1
|
Loaiza-Bonilla A, Thaker N, Chung C, Parikh RB, Stapleton S, Borkowski P. Driving Knowledge to Action: Building a Better Future With Artificial Intelligence-Enabled Multidisciplinary Oncology. Am Soc Clin Oncol Educ Book 2025; 45:e100048. [PMID: 40315375 DOI: 10.1200/edbk-25-100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Artificial intelligence (AI) is transforming multidisciplinary oncology at an unprecedented pace, redefining how clinicians detect, classify, and treat cancer. From earlier and more accurate diagnoses to personalized treatment planning, AI's impact is evident across radiology, pathology, radiation oncology, and medical oncology. By leveraging vast and diverse data-including imaging, genomic, clinical, and real-world evidence-AI algorithms can uncover complex patterns, accelerate drug discovery, and help identify optimal treatment regimens for each patient. However, realizing the full potential of AI also necessitates addressing concerns regarding data quality, algorithmic bias, explainability, privacy, and regulatory oversight-especially in low- and middle-income countries (LMICs), where disparities in cancer care are particularly pronounced. This study provides a comprehensive overview of how AI is reshaping cancer care, reviews its benefits and challenges, and outlines ethical and policy implications in line with ASCO's 2025 theme, Driving Knowledge to Action. We offer concrete calls to action for clinicians, researchers, industry stakeholders, and policymakers to ensure that AI-driven, patient-centric oncology is accessible, equitable, and sustainable worldwide.
Collapse
Affiliation(s)
- Arturo Loaiza-Bonilla
- St Luke's University Health Network, Bethlehem, PA
- Massive Bio, Inc, New York, NY
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | - Caroline Chung
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Shawn Stapleton
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
2
|
Yeghaian M, Bodalal Z, van den Broek D, Haanen JBAG, Beets-Tan RGH, Trebeschi S, van Gerven MAJ. Multimodal integration of longitudinal noninvasive diagnostics for survival prediction in immunotherapy using deep learning. J Am Med Inform Assoc 2025:ocaf074. [PMID: 40418276 DOI: 10.1093/jamia/ocaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/17/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVES Immunotherapies have revolutionized the landscape of cancer treatments. However, our understanding of response patterns in advanced cancers treated with immunotherapy remains limited. By leveraging routinely collected noninvasive longitudinal and multimodal data with artificial intelligence, we could unlock the potential to transform immunotherapy for cancer patients, paving the way for personalized treatment approaches. MATERIALS AND METHODS In this study, we developed a novel artificial neural network architecture, multimodal transformer-based simple temporal attention (MMTSimTA) network, building upon a combination of recent successful developments. We integrated pre- and on-treatment blood measurements, prescribed medications, and CT-based volumes of organs from a large pan-cancer cohort of 694 patients treated with immunotherapy to predict mortality at 3, 6, 9, and 12 months. Different variants of our extended MMTSimTA network were implemented and compared to baseline methods, incorporating intermediate and late fusion-based integration methods. RESULTS The strongest prognostic performance was demonstrated using a variant of the MMTSimTA model with area under the curves of 0.84 ± 0.04, 0.83 ± 0.02, 0.82 ± 0.02, 0.81 ± 0.03 for 3-, 6-, 9-, and 12-month survival prediction, respectively. DISCUSSION Our findings show that integrating noninvasive longitudinal data using our novel architecture yields an improved multimodal prognostic performance, especially in short-term survival prediction. CONCLUSION Our study demonstrates that multimodal longitudinal integration of noninvasive data using deep learning may offer a promising approach for personalized prognostication in immunotherapy-treated cancer patients.
