However, the specific predictive power and underlying mechanisms remain unclear. Objective This review systematically evaluates the application of multiparametric PET imaging metrics (e.g., SUVmax [Maximum Standardized Uptake Value], MTV [Metabolic Tumor Volume], and TLG [Total Lesion Glycolysis]) in predicting the efficacy of immunotherapies, including PD-1/PD-L1 inhibitors and CAR-T therapy, and explores their potential role in improving predictive accuracy when integrated with multi-omics data. Methods A systematic search of PubMed, Embase, and Web of Science databases identified studies evaluating the efficacy of immunotherapy using longitudinal PET/CT data and RECIST or iRECIST criteria. Only original prospective or retrospective studies were included for analysis. Review articles and meta-analyses were consulted for additional references but excluded from quantitative analysis. Studies lacking standardized efficacy evaluations were excluded to ensure data integrity and quality. Results Multiparametric PET imaging metrics exhibited high predictive capability for efficacy across various immunotherapies. Metabolic parameters such as SUVmax, MTV, and TLG were significantly correlated with treatment response rates, progression-free survival (PFS), and overall survival (OS). The integration of multi-omics data (including genomics and proteomics) with PET imaging enhanced the sensitivity and accuracy of efficacy prediction. Through integrated analysis, PET metabolic parameters demonstrated potential in predicting immune therapy response patterns, such as pseudo-progression and hyper-progression. Conclusion The integration of multiparametric PET imaging and multi-omics data holds broad potential for predicting the efficacy of immunotherapies and may support the development of personalized treatment strategies. Future validation using large-scale, multicenter datasets is needed to further advance precision medicine in cancer immunotherapy.
Collapse
Affiliation(s)
- Tingting Qiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Graduate School, Shandong First Medical University, Jinan, China
| | - Zhaoping Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yanhua Duan
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
19
|
Chang L, Liu J, Zhu J, Guo S, Wang Y, Zhou Z, Wei X. Advancing precision medicine: the transformative role of artificial intelligence in immunogenomics, radiomics, and pathomics for biomarker discovery and immunotherapy optimization. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0376. [PMID: 39749734 PMCID: PMC11795263 DOI: 10.20892/j.issn.2095-3941.2024.0376] [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: 09/18/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Artificial intelligence (AI) is significantly advancing precision medicine, particularly in the fields of immunogenomics, radiomics, and pathomics. In immunogenomics, AI can process vast amounts of genomic and multi-omic data to identify biomarkers associated with immunotherapy responses and disease prognosis, thus providing strong support for personalized treatments. In radiomics, AI can analyze high-dimensional features from computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT) images to discover imaging biomarkers associated with tumor heterogeneity, treatment response, and disease progression, thereby enabling non-invasive, real-time assessments for personalized therapy. Pathomics leverages AI for deep analysis of digital pathology images, and can uncover subtle changes in tissue microenvironments, cellular characteristics, and morphological features, and offer unique insights into immunotherapy response prediction and biomarker discovery. These AI-driven technologies not only enhance the speed, accuracy, and robustness of biomarker discovery but also significantly improve the precision, personalization, and effectiveness of clinical treatments, and are driving a shift from empirical to precision medicine. Despite challenges such as data quality, model interpretability, integration of multi-modal data, and privacy protection, the ongoing advancements in AI, coupled with interdisciplinary collaboration, are poised to further enhance AI's roles in biomarker discovery and immunotherapy response prediction. These improvements are expected to lead to more accurate, personalized treatment strategies and ultimately better patient outcomes, marking a significant step forward in the evolution of precision medicine.
Collapse
Affiliation(s)
- Luchen Chang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiamei Liu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuyue Guo
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yao Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhiwei Zhou
- Departments of Biochemistry and Radiation Oncology, UT Southwestern Medical Center, Dallas 75390, USA
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
20
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
21
|
Zhao X, Wang Y, Xue M, Ding Y, Zhang H, Wang K, Ren J, Li X, Xu M, Lv J, Wang Z, Sun D. Preoperative assessment of tertiary lymphoid structures in stage I lung adenocarcinoma using CT radiomics: a multicenter retrospective cohort study. Cancer Imaging 2024; 24:167. [PMID: 39696659 DOI: 10.1186/s40644-024-00813-5] [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: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE To develop a multimodal predictive model, Radiomics Integrated TLSs System (RAITS), based on preoperative CT radiomic features for the identification of TLSs in stage I lung adenocarcinoma patients and to evaluate its potential in prognosis stratification and guiding personalized treatment. METHODS The most recent preoperative chest CT thin-slice scans and postoperative hematoxylin and eosin-stained pathology sections of patients diagnosed with stage I LUAD were retrospectively collected. Tumor segmentation was achieved using an automatic virtual adversarial training segmentation algorithm based on a three-dimensional U-shape convolutional neural network (3D U-Net). Radiomic features were extracted from the tumor and peritumoral areas, with extensions of 2 mm, 4 mm, 6 mm, and 8 mm, respectively, and deep learning image features were extracted through a convolutional neural network. Subsequently, the RAITS was constructed. The performance of RAITS was then evaluated in both the train and validation cohorts. RESULTS RAITS demonstrated superior AUC, sensitivity, and specificity in both the training and external validation cohorts, outperforming traditional unimodal models. In the validation cohort, RAITS achieved an AUC of 0.78 (95% CI, 0.69-0.88) and showed higher net benefits across most threshold ranges. RAITS exhibited strong discriminative ability in risk stratification, with p < 0.01 in the training cohort and p = 0.02 in the validation cohort, consistent with the actual predictive performance of TLSs, where TLS-positive patients had significantly higher recurrence-free survival (RFS) compared to TLS-negative patients (p = 0.04 in the training cohort, p = 0.02 in the validation cohort). CONCLUSION As a multimodal predictive model based on preoperative CT radiomic features, RAITS demonstrated excellent performance in identifying TLSs in stage I LUAD and holds potential value in clinical decision-making.
Collapse
Affiliation(s)
| | - Yuhang Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Mengli Xue
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Yun Ding
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Han Zhang
- Chest hospital, Tianjin University, Tianjin, China
| | - Kai Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Jie Ren
- Department of Thoracic Surgery, Tianjin Jinnan Hospital, Tianjin, China
| | - Xin Li
- Chest hospital, Tianjin University, Tianjin, China
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Meilin Xu
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Jun Lv
- Department of Imaging, Tianjin Chest Hospital, Tianjin, China
| | - Zixiao Wang
- Department of Thoracic Surgery, Qinhuangdao First Hospital, Hebei Province, China
| | - Daqiang Sun
- Chest hospital, Tianjin University, Tianjin, China.
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China.
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
22
|
Qiao J, Liu B, Xin J, Shen S, Ma H, Pan S. Prediction of Prognosis and Response to Androgen Deprivation Therapy in Intermediate to High-Risk Prostate Cancer Using 18F-FDG PET/CT Radiomics. Acad Radiol 2024; 31:5008-5021. [PMID: 39019687 DOI: 10.1016/j.acra.2024.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 07/19/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to predict intermediate to high-risk prostate cancer (PCa) prognosis based on 18-fluoro-2-deoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) radiomics. Additionally, subgroup analysis will be performed on the androgen deprivation therapy (ADT) group and the metastatic PCa group. MATERIALS AND METHODS In the retrospective analysis of 104 intermediate to high-risk PCa patients who underwent 18F-FDG PET/CT prior to treatment. The data set was divided into a training set (n = 72) and a testing set (n = 32). Two different PET/CT models were constructed using multivariate logistic regression with cross-validation: radiomics model A and an alternative ensemble learning-based model B. The superior model was then selected to develop a radiomics nomogram. Separate models were also developed for the ADT and metastatic PCa subgroups. RESULTS Model A, which integrates eight radiomics features showed excellent performance with an area under curve (AUC) of 0.844 in the training set and 0.804 in the testing set. The radiomics nomogram incorporating the radiomics score (radscore) from model A and the tumor-to-liver ratio (TLR) showed good prognostic accuracy in the testing set with an AUC of 0.827. In the subgroup analyses for endocrine therapy and metastatic cancer, the PET/CT radiomics model showed AUCs of 0.845 and 0.807 respectively, suggesting its potential effectiveness. CONCLUSION The study establishes the utility of the 18F-FDG PET/CT radiomics nomogram in predicting the prognosis of intermediate to high-risk PCa patients, indicating its potential for clinical application.