Collapse
Affiliation(s)
- Melda Yeghaian
- Department of Machine Learning and Neural Computing, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 GD, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Zuhir Bodalal
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- Division of Molecular Oncology and Immunology, Oncode Institute, Amsterdam 1066 CX, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden 2333 ZG, The Netherlands
- Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne 1005, Switzerland
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
- Faculty of Health Science, University of Southern Denmark, Odense 5230, Denmark
| | - Stefano Trebeschi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Marcel A J van Gerven
- Department of Machine Learning and Neural Computing, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 GD, The Netherlands
| |
Collapse
|
3
|
Zhang P, Wei L, Nie Z, Hu P, Zheng J, Lv J, Cui T, Liu C, Lan X. Research on the developments of artificial intelligence in radiomics for oncology over the past decade: a bibliometric and visualized analysis. Discov Oncol 2025; 16:763. [PMID: 40366503 PMCID: PMC12078899 DOI: 10.1007/s12672-025-02590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE To assess the publications' bibliographic features and look into how the advancement of artificial intelligence (AI) and its subfields in radiomics has affected the growth of oncology. METHODS The researchers conducted a search in the Web of Science (WoS) for scientific publications in cancer pertaining to AI and radiomics, published in English from 1 January 2015 to 31 December 2024.The research included a scientometric methodology and comprehensive data analysis utilising scientific visualization tools, including the Bibliometrix R software package, VOSviewer, and CiteSpace. Bibliometric techniques utilised were co-authorship, co-citation, co-occurrence, citation burst, and performance Analysis. RESULTS The final study encompassed 4,127 publications authored by 5,026 individuals and published across 597 journals. China (2087;50.57%) and USA (850;20.6%) were the two most productive countries. The authors with the highest publication counts were Tian Jie (60) and Cuocolo Renato (30). Fudan University (169;4.09%) and Sun Yat-sen University (162;3.93%) were the most active institutions. The foremost journals were Frontiers in Oncology and Cancer. The predominant author keywords were radiomics, artificial intelligence, and oncology research. CONCLUSION Investigations into the integration of AI with radiomics in oncology remain nascent, with numerous studies concentrating on biology, diagnosis, treatment, and cancer risk evaluation.
Collapse
Affiliation(s)
- Pengyu Zhang
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China
- School of Qingdao Medical College, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Lili Wei
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China
| | - Zonglong Nie
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China
| | - Pengcheng Hu
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China
| | - Jilu Zheng
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China
| | - Ji Lv
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China.
| | - Tao Cui
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China.
| | - Chunlei Liu
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China.
| | - Xiaopeng Lan
- Department of Urology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, People's Republic of China.
| |
Collapse
|
4
|
Wu Y, Wang H, Gu Y, Zhang Y, Li G, Huang Y, Cao M, Chen X, Guan Y, Xu D, Wei W, Xie F. Head-to-head comparison of peptide-based and nanobody-based radiotracers in detecting PD-L1 expression in non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07316-w. [PMID: 40338301 DOI: 10.1007/s00259-025-07316-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Immunotherapy based on programmed cell death protein receptor 1 and its ligand (PD-1/PD-L1) has become an important method for treating non-small cell lung cancer (NSCLC). Peptide-based and nanobody-based PET tracers offer potential advantages in PD-L1 detection, yet their comparative tumor uptake and biodistribution remain unclear. This study aimed to evaluate and compare [68Ga]Ga-DOTA-WL12 (a peptide-based tracer) and [68Ga]Ga-NOTA-RW102 (a nanobody-based tracer) in assessing PD-L1 expression in primary and metastatic NSCLC, providing insights for future radiotracer design and theranostic applications. METHODS Ten patients diagnosed with NSCLC underwent [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 PET/CT scans, with four of these patients also receiving [18F]FDG PET/CT scans. The tracer uptakes, quantified by maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), and target-to-background ratio (TBR), were compared between [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 PET/CT. RESULTS DOTA-WL12 and NOTA-RW102 exhibited favorable binding affinities with PD-L1, with equilibrium dissociation constant (KD) values of 0.2 nM and 0.0047 nM, respectively. Subsequent human studies revealed significant variations (P < 0.05) in the uptake of [68Ga]Ga-DOTA-WL12 and [68Ga]Ga-NOTA-RW102 across the liver (SUVmean: 20.43 ± 4.26 vs. 6.12 ± 1.36, p = 0.015), kidney (SUVmean: 2.40 ± 0.34 vs. 22.37 ± 2.88, P = 0.015), spleen (SUVmean: 2.44 ± 0.67 vs. 18.49 ± 3.90, P = 0.015), and lung background (SUVmean: 0.18 ± 0.12 vs. 1.09 ± 0.29, P = 0.015). Meanwhile, we found that the correlation between SUVmax and PD-L1 TPS was significantly stronger with [68Ga]Ga-DOTA-WL12 compared to [68Ga]Ga-NOTA-RW102 (P < 0.0001, r = 0.9471 vs. P = 0.0241, r = 0.5235). CONCLUSION The uptake of peptide-based [68Ga]Ga-DOTA-WL12 was more strongly correlated with PD-L1 TPS in primary and metastatic tumor lesions compared to [68Ga]Ga-NOTA-RW102. They also displayed different distribution, suggesting that peptide-based and nanobody-based radiotracers may have different clinical implications, particularly in radiotherapy.
Collapse
Affiliation(s)
- Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yue Gu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Guanglei Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuan Huang
- Rehabilitation Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Cao
- Department of Thoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaofeng Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Wu Y, Zhang W, Liang X, Zhang P, Zhang M, Jiang Y, Cui Y, Chen Y, Zhou W, Liang Q, Dai J, Zhang C, Xu J, Li J, Yu T, Zhang Z, Guo R. Habitat radiomics analysis for progression free survival and immune-related adverse reaction prediction in non-small cell lung cancer treated by immunotherapy. J Transl Med 2025; 23:393. [PMID: 40181378 PMCID: PMC11970015 DOI: 10.1186/s12967-024-06057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/25/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is highly heterogeneous, leading to varied treatment responses and immune-related adverse reactions (irAEs) among patients. Habitat radiomics allows non-invasive quantitative assessment of intratumor heterogeneity (ITH). Therefore, our objective is to employ habitat radiomics techniques to develop a robust approach for predicting the efficacy of Immune checkpoint inhibitors (ICIs) and the likelihood of irAEs in advanced NSCLC patients. METHODS In this retrospective two center study, two independent cohorts of patients with NSCLC were used to develop (n = 248) and validate signatures (n = 95). After applying four kinds of machine learning algorithms to select the key preoperative CT radiomic features, we used clinical, radiomics and habitat radiomic features to develop the clinical signature, radiomics signature and habitat radiomic signature for ICIs prognostics and irAEs prediction. By combining habitat radiomic features with corresponding clinicopathologic information, the nomogram signature was constructed in the training cohort. Next, the internal validation cohort (n = 75) of patients, and the external validation cohort (n = 20) of patients treated with ICIs were included to evaluate the predictive value of the four signatures, and their predictive performance was assessed by the area under operating characteristic curve (AUC). RESULTS Our study introduces a radiomic nomogram model that integrates clinical and habitat radiomic features to identify patients who may benefit from ICIs or experience irAEs. The Radiomics Nomogram model exhibited superior predictive performance in the training, validation, and external validation sets, with AUCs of 0.923, 0.817, and 0.899, respectively. This model outperformed both the Whole-tumor Radiomics Signature model (AUCs of 0.870, 0.736, and 0.626) and the Habitat Signature model (AUCs of 0.900, 0.804, and 0.808). The radiomics model focusing on tumor sub-regional habitat showed better predictive performance than the model derived from the entire tumor. Decision Curve Analysis (DCA) and calibration curves confirmed the nomogram's effectiveness. CONCLUSION By leveraging machine learning to predict the outcomes of ICIs, we can move closer to achieving tailored ICIs for lung cancer. This advancement will assist physicians in selecting and managing subsequent treatment strategies, thereby facilitating clinical decision-making.
Collapse
Affiliation(s)
- Yuemin Wu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Institute, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuqin Jiang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yanan Cui
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Chen
- Department of Oncology, Pukou Branch of Jiangsu People's Hospital, Nanjing Pukou District Central Hospital, Nanjing, China
| | - Wenxin Zhou
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qi Liang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Dai
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chen Zhang
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiali Xu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tongfu Yu
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhihong Zhang
- Department of Pathology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Renhua Guo
- Department of Radiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Yu X, Yang D, Xu G, Tian F, Shi H, Xie Z, Cao Z, Wang J. A model for prediction of recurrence of non-small cell lung cancer based on clinical data and CT imaging characteristics. Clin Imaging 2025; 120:110416. [PMID: 39904004 DOI: 10.1016/j.clinimag.2025.110416] [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: 09/10/2024] [Revised: 01/16/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVES To establish a model for prediction of recurrence of non-small cell lung cancer (NSCLC) based on clinical data and computed tomography (CT) imaging characteristics. METHODS A total of 695 patients with surgically resected NSCLC confirmed by pathology at three centers were retrospectively investigated. 626 patients from center 1 were randomly divided into two sets in a ratio of 7:3 (training set, n = 438; testing set, n = 188), 69 patients from center 2 and 3 were assigned in the external validation set. Univariate and binary logistic regression analyses of clinical and CT imaging features determined the independent risk factors used to construct the model. The receiver-operating characteristic curve nomogram and decision curves analysis were used to evaluate the predictive ability of the model. RESULTS The mean patient age was 63.3 ± 10.1 years, and 44.7 % (311/695) were male. The univariate and binary logistic regression analyses identified four independent risk factors (age, tumor markers, consolidation/tumor ratio, and pleural effusion), which were used to construct the prediction model. In the training set, the model had an area under the curve of 0.857, an accuracy of 71.7 %, a sensitivity of 88.1 %, and a specificity of 70.0 %; in the testing set, the respective values were 0.867, 75.5 %, 94.4 %, and 73.5 %; in the external validation set, the respective values were 0.852, 79.7 %, 83.3 %, 78.9 %. CONCLUSION A prediction model based on clinical data and CT imaging characteristics showed excellent efficiency in prediction of recurrence of NSCLC. Clinical use of this model could be useful for selection of appropriate treatment options.