Collapse
Affiliation(s)
- Jianyi Qiao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Siang Shen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Han Ma
- Department of Nuclear Medicine, People's Hospital of Liaoning Province, Shenyang, China
| | - Shen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
23
|
Xv Y, Wei Z, Lv F, Jiang Q, Guo H, Zheng Y, Zhang X, Xiao M. Multiparameter computed tomography (CT) radiomics signature fusion-based model for the preoperative prediction of clear cell renal cell carcinoma nuclear grade: a multicenter development and external validation study. Quant Imaging Med Surg 2024; 14:7031-7045. [PMID: 39429571 PMCID: PMC11485359 DOI: 10.21037/qims-24-35] [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: 01/07/2024] [Accepted: 08/01/2024] [Indexed: 10/22/2024]
Abstract
Background The preoperative prediction of the pathological nuclear grade of clear cell renal cell carcinoma (CCRCC) is crucial for clinical decision making. However, radiomics features from one or two computed tomography (CT) phases are required to predict the CCRCC grade, which reduces the predictive performance and generalizability of this method. We aimed to develop and externally validate a multiparameter CT radiomics-based model for predicting the World Health Organization/International Society of Urological Pathology (WHO/ISUP) grade of CCRCC. Methods A total of 500 CCRCC patients at The First, Second, and Yongchuan Hospitals of Chongqing Medical University between January 2016 and May 2022 were retrospectively enrolled in this study. The patients were divided into the training set (n=268), internal testing set (n=115), and two external testing sets (testing set 1, n=62; testing set 2, n=55). Radiomics features were extracted from multi-phase CT images, and radiomics signatures (RSs) were created by least absolute shrinkage and selection operator (LASSO) regression. In addition, a clinical model was developed. A combined model was also established that integrated the RSs with the clinical factors, and was visualized via a nomogram. The performance of the established model was assessed using area under the curve (AUC) values, a calibration curve analysis, and a decision curve analysis (DCA). Results Among the four RSs and the clinical model, the RS-Triphasic had the best predictive performance with AUCs of 0.88 [95% confidence interval (CI): 0.85-0.91] and 0.84 (95% CI: 0.74-0.95) in the training and testing sets, respectively, and 0.82 (95% CI: 0.72-0.93) and 0.82 (95% CI: 0.71-0.93) in external testing sets 1 and 2. Integrating the RS-Triphasic, RS-corticomedullary phase (CMP), RS-nephrographic phase (NP), RS-non-contrast phase (NCP) with the clinical risk factors, a combined model was established with AUCs of 0.92 (95% CI: 0.89-0.94), 0.86 (95% CI: 0.76-0.95), 0.84 (95% CI: 0.73-0.95), and 0.82 (95% CI: 0.70-0.94) for the training, internal testing, and external testing sets 1 and 2, respectively. The DCA indicated that the nomogram had a greater overall net benefit than the clinical and radiomics models. Conclusions The multiparameter CT RS fusion-based model had high accuracy in differentiating between high- and low-grade CCRCC preoperatively. Thus, it has great potential as a useful tool for personalized treatment planning and clinical decision making for CCRCC patients.
Collapse
Affiliation(s)
- Yingjie Xv
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongjie Wei
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Jiang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoming Guo
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yineng Zheng
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Zhang
- Department of Urology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Mingzhao Xiao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
McGale JP, Chen DL, Trebeschi S, Farwell MD, Wu AM, Cutler CS, Schwartz LH, Dercle L. Artificial intelligence in immunotherapy PET/SPECT imaging. Eur Radiol 2024; 34:5829-5841. [PMID: 38355986 DOI: 10.1007/s00330-024-10637-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVE Immunotherapy has dramatically altered the therapeutic landscape for oncology, but more research is needed to identify patients who are likely to achieve durable clinical benefit and those who may develop unacceptable side effects. We investigated the role of artificial intelligence in PET/SPECT-guided approaches for immunotherapy-treated patients. METHODS We performed a scoping review of MEDLINE, CENTRAL, and Embase databases using key terms related to immunotherapy, PET/SPECT imaging, and AI/radiomics through October 12, 2022. RESULTS Of the 217 studies identified in our literature search, 24 relevant articles were selected. The median (interquartile range) sample size of included patient cohorts was 63 (157). Primary tumors of interest were lung (n = 14/24, 58.3%), lymphoma (n = 4/24, 16.7%), or melanoma (n = 4/24, 16.7%). A total of 28 treatment regimens were employed, including anti-PD-(L)1 (n = 13/28, 46.4%) and anti-CTLA-4 (n = 4/28, 14.3%) monoclonal antibodies. Predictive models were built from imaging features using univariate radiomics (n = 7/24, 29.2%), radiomics (n = 12/24, 50.0%), or deep learning (n = 5/24, 20.8%) and were most often used to prognosticate (n = 6/24, 25.0%) or describe tumor phenotype (n = 5/24, 20.8%). Eighteen studies (75.0%) performed AI model validation. CONCLUSION Preliminary results suggest broad potential for the application of AI-guided immunotherapy management after further validation of models on large, prospective, multicenter cohorts. CLINICAL RELEVANCE STATEMENT This scoping review describes how artificial intelligence models are built to make predictions based on medical imaging and explores their application specifically in the PET and SPECT examination of immunotherapy-treated cancers. KEY POINTS • Immunotherapy has drastically altered the cancer treatment landscape but is known to precipitate response patterns that are not accurately accounted for by traditional imaging methods. • There is an unmet need for better tools to not only facilitate in-treatment evaluation but also to predict, a priori, which patients are likely to achieve a good response with a certain treatment as well as those who are likely to develop side effects. • Artificial intelligence applied to PET/SPECT imaging of immunotherapy-treated patients is mainly used to make predictions about prognosis or tumor phenotype and is built from baseline, pre-treatment images. Further testing is required before a true transition to clinical application can be realized.
Collapse
Affiliation(s)
- Jeremy P McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Delphine L Chen
- Department of Molecular Imaging and Therapy, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School of Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna M Wu
- Department of Immunology and Theranostics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Cathy S Cutler
- Collider Accelerator Department, Brookhaven National Laboratory, Upton, NY, USA
| | - Lawrence H Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
25
|
Li C, Zhou Z, Hou L, Hu K, Wu Z, Xie Y, Ouyang J, Cai X. A novel machine learning model for efficacy prediction of immunotherapy-chemotherapy in NSCLC based on CT radiomics. Comput Biol Med 2024; 178:108638. [PMID: 38897152 DOI: 10.1016/j.compbiomed.2024.108638] [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: 01/29/2024] [Revised: 04/16/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024]
Abstract
Lung cancer is categorized into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer. Of these, NSCLC accounts for approximately 85% of all cases and encompasses varieties such as squamous cell carcinoma and adenocarcinoma. For patients with advanced NSCLC that do not have oncogene addiction, the preferred treatment approach is a combination of immunotherapy and chemotherapy. However, the progression-free survival (PFS) typically ranges only from about 6 to 8 months, accompanied by certain adverse events. In order to carry out individualized treatment more effectively, it is urgent to accurately screen patients with PFS for more than 12 months under this treatment regimen. Therefore, this study undertook a retrospective collection of pulmonary CT images from 60 patients diagnosed with NSCLC treated at the First Affiliated Hospital of Wenzhou Medical University. It developed a machine learning model, designated as bSGSRIME-SVM, which integrates the rime optimization algorithm with self-adaptive Gaussian kernel probability search (SGSRIME) and support vector machine (SVM) classifier. Specifically, the model initiates its process by employing the SGSRIME algorithm to identify pivotal image features. Subsequently, it utilizes an SVM classifier to assess these features, aiming to enhance the model's predictive accuracy. Initially, the superior optimization capability and robustness of SGSRIME in IEEE CEC 2017 benchmark functions were validated. Subsequently, employing color moments and gray-level co-occurrence matrix methods, image features were extracted from images of 60 NSCLC patients undergoing immunotherapy combined with chemotherapy. The developed model was then utilized for analysis. The results indicate a significant advantage of the model in predicting the efficacy of immunotherapy combined with chemotherapy for NSCLC, with an accuracy of 92.381% and a specificity of 96.667%. This lays the foundation for more accurate PFS predictions and personalized treatment plans.
Collapse
Affiliation(s)
- Chengye Li
- Department of Pulmonary and Critical Care Medicine, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Zhifeng Zhou
- Wenzhou University Library, Wenzhou, 325035, China.
| | - Lingxian Hou
- Rehabilitation Department, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, China.
| | - Keli Hu
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, 312000, China; Information Technology R&D Innovation Center of Peking University, Shaoxing, 312000, China.
| | - Zongda Wu
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, 312000, China.
| | - Yupeng Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jinsheng Ouyang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xueding Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
26
|
Liu J, Sui C, Bian H, Li Y, Wang Z, Fu J, Qi L, Chen K, Xu W, Li X. Radiomics based on 18F-FDG PET/CT for prediction of pathological complete response to neoadjuvant therapy in non-small cell lung cancer. Front Oncol 2024; 14:1425837. [PMID: 39132503 PMCID: PMC11310012 DOI: 10.3389/fonc.2024.1425837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose This study aimed to establish and evaluate the value of integrated models involving 18F-FDG PET/CT-based radiomics and clinicopathological information in the prediction of pathological complete response (pCR) to neoadjuvant therapy (NAT) for non-small cell lung cancer (NSCLC). Methods A total of 106 eligible NSCLC patients were included in the study. After volume of interest (VOI) segmentation, 2,016 PET-based and 2,016 CT-based radiomic features were extracted. To select an optimal machine learning model, a total of 25 models were constructed based on five sets of machine learning classifiers combined with five sets of predictive feature resources, including PET-based alone radiomics, CT-based alone radiomics, PET/CT-based radiomics, clinicopathological features, and PET/CT-based radiomics integrated with clinicopathological features. Area under the curves (AUCs) of receiver operator characteristic (ROC) curves were used as the main outcome to assess the model performance. Results The hybrid PET/CT-derived radiomic model outperformed PET-alone and CT-alone radiomic models in the prediction of pCR to NAT. Moreover, addition of clinicopathological information further enhanced the predictive performance of PET/CT-derived radiomic model. Ultimately, the support vector machine (SVM)-based PET/CT radiomics combined clinicopathological information presented an optimal predictive efficacy with an AUC of 0.925 (95% CI 0.869-0.981) in the training cohort and an AUC of 0.863 (95% CI 0.740-0.985) in the test cohort. The developed nomogram involving radiomics and pathological type was suggested as a convenient tool to enable clinical application. Conclusions The 18F-FDG PET/CT-based SVM radiomics integrated with clinicopathological information was an optimal model to non-invasively predict pCR to NAC for NSCLC.
Collapse
Affiliation(s)
- Jianjing Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Haiman Bian
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yue Li
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Jie Fu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Lisha Qi
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
27
|
Zhang M, Kuang B, Zhang J, Peng J, Xia H, Feng X, Peng L. Enhancing prognostic prediction in hepatocellular carcinoma post-TACE: a machine learning approach integrating radiomics and clinical features. Front Med (Lausanne) 2024; 11:1419058. [PMID: 39086938 PMCID: PMC11289890 DOI: 10.3389/fmed.2024.1419058] [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: 04/17/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Objective This study aimed to investigate the use of radiomics features and clinical information by four machine learning algorithms for predicting the prognosis of patients with hepatocellular carcinoma (HCC) who have been treated with transarterial chemoembolization (TACE). Methods A total of 105 patients with HCC treated with TACE from 2002 to 2012 were enrolled retrospectively and randomly divided into two cohorts for training (n = 74) and validation (n = 31) according to a ratio of 7:3. The Spearman rank, random forest, and univariate Cox regression were used to select the optimal radiomics features. Univariate Cox regression was used to select clinical features. Four machine learning algorithms were used to develop the models: random survival forest, eXtreme gradient boosting (XGBoost), gradient boosting, and the Cox proportional hazard regression model. The area under the curve (AUC) and C-index were devoted to assessing the performance of the models in predicting HCC prognosis. Results A total of 1,834 radiomics features were extracted from the computed tomography images of each patient. The clinical risk factors for HCC prognosis were age at diagnosis, TNM stage, and metastasis, which were analyzed using univariate Cox regression. In various models, the efficacy of the combined models generally surpassed that of the radiomics and clinical models. Among four machine learning algorithms, XGBoost exhibited the best performance in combined models, achieving an AUC of 0.979 in the training set and 0.750 in the testing set, demonstrating its strong prognostic prediction capability. Conclusion The superior performance of the XGBoost-based combined model underscores its potential as a powerful tool for enhancing the precision of prognostic assessments for patients with HCC.
Collapse
Affiliation(s)
- Mingqi Zhang
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Second Clinical School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bingling Kuang
- Nanshan College, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingxuan Zhang
- Nanshan College, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingyi Peng
- The Second Clinical School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haoming Xia
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiaobin Feng
- School of Clinical Medicine, Tsinghua University, Beijing, China
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Liang Peng
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
28
|
Chen L, Yin G, Wang Z, Liu Z, Sui C, Chen K, Song T, Xu W, Qi L, Li X. A predictive radiotranscriptomics model based on DCE-MRI for tumor immune landscape and immunotherapy in cholangiocarcinoma. Biosci Trends 2024; 18:263-276. [PMID: 38853000 DOI: 10.5582/bst.2024.01121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.
Collapse
Affiliation(s)
- Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guotao Yin
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
29
|
Xin W, Rixin S, Linrui L, Zhihui Q, Long L, Yu Z. Machine learning-based radiomics for predicting outcomes in cervical cancer patients undergoing concurrent chemoradiotherapy. Comput Biol Med 2024; 177:108593. [PMID: 38801795 DOI: 10.1016/j.compbiomed.2024.108593] [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: 11/22/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
PURPOSES To investigate the value of machine learning-based radiomics for predicting disease-free survival (DFS) and overall survival (OS) undergoing concurrent chemoradiotherapy (CCRT) for patients with locally advanced cervical cancer (LACC). MATERIALS AND METHODS In this multicentre study, 700 patients with IB2-IVA cervical cancer who underwent CCRT with ongoing follow-up were retrospectively analyzed. Three-dimensional radiomics features of primary lesions and its surrounding 5 mm region in T2WI sequences were collected. Six machine learning methods were used to construct the optimal radiomics model for accurate prediction of DFS and OS after CCRT in LACC patients. Eventually, TCGA and GEO databases were used to explore the mechanisms of radiomics in predicting the progression and survival of cervical cancer. This study adhered CLEAR for reporting and its quality was assessed using RQS and METRICS. RESULTS In the prediction of DFS, the RSF model combined tumor and peritumor radiomics demonstrated the best predictive efficacy, with the AUC for predicting 1-year, 3-year, and 5-year DFS in the training, validation, and test sets of 0.986, 0.989, 0.990, and 0.884, 0.838, 0.823, and 0.829, 0.809, 0.841, respectively. In the prediction of OS, the GBM model best performer, with AUC of 0.999, 0.995, 0.978, and 0.981, 0.975, 0.837, and 0.904, 0.860, 0.905. Differential genes in TCGA and GEO suggest that the prediction of radiomics model may be associated with KDELR2 and HK2. CONCLUSION Machine learning-based radiomics models help to predict DFS and OS after CCRT in LACC patients, and the combination of tumor and peritumor information has higher predictive efficacy, which can provide a reliable basis for therapeutic decision-making in cervical cancer patients.
Collapse
Affiliation(s)
- Wang Xin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Su Rixin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Li Linrui
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Qin Zhihui
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Liu Long
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Zhejiang University, Hangzhou, 310000, Zhejiang, China.
| | - Zhang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
30
|
Yao W, Wang Y, Zhao X, He M, Wang Q, Liu H, Zhao J. Automatic diagnosis of pediatric supracondylar humerus fractures using radiomics-based machine learning. Medicine (Baltimore) 2024; 103:e38503. [PMID: 38847664 PMCID: PMC11155539 DOI: 10.1097/md.0000000000038503] [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: 12/05/2023] [Accepted: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
The aim of this study was to construct a classification model for the automatic diagnosis of pediatric supracondylar humerus fractures using radiomics-based machine learning. We retrospectively collected elbow joint Radiographs of children aged 3 to 14 years and manually delineated regions of interest (ROI) using ITK-SNAP. Radiomics features were extracted using pyradiomics, a python-based feature extraction tool. T-tests and the least absolute shrinkage and selection operator (LASSO) algorithm were used to further select the most valuable radiomics features. A logistic regression (LR) model was trained, with an 8:2 split into training and testing sets, and 5-fold cross-validation was performed on the training set. The diagnostic performance of the model was evaluated using receiver operating characteristic curves (ROC) on the testing set. A total of 411 fracture samples and 190 normal samples were included. 1561 features were extracted from each ROI. After dimensionality reduction screening, 40 and 94 features with the most diagnostic value were selected for further classification modeling in anteroposterior and lateral elbow radiographs. The area under the curve (AUC) of anteroposterior and lateral elbow radiographs is 0.65 and 0.72. Radiomics can extract and select the most valuable features from a large number of image features. Supervised machine-learning models built using these features can be used for the diagnosis of pediatric supracondylar humerus fractures.