Collapse
Affiliation(s)
- Xinjie Yu
- Department of Radiology, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China
| | - Dengfa Yang
- Department of Radiology, Taizhou Municipal Hospital, Taizhou, Zhejiang Province, China
| | - Gang Xu
- Department of Radiology, Xin Hua Hospital of Huainan, Huainan, Anhui Province, China
| | - Fengjuan Tian
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hengfeng Shi
- Department of Radiology, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhenyu Cao
- Department of Radiology, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China
| | - Jian Wang
- Department of Radiology, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China.
| |
Collapse
|
7
|
Li S, Hu Y, Tian C, Luan J, Zhang X, Wei Q, Li X, Bian Y. Prediction of EGFR-TP53 genes co-mutations in patients with lung adenocarcinoma (LUAD) by 18F-FDG PET/CT radiomics. Clin Transl Oncol 2025; 27:1506-1515. [PMID: 39251494 DOI: 10.1007/s12094-024-03685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE This retrospective study was undertaken to assess the predictive efficacy of 18F-FDG PET/CT -derived radiomic features concerning the co-mutation status of epidermal growth factor receptor (EGFR) and TP53 in LUAD. METHODS A cohort of 150 LUAD patients underwent pretreatment 18F-FDG PET/CT scans with known mutation status of EGFR and TP53 were collected. The feature extraction based on their PET/CT images utilized the Pyradiomics package based on the 3D Slicer. The optimal radiomic features were selected through correlation analysis and the Gradient Boosting Decision Tree (GBDT) algorithm, followed by the construction of the radiomic model. The clinical model incorporated meaningful clinical variables, whereas the complex model integrated both the radiomic and clinical models. The area under the receiver operating characteristic curve (AUC) facilitated the comparison of prediction performance across the three models. The DCA gauged the clinical utility of these models. RESULTS The patient cohort was randomly allocated into a training set (n = 105) and a validation set (n = 45) in a 7:3 ratio. Eleven PET and eleven CT optimal radiomic features were selected to construct the radiomic model. The model showed a good ability to discriminate the co-occurrence of EGFR and TP53, with AUC equal to 0.850 in the training set, and 0.748 in the validation set, compared with 0.750 and 0.626 for the clinical model. The complex model exhibited the highest AUC values, with 0.880 and 0.794 in both sets, but there were no significant differences compared to the radiomic model. The DCA revealed favorable clinical value. CONCLUSION
Collapse
Affiliation(s)
- Shuheng Li
- Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Congna Tian
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiusong Luan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanzhu Bian
- Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
8
|
Lin S, Ma Z, Yao Y, Huang H, Chen W, Tang D, Gao W. Automatic machine learning accurately predicts the efficacy of immunotherapy for patients with inoperable advanced non-small cell lung cancer using a computed tomography-based radiomics model. Diagn Interv Radiol 2025; 31:130-140. [PMID: 39817633 PMCID: PMC11880869 DOI: 10.4274/dir.2024.242972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE Patients with advanced non-small cell lung cancer (NSCLC) have varying responses to immunotherapy, but there are no reliable, accepted biomarkers to accurately predict its therapeutic efficacy. The present study aimed to construct individualized models through automatic machine learning (autoML) to predict the efficacy of immunotherapy in patients with inoperable advanced NSCLC. METHODS A total of 63 eligible participants were included and randomized into training and validation groups. Radiomics features were extracted from the volumes of interest of the tumor circled in the preprocessed computed tomography (CT) images. Golden feature, clinical, radiomics, and fusion models were generated using a combination of various algorithms through autoML. The models were evaluated using a multi-class receiver operating characteristic curve. RESULTS In total, 1,219 radiomics features were extracted from regions of interest. The ensemble algorithm demonstrated superior performance in model construction. In the training cohort, the fusion model exhibited the highest accuracy at 0.84, with an area under the curve (AUC) of 0.89-0.98. In the validation cohort, the radiomics model had the highest accuracy at 0.89, with an AUC of 0.98-1.00; its prediction performance in the partial response subgroup outperformed that in both the clinical and radiomics models. Patients with low rad scores achieved improved progression-free survival (PFS); (median PFS 16.2 vs. 13.4, P = 0.009). CONCLUSION autoML accurately and robustly predicted the short-term outcomes of patients with inoperable NSCLC treated with immune checkpoint inhibitor immunotherapy by constructing CT-based radiomics models, confirming it as a powerful tool to assist in the individualized management of patients with advanced NSCLC. CLINICAL SIGNIFICANCE This article highlights that autoML promotes the accuracy and efficiency of feature selection and model construction. The radiomics model generated by autoML predicted the efficacy of immunotherapy in patients with advanced NSCLC effectively. This may provide a rapid and non-invasive method for making personalized clinical decisions.