Collapse
Affiliation(s)
- Wuyi Yao
- Department of Orthopedics, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| | - Yu Wang
- Department of Orthopedics, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| | - Xiaobin Zhao
- Department of Radiology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| | - Man He
- Department of Rehabilitation, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| | - Qian Wang
- Department of Orthopedics, Tianjin Beichen Hospital, Tianjin, PR China
| | - Hanjie Liu
- Department of Orthopedics, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| | - Jingxin Zhao
- Department of Orthopedics, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, PR China
| |
Collapse
|
31
|
Anuntakarun S, Khamjerm J, Tangkijvanich P, Chuaypen N. Classification of Long Non-Coding RNAs s Between Early and Late Stage of Liver Cancers From Non-coding RNA Profiles Using Machine-Learning Approach. Bioinform Biol Insights 2024; 18:11779322241258586. [PMID: 38846329 PMCID: PMC11155358 DOI: 10.1177/11779322241258586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Long non-coding RNAs (lncRNAs), which are RNA sequences greater than 200 nucleotides in length, play a crucial role in regulating gene expression and biological processes associated with cancer development and progression. Liver cancer is a major cause of cancer-related mortality worldwide, notably in Thailand. Although machine learning has been extensively used in analyzing RNA-sequencing data for advanced knowledge, the identification of potential lncRNA biomarkers for cancer, particularly focusing on lncRNAs as molecular biomarkers in liver cancer, remains comparatively limited. In this study, our objective was to identify candidate lncRNAs in liver cancer. We employed an expression data set of lncRNAs from patients with liver cancer, which comprised 40 699 lncRNAs sourced from The CancerLivER database. Various feature selection methods and machine-learning approaches were used to identify these candidate lncRNAs. The results showed that the random forest algorithm could predict lncRNAs using features extracted from the database, which achieved an area under the curve (AUC) of 0.840 for classifying lncRNAs between early (stage 1) and late stages (stages 2, 3, and 4) of liver cancer. Five of 23 significant lncRNAs (WAC-AS1, MAPKAPK5-AS1, ARRDC1-AS1, AC133528.2, and RP11-1094M14.11) were differentially expressed between early and late stage of liver cancer. Based on the Gene Expression Profiling Interactive Analysis (GEPIA) database, higher expression of WAC-AS1, MAPKAPK5-AS1, and ARRDC1-AS1 was associated with shorter overall survival. In conclusion, the classification model could predict the early and late stages of liver cancer using the signature expression of lncRNA genes. The identified lncRNAs might be used as early diagnostic and prognostic biomarkers for patients with liver cancer.
Collapse
Affiliation(s)
- Songtham Anuntakarun
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jakkrit Khamjerm
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biomedical Engineering Program, Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natthaya Chuaypen
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
32
|
Wang P, Luo Z, Luo C, Wang T. Application of a Comprehensive Model Based on CT Radiomics and Clinical Features for Postoperative Recurrence Risk Prediction in Non-small Cell Lung Cancer. Acad Radiol 2024; 31:2579-2590. [PMID: 38172022 DOI: 10.1016/j.acra.2023.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 01/05/2024]
Abstract
RATIONALE AND OBJECTIVES We constructed a comprehensive model by combining the radiomics and clinical features of tumors to predict the recurrence risk of patients with operable stage IA-IIIA non-small cell lung cancer (NSCLC). Our aim was to improve the accuracy of prognostic prediction and provide personalized treatment plans to enhance patient outcomes. MATERIALS AND METHODS We retrospectively analyzed 152 surgically treated patients with pathologically confirmed stage IA-IIIA NSCLC. These patients were randomly divided into a training cohort and a test cohort in an 8:2 ratio. Using the 3D Slicer image computing platform, we manually delineated the regions of interest (ROI) for all lesions and extracted radiomics features using Python. We used the Least Absolute Shrinkage and Selection Operator (LASSO) to select the radiomics features, while the COX multivariate regression model was employed to identify independent clinical risk factors for recurrence. Finally, we utilized logistic regression (LR) to build the model and validated it using the receiver operating characteristic curve (ROC). The predictive performance of the model was evaluated using the concordance index (C-index), and the clinical value of the model was compared through decision curve analysis (DCA). RESULTS We extracted a total of 1562 radiomics features. After feature selection, we retained 29 features. The COX multivariate regression model demonstrated that the N stage was an independent risk factor for postoperative recurrence. In the training and test cohorts, the area under the curve (AUC) values of the radiomics-clinical comprehensive model were 0.972 and 0.937, respectively, while the C-index values were 0.815 and 0.847. These values surpassed those of the standalone clinical model or radiomics model. CONCLUSION Our study demonstrates that a comprehensive model based on CT radiomics and clinical features can effectively stratify the risk of postoperative recurrence in patients with operable NSCLC. It provides a powerful tool for accurately stratifying the risk of high-risk patients after surgery.
Collapse
Affiliation(s)
- Peiwen Wang
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhilin Luo
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chengwen Luo
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Tianhu Wang
- Department of Thoracic Surgery, Third Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China.
| |
Collapse
|
33
|
Yuan L, An L, Zhu Y, Duan C, Kong W, Jiang P, Yu QQ. Machine Learning in Diagnosis and Prognosis of Lung Cancer by PET-CT. Cancer Manag Res 2024; 16:361-375. [PMID: 38699652 PMCID: PMC11063459 DOI: 10.2147/cmar.s451871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
As a disease with high morbidity and high mortality, lung cancer has seriously harmed people's health. Therefore, early diagnosis and treatment are more important. PET/CT is usually used to obtain the early diagnosis, staging, and curative effect evaluation of tumors, especially lung cancer, due to the heterogeneity of tumors and the differences in artificial image interpretation and other reasons, it also fails to entirely reflect the real situation of tumors. Artificial intelligence (AI) has been applied to all aspects of life. Machine learning (ML) is one of the important ways to realize AI. With the help of the ML method used by PET/CT imaging technology, there are many studies in the diagnosis and treatment of lung cancer. This article summarizes the application progress of ML based on PET/CT in lung cancer, in order to better serve the clinical. In this study, we searched PubMed using machine learning, lung cancer, and PET/CT as keywords to find relevant articles in the past 5 years or more. We found that PET/CT-based ML approaches have achieved significant results in the detection, delineation, classification of pathology, molecular subtyping, staging, and response assessment with survival and prognosis of lung cancer, which can provide clinicians a powerful tool to support and assist in critical daily clinical decisions. However, ML has some shortcomings such as slightly poor repeatability and reliability.
Collapse
Affiliation(s)
- Lili Yuan
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Lin An
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Yandong Zhu
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Chongling Duan
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Weixiang Kong
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Qing-Qing Yu
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| |
Collapse
|
34
|
Li L, Jiang H, Zeng B, Wang X, Bao Y, Chen C, Ma L, Yuan J. Liquid biopsy in lung cancer. Clin Chim Acta 2024; 554:117757. [PMID: 38184141 DOI: 10.1016/j.cca.2023.117757] [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: 10/23/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Lung cancer is a highly prevalent malignancy worldwide and the primary cause of mortality. The absence of systematic and standardized diagnostic approaches for identifying potential pulmonary nodules, early-stage cancers, and indeterminate tumors has led clinicians to consider tissue biopsy and pathological sections as the preferred method for clinical diagnosis, often regarded as the gold standard. The conventional tissue biopsy is an invasive procedure that does not adequately capture the diverse characteristics and evolving nature of tumors. Recently, the concept of 'liquid biopsy' has gained considerable attention as a promising solution. Liquid biopsy is a non-invasive approach that facilitates repeated analysis, enabling real-time monitoring of tumor recurrence, metastasis, and response to treatment. Currently, liquid biopsy includes circulating tumor cells, circulating cell-free DNA, circulating tumor DNA, circulating cell-free RNA, extracellular vesicles, and other proteins and metabolites. With rapid progress in molecular technology, liquid biopsy has emerged as a highly promising and intriguing approach, yielding compelling results. This article critically examines the significant role and potential clinical implications of liquid biopsy in the diagnosis, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Lan Li
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haixia Jiang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Bingjie Zeng
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Xianzhao Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Yunxia Bao
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Changqiang Chen
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Jin Yuan
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
35
|
Yu Y, Wang Z, Wang Q, Su X, Li Z, Wang R, Guo T, Gao W, Wang H, Zhang B. Radiomic model based on magnetic resonance imaging for predicting pathological complete response after neoadjuvant chemotherapy in breast cancer patients. Front Oncol 2024; 13:1249339. [PMID: 38357424 PMCID: PMC10865896 DOI: 10.3389/fonc.2023.1249339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/02/2023] [Indexed: 02/16/2024] Open
Abstract
Purpose To establish a model combining radiomic and clinicopathological factors based on magnetic resonance imaging to predict pathological complete response (pCR) after neoadjuvant chemotherapy in breast cancer patients. Method MRI images and clinicopathologic data of 329 eligible breast cancer patients from the Affiliated Hospital of Qingdao University from August 2018 to August 2022 were included in this study. All patients received neoadjuvant chemotherapy (NAC), and imaging examinations were performed before and after NAC. A total of 329 patients were randomly allocated to a training set and a test set at a ratio of 7:3. We mainly studied the following three types of prediction models: radiomic models, clinical models, and clinical-radiomic models. All models were evaluated using subject operating characteristic curve analysis and area under the curve (AUC), decision curve analysis (DCA) and calibration curves. Results The AUCs of the clinical prediction model, independent imaging model and clinical combined imaging model in the training set were 0.864 0.968 and 0.984, and those in the test set were 0.724, 0.754 and 0.877, respectively. According to DCA and calibration curves, the clinical-radiomic model showed good predictive performance in both the training set and the test set, and we found that we had developed a more concise clinical-radiomic nomogram. Conclusion We have developed a clinical-radiomic model by integrating radiomic features and clinical factors to predict pCR after NAC in breast cancer patients, thereby contributing to the personalized treatment of patients.
Collapse
Affiliation(s)
- Yimiao Yu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhibo Wang
- Department of Gastroenterological Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaohui Su
- Department of Galactophore, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenghao Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Galactophore, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruifeng Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianhui Guo
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen Gao
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiji Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Biyuan Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
36
|
Meng X, Xu H, Liang Y, Liang M, Song W, Zhou B, Shi J, Du M, Gao Y. Enhanced CT-based radiomics model to predict natural killer cell infiltration and clinical prognosis in non-small cell lung cancer. Front Immunol 2024; 14:1334886. [PMID: 38283362 PMCID: PMC10811188 DOI: 10.3389/fimmu.2023.1334886] [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: 11/08/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Background Natural killer (NK) cells are crucial for tumor prognosis; however, their role in non-small-cell lung cancer (NSCLC) remains unclear. The current detection methods for NSCLC are inefficient and costly. Therefore, radiomics represent a promising alternative. Methods We analyzed the radiogenomics datasets to extract clinical, radiological, and transcriptome data. The effect of NK cells on the prognosis of NSCLC was assessed. Tumors were delineated using a 3D Slicer, and features were extracted using pyradiomics. A radiomics model was developed and validated using five-fold cross-validation. A nomogram model was constructed using the selected clinical variables and a radiomic score (RS). The CIBERSORTx database and gene set enrichment analysis were used to explore the correlations of NK cell infiltration and molecular mechanisms. Results Higher infiltration of NK cells was correlated with better overall survival (OS) (P = 0.002). The radiomic model showed an area under the curve of 0.731, with 0.726 post-validation. The RS differed significantly between high and low infiltration of NK cells (P < 0.01). The nomogram, using RS and clinical variables, effectively predicted 3-year OS. NK cell infiltration was correlated with the ICOS and BTLA genes (P < 0.001) and macrophage M0/M2 levels. The key pathways included TNF-α signaling via NF-κB and Wnt/β-catenin signaling. Conclusions Our radiomic model accurately predicted NK cell infiltration in NSCLC. Combined with clinical characteristics, it can predict the prognosis of patients with NSCLC. Bioinformatic analysis revealed the gene expression and pathways underlying NK cell infiltration in NSCLC.
Collapse
Affiliation(s)
- Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijun Xu
- 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, China
| | - Yicheng Liang
- Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Shi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Zhou L, Sun J, Long H, Zhou W, Xia R, Luo Y, Fang J, Wang Y, Chen X. Imaging phenotyping using 18F-FDG PET/CT radiomics to predict micropapillary and solid pattern in lung adenocarcinoma. Insights Imaging 2024; 15:5. [PMID: 38185779 PMCID: PMC10772036 DOI: 10.1186/s13244-023-01573-9] [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: 09/10/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
OBJECTIVES To develop and validate a machine learning model using 18F-FDG PET/CT radiomics signature and clinical features to predict the presence of micropapillary and solid (MP/S) components in lung adenocarcinoma. METHODS Eight hundred and forty-six patients who underwent preoperative PET/CT with pathologically confirmed adenocarcinoma were enrolled. After segmentation, 1688 radiomics features were extracted from PET/CT and selected to construct predictive models. Then, we developed a nomogram based on PET/CT radiomics integrated with clinical features. Receiver operating curves, calibration curves, and decision curve analysis (DCA) were performed for diagnostics assessment and test of the developed models for distinguishing patients with MP/S components from the patients without. RESULTS PET/CT radiomics-clinical combined model could well distinguish patients with MP/S components from those without MP/S components (AUC = 0.87), which performed better than PET (AUC = 0.829, p < 0.05) or CT (AUC = 0.827, p < 0.05) radiomics models in the training cohort. In test cohorts, radiomics-clinical combined model outperformed the PET radiomics model in test cohort 1 (AUC = 0.859 vs 0.799, p < 0.05) and the CT radiomics model in test cohort 2 (AUC = 0.880 vs 0.829, p < 0.05). Calibration curve indicated good coherence between all model prediction and the actual observation in training and test cohorts. DCA revealed PET/CT radiomics-clinical model exerted the highest clinical benefit. CONCLUSION 18F-FDG PET/CT radiomics signatures could achieve promising prediction efficiency to identify the presence of MP/S components in adenocarcinoma patients to help the clinician decide on personalized treatment and surveillance strategies. The PET/CT radiomics-clinical combined model performed best. CRITICAL RELEVANCE STATEMENT: 18F-FDG PET/CT radiomics signatures could achieve promising prediction efficiency to identify the presence of micropapillary and solid components in adenocarcinoma patients to help the clinician decide on personalized treatment and surveillance strategies.
Collapse
Affiliation(s)
- Linyi Zhou
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Jinju Sun
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - He Long
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Weicheng Zhou
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Renxiang Xia
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Luo
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingqin Fang
- Department of Ultrasound, Daping Hospital, Army Medical University, Chongqing, China.
| | - Yi Wang
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China.
| |
Collapse
|
38
|
Sun J, Cong C, Li X, Zhou W, Xia R, Liu H, Wang Y, Xu Z, Chen X. Identification of Parkinson's disease and multiple system atrophy using multimodal PET/MRI radiomics. Eur Radiol 2024; 34:662-672. [PMID: 37535155 DOI: 10.1007/s00330-023-10003-9] [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: 09/28/2022] [Revised: 05/08/2023] [Accepted: 06/06/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVES To construct a machine learning model for differentiating Parkinson's disease (PD) and multiple system atrophy (MSA) by using multimodal PET/MRI radiomics and clinical characteristics. METHODS One hundred and nineteen patients (81 with PD and 38 with MSA) underwent brain PET/CT and MRI to obtain metabolic images ([18F]FDG, [11C]CFT PET) and structural MRI (T1WI, T2WI, and T2-FLAIR). Image analysis included automatic segmentation on MRI, co-registration of PET images onto the corresponding MRI. Radiomics features were then extracted from the putamina and caudate nuclei and selected to construct predictive models. Moreover, based on PET/MRI radiomics and clinical characteristics, we developed a nomogram. Receiver operating characteristic (ROC) curves were performed to evaluate the performance of the models. Decision curve analysis (DCA) was employed to access the clinical usefulness of the models. RESULTS The combined PET/MRI radiomics model of five sequences outperformed monomodal radiomics models alone. Further, PET/MRI radiomics-clinical combined model could perfectly distinguish PD from MSA (AUC = 0.993), which outperformed the clinical model (AUC = 0.923, p = 0.028) in training set, with no significant difference in test set (AUC = 0.860 vs 0.917, p = 0.390). However, no significant difference was found between PET/MRI radiomics-clinical model and PET/MRI radiomics model in training (AUC = 0.988, p = 0.276) and test sets (AUC = 0.860 vs 0.845, p = 0.632). DCA demonstrated the highest clinical benefit of PET/MRI radiomics-clinical model. CONCLUSIONS Our study indicates that multimodal PET/MRI radiomics could achieve promising performance to differentiate between PD and MSA in clinics. CLINICAL RELEVANCE STATEMENT This study developed an optimal radiomics signature and construct model to distinguish PD from MSA by multimodal PET/MRI imaging methods in clinics for parkinsonian syndromes, which achieved an excellent performance. KEY POINTS •Multimodal PET/MRI radiomics from putamina and caudate nuclei increase the diagnostic efficiency for distinguishing PD from MSA. •The radiomics-based nomogram was developed to differentiate between PD and MSA. •Combining PET/MRI radiomics-clinical model achieved promising performance to identify PD and MSA.
Collapse
Affiliation(s)
- Jinju Sun
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Chao Cong
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
- School of Electrical and Electronic Engineering, Chongqing University of Technology, Chongqing, China
| | - Xinpeng Li
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing, China
| | - Weicheng Zhou
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Renxiang Xia
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Yi Wang
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhiqiang Xu
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing, China.
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China.
| |
Collapse
|
39
|
Nakajo M, Jinguji M, Ito S, Tani A, Hirahara M, Yoshiura T. Clinical application of 18F-fluorodeoxyglucose positron emission tomography/computed tomography radiomics-based machine learning analyses in the field of oncology. Jpn J Radiol 2024; 42:28-55. [PMID: 37526865 PMCID: PMC10764437 DOI: 10.1007/s11604-023-01476-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
Machine learning (ML) analyses using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) radiomics features have been applied in the field of oncology. The current review aimed to summarize the current clinical articles about 18F-FDG PET/CT radiomics-based ML analyses to solve issues in classifying or constructing prediction models for several types of tumors. In these studies, lung and mediastinal tumors were the most commonly evaluated lesions, followed by lymphatic, abdominal, head and neck, breast, gynecological, and other types of tumors. Previous studies have commonly shown that 18F-FDG PET radiomics-based ML analysis has good performance in differentiating benign from malignant tumors, predicting tumor characteristics and stage, therapeutic response, and prognosis by examining significant differences in the area under the receiver operating characteristic curves, accuracies, or concordance indices (> 0.70). However, these studies have reported several ML algorithms. Moreover, different ML models have been applied for the same purpose. Thus, various procedures were used in 18F-FDG PET/CT radiomics-based ML analysis in oncology, and 18F-FDG PET/CT radiomics-based ML models, which are easy and universally applied in clinical practice, would be expected to be established.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Megumi Jinguji
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Soichiro Ito
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
40
|
Zhao Z, Li W, Liu P, Zhang A, Sun J, Xu LX. Survival Analysis for Multimode Ablation Using Self-Adapted Deep Learning Network Based on Multisource Features. IEEE J Biomed Health Inform 2024; 28:19-30. [PMID: 37015120 DOI: 10.1109/jbhi.2023.3260776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Novel multimode thermal therapy by freezing before radio-frequency heating has achieved a desirable therapeutic effect in liver cancer. Compared with surgical resection, ablation treatment has a relatively high risk of tumor recurrence. To monitor tumor progression after ablation, we developed a novel survival analysis framework for survival prediction and efficacy assessment. We extracted preoperative and postoperative MRI radiomics features and vision transformer-based deep learning features. We also combined the immune features extracted from peripheral blood immune responses using flow cytometry and routine blood tests before and after treatment. We selected features using random survival forest and improved the deep Cox mixture (DCM) for survival analysis. To properly accommodate multitype input features, we proposed a self-adapted fully connected layer for locally and globally representing features. We evaluated the method using our clinical dataset. Of note, the immune features rank the highest feature importance and contribute significantly to the prediction accuracy. The results showed a promising C$^{\mathit{td}}$-index of 0.885 $\pm$ 0.040 and an integrated Brier score of 0.041 $\pm$ 0.014, which outperformed state-of-the-art method combinations of survival prediction. For each patient, individual survival probability was accurately predicted over time, which provided clinicians with trustable prognosis suggestions.
Collapse
|
41
|
Tang X, Wu F, Chen X, Ye S, Ding Z. Current status and prospect of PET-related imaging radiomics in lung cancer. Front Oncol 2023; 13:1297674. [PMID: 38164195 PMCID: PMC10757959 DOI: 10.3389/fonc.2023.1297674] [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: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Lung cancer is highly aggressive, which has a high mortality rate. Major types encompass lung adenocarcinoma, lung squamous cell carcinoma, lung adenosquamous carcinoma, small cell carcinoma, and large cell carcinoma. Lung adenocarcinoma and lung squamous cell carcinoma together account for more than 80% of cases. Diverse subtypes demand distinct treatment approaches. The application of precision medicine necessitates prompt and accurate evaluation of treatment effectiveness, contributing to the improvement of treatment strategies and outcomes. Medical imaging is crucial in the diagnosis and management of lung cancer, with techniques such as fluoroscopy, computed radiography (CR), digital radiography (DR), computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET)/CT, and PET/MRI being essential tools. The surge of radiomics in recent times offers fresh promise for cancer diagnosis and treatment. In particular, PET/CT and PET/MRI radiomics, extensively studied in lung cancer research, have made advancements in diagnosing the disease, evaluating metastasis, predicting molecular subtypes, and forecasting patient prognosis. While conventional imaging methods continue to play a primary role in diagnosis and assessment, PET/CT and PET/MRI radiomics simultaneously provide detailed morphological and functional information. This has significant clinical potential value, offering advantages for lung cancer diagnosis and treatment. Hence, this manuscript provides a review of the latest developments in PET-related radiomics for lung cancer.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, Hangzhou Wuyunshan Hospital (Hangzhou Health Promotion Research Institute), Hangzhou, China
| | - Fan Wu
- Department of Nuclear Medicine and Radiology, Shulan Hangzhou Hospital affiliated to Shulan International Medical College of Zhejiang Shuren University, Hangzhou, China
| | - Xiaofen Chen
- Department of Radiology, Hangzhou Wuyunshan Hospital (Hangzhou Health Promotion Research Institute), Hangzhou, China
| | - Shengli Ye
- Department of Nuclear Medicine and Radiology, Shulan Hangzhou Hospital affiliated to Shulan International Medical College of Zhejiang Shuren University, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Hangzhou First People’s Hospital, Hangzhou, China
| |
Collapse
|
42
|
Zschaeck S, Klinger B, van den Hoff J, Cegla P, Apostolova I, Kreissl MC, Cholewiński W, Kukuk E, Strobel H, Amthauer H, Blüthgen N, Zips D, Hofheinz F. Combination of tumor asphericity and an extracellular matrix-related prognostic gene signature in non-small cell lung cancer patients. Sci Rep 2023; 13:20840. [PMID: 38012155 PMCID: PMC10681996 DOI: 10.1038/s41598-023-46405-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
One important aim of precision oncology is a personalized treatment of patients. This can be achieved by various biomarkers, especially imaging parameters and gene expression signatures are commonly used. So far, combination approaches are sparse. The aim of the study was to independently validate the prognostic value of the novel positron emission tomography (PET) parameter tumor asphericity (ASP) in non small cell lung cancer (NSCLC) patients and to investigate associations between published gene expression profiles and ASP. This was a retrospective evaluation of PET imaging and gene expression data from three public databases and two institutional datasets. The whole cohort comprised 253 NSCLC patients, all treated with curative intent surgery. Clinical parameters, standard PET parameters and ASP were evaluated in all patients. Additional gene expression data were available for 120 patients. Univariate Cox regression and Kaplan-Meier analysis was performed for the primary endpoint progression-free survival (PFS) and additional endpoints. Furthermore, multivariate cox regression testing was performed including clinically significant parameters, ASP, and the extracellular matrix-related prognostic gene signature (EPPI). In the whole cohort, a significant association with PFS was observed for ASP (p < 0.001) and EPPI (p = 0.012). Upon multivariate testing, EPPI remained significantly associated with PFS (p = 0.018) in the subgroup of patients with additional gene expression data, while ASP was significantly associated with PFS in the whole cohort (p = 0.012). In stage II patients, ASP was significantly associated with PFS (p = 0.009), and a previously published cutoff value for ASP (19.5%) was successfully validated (p = 0.008). In patients with additional gene expression data, EPPI showed a significant association with PFS, too (p = 0.033). The exploratory combination of ASP and EPPI showed that the combinatory approach has potential to further improve patient stratification compared to the use of only one parameter. We report the first successful validation of EPPI and ASP in stage II NSCLC patients. The combination of both parameters seems to be a very promising approach for improvement of risk stratification in a group of patients with urgent need for a more personalized treatment approach.
Collapse
Affiliation(s)
- Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Bertram Klinger
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
- Computational Modelling in Medicine, Instiute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Jörg van den Hoff
- Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Paulina Cegla
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Ivayla Apostolova
- Department for Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Division of Nuclear Medicine, Department of Radiology and Nuclear Medicine, Otto Von Guericke University, Magdeburg, Germany
| | - Michael C Kreissl
- Division of Nuclear Medicine, Department of Radiology and Nuclear Medicine, Otto Von Guericke University, Magdeburg, Germany
| | - Witold Cholewiński
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Emily Kukuk
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helen Strobel
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Holger Amthauer
- Division of Nuclear Medicine, Department of Radiology and Nuclear Medicine, Otto Von Guericke University, Magdeburg, Germany
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Nils Blüthgen
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
- Computational Modelling in Medicine, Instiute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Frank Hofheinz
- Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.
| |
Collapse
|
43
|
Nguyen TM, Bertolus C, Giraud P, Burgun A, Saintigny P, Bibault JE, Foy JP. A Radiomics Approach to Identify Immunologically Active Tumor in Patients with Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2023; 15:5369. [PMID: 38001629 PMCID: PMC10670096 DOI: 10.3390/cancers15225369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND We recently developed a gene-expression-based HOT score to identify the hot/cold phenotype of head and neck squamous cell carcinomas (HNSCCs), which is associated with the response to immunotherapy. Our goal was to determine whether radiomic profiling from computed tomography (CT) scans can distinguish hot and cold HNSCC. METHOD We included 113 patients from The Cancer Genome Atlas (TCGA) and 20 patients from the Groupe Hospitalier Pitié-Salpêtrière (GHPS) with HNSCC, all with available pre-treatment CT scans. The hot/cold phenotype was computed for all patients using the HOT score. The IBEX software (version 4.11.9, accessed on 30 march 2020) was used to extract radiomic features from the delineated tumor region in both datasets, and the intraclass correlation coefficient (ICC) was computed to select robust features. Machine learning classifier models were trained and tested in the TCGA dataset and validated using the area under the receiver operator characteristic curve (AUC) in the GHPS cohort. RESULTS A total of 144 radiomic features with an ICC >0.9 was selected. An XGBoost model including these selected features showed the best performance prediction of the hot/cold phenotype with AUC = 0.86 in the GHPS validation dataset. CONCLUSIONS AND RELEVANCE We identified a relevant radiomic model to capture the overall hot/cold phenotype of HNSCC. This non-invasive approach could help with the identification of patients with HNSCC who may benefit from immunotherapy.
Collapse
Affiliation(s)
- Tan Mai Nguyen
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Chloé Bertolus
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
| | - Paul Giraud
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Sorbonne Université, Department of Radiation Oncology, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France
| | - Anita Burgun
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Jean-Emmanuel Bibault
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Department of Radiation Oncology, Hôpital Européen Georges-Pompidou, Université Paris Cité, 75015 Paris, France
| | - Jean-Philippe Foy
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Sorbonne Université, INSERM UMRS 938, Centre de Recherche de Saint Antoine, Team Cancer Biology and Therapeutics, 75011 Paris, France
| |
Collapse
|
44
|
Sang J, Ye X. Potential biomarkers for predicting immune response and outcomes in lung cancer patients undergoing thermal ablation. Front Immunol 2023; 14:1268331. [PMID: 38022658 PMCID: PMC10646301 DOI: 10.3389/fimmu.2023.1268331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Thermal ablation is a promising alternative treatment for lung cancer. It disintegrates cancer cells and releases antigens, followed by the remodeling of local tumor immune microenvironment and the activation of anti-tumor immune responses, enhancing the overall effectiveness of the treatment. Biomarkers can offer insights into the patient's immune response and outcomes, such as local tumor control, recurrence, overall survival, and progression-free survival. Identifying and validating such biomarkers can significantly impact clinical decision-making, leading to personalized treatment strategies and improved patient outcomes. This review provides a comprehensive overview of the current state of research on potential biomarkers for predicting immune response and outcomes in lung cancer patients undergoing thermal ablation, including their potential role in lung cancer management, and the challenges and future directions.
Collapse
Affiliation(s)
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| |
Collapse
|
45
|
Deininger K, Raacke JN, Yousefzadeh-Nowshahr E, Kropf-Sanchen C, Muehling B, Beer M, Glatting G, Beer AJ, Thaiss W. Combined morphologic-metabolic biomarkers from [18F]FDG-PET/CT stratify prognostic groups in low-risk NSCLC. Nuklearmedizin 2023; 62:284-292. [PMID: 37696296 DOI: 10.1055/a-2150-4130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
AIM The aim of this study was to derive prognostic parameters from 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG-PET/CT) in patients with low-risk NSCLC and determine their prognostic value. METHODS 81 (21 female, mean age 66 a) therapy-naive patients that underwent [18F]FDG-PET/CT before histologic confirmation of NSCLC with stadium I and II between 2008-2016 were included. A mean follow-up time of 58 months (13-176), overall and progression free survival (OS, PFS) were registered. A volume of interest for the primary tumor was defined on PET and CT images. Parameters SUVmax, PET-solidity, PET-circularity, and CT-volume were analyzed. To evaluate the prognostic value of each parameter for OS, a minimum p-value approach was used to define cutoff values, survival analysis, and log-rank tests were performed, including subgroup analysis for combinations of parameters. RESULTS Mean OS was 58±28 months. Poor OS was associated with a tumor CT-volume >14.3 cm3 (p=0.02, HR=7.0, CI 2.7-17.7), higher SUVmax values >12.2 (p=0.003; HR=3.0, CI 1.3-6.7) and PET-solidity >0.919 (p=0.004; HR=3.0, CI 1.0-8.9). Combined parameter analysis revealed worse prognosis in larger volume/high SUVmax tumors compared to larger volume/lower SUVmax (p=0.028; HR=2.5, CI 1.1-5.5), high PET-solidity/low volume (p=0.01; HR=2.4, CI 0.8-6.6) and low SUVmax/high PET-solidity (p=0.02, HR=4.0, CI 0.8-19.0). CONCLUSION Even in this group of low-risk NSCLC patients, we identified a subgroup with a significantly worse prognosis by combining morphologic-metabolic biomarkers from [18F]FDG-PET/CT. The combination of SUVmax and CT-volume performed best. Based on these preliminary data, future prospective studies to validate this combined morphologic-metabolic imaging biomarker for potential therapeutic decisions seem promising.
Collapse
Affiliation(s)
| | - Joel Niclas Raacke
- Nuclear Medicine, Ulm University Hospital, Ulm, Germany
- Urology, Clinical Centre St. Elisabethen, Ravensburg, Germany
| | | | | | - Bernd Muehling
- Cardiac and Thoracic Surgery, Section Thoracic and Vascular Surgery, Ulm University Hospital, Ulm, Germany
| | - Meinrad Beer
- Diagnostic and Interventional Radiology, Ulm University Hospital, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Gerhard Glatting
- Nuclear Medicine Medical Radiation Physics, Ulm University, Ulm, Germany
| | - Ambros J Beer
- Nuclear Medicine, Ulm University Hospital, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Wolfgang Thaiss
- Nuclear Medicine, Ulm University Hospital, Ulm, Germany
- Diagnostic and Interventional Radiology, Ulm University Hospital, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| |
Collapse
|
46
|
Kang W, Qiu X, Luo Y, Luo J, Liu Y, Xi J, Li X, Yang Z. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J Transl Med 2023; 21:598. [PMID: 37674169 PMCID: PMC10481579 DOI: 10.1186/s12967-023-04437-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
The advent of immunotherapy, a groundbreaking advancement in cancer treatment, has given rise to the prominence of the tumor microenvironment (TME) as a critical area of research. The clinical implications of an improved understanding of the TME are significant and far-reaching. Radiomics has been increasingly utilized in the comprehensive assessment of the TME and cancer prognosis. Similarly, the advancement of pathomics, which is based on pathological images, can offer additional insights into the panoramic view and microscopic information of tumors. The combination of pathomics and radiomics has revolutionized the concept of a "digital biopsy". As genomics and transcriptomics continue to evolve, integrating radiomics with genomic and transcriptomic datasets can offer further insights into tumor and microenvironment heterogeneity and establish correlations with biological significance. Therefore, the synergistic analysis of digital image features (radiomics, pathomics) and genetic phenotypes (genomics) can comprehensively decode and characterize the heterogeneity of the TME as well as predict cancer prognosis. This review presents a comprehensive summary of the research on important radiomics biomarkers for predicting the TME, emphasizing the interplay between radiomics, genomics, transcriptomics, and pathomics, as well as the application of multiomics in decoding the TME and predicting cancer prognosis. Finally, we discuss the challenges and opportunities in multiomics research. In conclusion, this review highlights the crucial role of radiomics and multiomics associations in the assessment of the TME and cancer prognosis. The combined analysis of radiomics, pathomics, genomics, and transcriptomics is a promising research direction with substantial research significance and value for comprehensive TME evaluation and cancer prognosis assessment.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital of, Fudan University, Shanghai, 200011, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Jianwei Luo
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yang Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junqing Xi
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
47
|
Gao J, Zhang C, Wei Z, Ye X. Immunotherapy for early-stage non-small cell lung cancer: A system review. J Cancer Res Ther 2023; 19:849-865. [PMID: 37675709 DOI: 10.4103/jcrt.jcrt_723_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/06/2023] [Indexed: 09/08/2023]
Abstract
With the addition of immunotherapy, lung cancer, one of the most common cancers with high mortality rates, has broadened the treatment landscape. Immune checkpoint inhibitors have demonstrated significant efficacy in the treatment of non-small cell lung cancer (NSCLC) and are now used as the first-line therapy for metastatic disease, consolidation therapy after radiotherapy for unresectable locally advanced disease, and adjuvant therapy after surgical resection and chemotherapy for resectable disease. The use of adjuvant and neoadjuvant immunotherapy in patients with early-stage NSCLC, however, is still debatable. We will address several aspects, namely the initial efficacy of monotherapy, the efficacy of combination chemotherapy, immunotherapy-related biomarkers, adverse effects, ongoing randomized controlled trials, and current issues and future directions for immunotherapy in early-stage NSCLC will be discussed here.
Collapse
Affiliation(s)
- Jingyi Gao
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong; Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| | - Chao Zhang
- Department of Oncology, Affiliated Qujing Hospital of Kunming Medical University, QuJing, Yunnan Province, China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong Province, China
| |
Collapse
|
48
|
Evangelista L, Fiz F, Laudicella R, Bianconi F, Castello A, Guglielmo P, Liberini V, Manco L, Frantellizzi V, Giordano A, Urso L, Panareo S, Palumbo B, Filippi L. PET Radiomics and Response to Immunotherapy in Lung Cancer: A Systematic Review of the Literature. Cancers (Basel) 2023; 15:3258. [PMID: 37370869 PMCID: PMC10296704 DOI: 10.3390/cancers15123258] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this review is to provide a comprehensive overview of the existing literature concerning the applications of positron emission tomography (PET) radiomics in lung cancer patient candidates or those undergoing immunotherapy. MATERIALS AND METHODS A systematic review was conducted on databases and web sources. English-language original articles were considered. The title and abstract were independently reviewed to evaluate study inclusion. Duplicate, out-of-topic, and review papers, or editorials, articles, and letters to editors were excluded. For each study, the radiomics analysis was assessed based on the radiomics quality score (RQS 2.0). The review was registered on the PROSPERO database with the number CRD42023402302. RESULTS Fifteen papers were included, thirteen were qualified as using conventional radiomics approaches, and two used deep learning radiomics. The content of each study was different; indeed, seven papers investigated the potential ability of radiomics to predict PD-L1 expression and tumor microenvironment before starting immunotherapy. Moreover, two evaluated the prediction of response, and four investigated the utility of radiomics to predict the response to immunotherapy. Finally, two papers investigated the prediction of adverse events due to immunotherapy. CONCLUSIONS Radiomics is promising for the evaluation of TME and for the prediction of response to immunotherapy, but some limitations should be overcome.
Collapse
Affiliation(s)
- Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Francesco Fiz
- Nuclear Medicine Department, E.O. “Ospedali Galliera”, 16128 Genoa, Italy;
- Nuclear Medicine Department and Clinical Molecular Imaging, University Hospital, 72076 Tübingen, Germany
| | - Riccardo Laudicella
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90100 Palermo, Italy;
| | - Francesco Bianconi
- Department of Engineering, Università degli Studi di Perugia, Via Goffredo Duranti, 06125 Perugia, Italy;
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Priscilla Guglielmo
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy;
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy;
| | - Luigi Manco
- Medical Physics Unit, Azienda USL of Ferrara, 45100 Ferrara, Italy;
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy;
| | - Alessia Giordano
- Nuclear Medicine Unit, IRCCS CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Luca Urso
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, 41124 Modena, Italy;
| | - Barbara Palumbo
- Section of Nuclear Medicine and Health Physics, Department of Medicine and Surgery, Università degli Studi di Perugia, 06125 Perugia, Italy;
| | - Luca Filippi
- Nuclear Medicine Section, Santa Maria Goretti Hospital, 04100 Latina, Italy;
| |
Collapse
|
49
|
Gao Q, Yang L, Lu M, Jin R, Ye H, Ma T. The artificial intelligence and machine learning in lung cancer immunotherapy. J Hematol Oncol 2023; 16:55. [PMID: 37226190 DOI: 10.1186/s13045-023-01456-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Since the past decades, more lung cancer patients have been experiencing lasting benefits from immunotherapy. It is imperative to accurately and intelligently select appropriate patients for immunotherapy or predict the immunotherapy efficacy. In recent years, machine learning (ML)-based artificial intelligence (AI) was developed in the area of medical-industrial convergence. AI can help model and predict medical information. A growing number of studies have combined radiology, pathology, genomics, proteomics data in order to predict the expression levels of programmed death-ligand 1 (PD-L1), tumor mutation burden (TMB) and tumor microenvironment (TME) in cancer patients or predict the likelihood of immunotherapy benefits and side effects. Finally, with the advancement of AI and ML, it is believed that "digital biopsy" can replace the traditional single assessment method to benefit more cancer patients and help clinical decision-making in the future. In this review, the applications of AI in PD-L1/TMB prediction, TME prediction and lung cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Luyu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Renjing Jin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Huan Ye
- Department of Respiratory and Critical Care Medicine, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
50
|
Dai J, Wang H, Xu Y, Chen X, Tian R. Clinical application of AI-based PET images in oncological patients. Semin Cancer Biol 2023; 91:124-142. [PMID: 36906112 DOI: 10.1016/j.semcancer.2023.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Based on the advantages of revealing the functional status and molecular expression of tumor cells, positron emission tomography (PET) imaging has been performed in numerous types of malignant diseases for diagnosis and monitoring. However, insufficient image quality, the lack of a convincing evaluation tool and intra- and interobserver variation in human work are well-known limitations of nuclear medicine imaging and restrict its clinical application. Artificial intelligence (AI) has gained increasing interest in the field of medical imaging due to its powerful information collection and interpretation ability. The combination of AI and PET imaging potentially provides great assistance to physicians managing patients. Radiomics, an important branch of AI applied in medical imaging, can extract hundreds of abstract mathematical features of images for further analysis. In this review, an overview of the applications of AI in PET imaging is provided, focusing on image enhancement, tumor detection, response and prognosis prediction and correlation analyses with pathology or specific gene mutations in several types of tumors. Our aim is to describe recent clinical applications of AI-based PET imaging in malignant diseases and to focus on the description of possible future developments.
Collapse
Affiliation(s)
- Jiaona Dai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang City 421001, China
| | - Xiyang Chen
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|