Collapse
Affiliation(s)
- Siyun Lin
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Zhuangxuan Ma
- Huadong Hospital, Fudan University, Department of Radiology, Shanghai, China
| | - Yuanshan Yao
- Shanghai Chest Hospital, Shanghai JiaoTong University School of Medicine, Department of Thoracic Surgery, Shanghai, China
| | - Hou Huang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Wufei Chen
- Huadong Hospital, Fudan University, Department of Radiology, Shanghai, China
| | - Dongfang Tang
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
| | - Wen Gao
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
| |
Collapse
|
9
|
Ligero M, El Nahhas OSM, Aldea M, Kather JN. Artificial intelligence-based biomarkers for treatment decisions in oncology. Trends Cancer 2025; 11:232-244. [PMID: 39814650 DOI: 10.1016/j.trecan.2024.12.001] [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: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The development of new therapeutic strategies such as immune checkpoint inhibitors (ICIs) and targeted therapies has increased the complexity of the treatment landscape for solid tumors. At the current rate of annual FDA approvals, the potential treatment options could increase by tenfold over the next 5 years. The cost of personalized medicine technologies limits its accessibility, thus increasing socioeconomic disparities in the treated population. In this review we describe artificial intelligence (AI)-based solutions - including deep learning (DL) methods for routine medical imaging and large language models (LLMs) for electronic health records (EHRs) - to support cancer treatment decisions with cost-effective biomarkers. We address the current limitations of these technologies and propose the next steps towards their adoption in routine clinical practice.
Collapse
Affiliation(s)
- Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Mihaela Aldea
- Department of Cancer Medicine, Institut Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Thoracic Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
10
|
Richlitzki C, Manapov F, Holzgreve A, Rabe M, Werner RA, Belka C, Unterrainer M, Eze C. Advances of PET/CT in Target Delineation of Lung Cancer Before Radiation Therapy. Semin Nucl Med 2025; 55:190-201. [PMID: 40064578 DOI: 10.1053/j.semnuclmed.2025.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
In the clinical management of lung cancer, radiotherapy remains a cornerstone of multimodal treatment strategies, often used alongside surgery or in combination with systemic therapies such as chemotherapy, tyrosine kinase inhibitors, and immune checkpoint inhibitors. While conventional imaging modalities like computed tomography (CT) and magnetic resonance imaging (MRI) continue to play a central role in staging, response assessment, and radiotherapy planning, advanced imaging techniques, particularly [18F]FDG PET/CT, are being increasingly integrated into routine clinical practice. These advanced techniques address the limitations of standard imaging by providing insight into molecular and metabolic tumor characteristics, enabling precise tumor visualization, accurate target volume delineation, and early treatment response assessment. This review examines the role of radiotherapy in the multidisciplinary management of lung cancer, detailing current concepts of morphological and functional imaging for staging and treatment planning. It also highlights the growing importance of PET-based radiotherapy planning, emphasizing its contributions to target volume definition and predictive value for treatment outcomes. Recent methodological advances, including the integration of artificial intelligence (AI), radiomics, technical innovations, and novel PET ligands, are discussed, highlighting their potential to improve the precision, efficacy, and personalization of lung cancer radiotherapy planning.
Collapse
Affiliation(s)
- Cedric Richlitzki
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Moritz Rabe
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rudolf Alexander Werner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany; Bavarian Cancer Research Center, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Die Radiologie, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
11
|
Stüber AT, Heimer MM, Ta J, Fabritius MP, Hoppe BF, Sheikh G, Brendel M, Unterrainer L, Jurmeister P, Tufman A, Ricke J, Cyran CC, Ingrisch M. Replication study of PD-L1 status prediction in NSCLC using PET/CT radiomics. Eur J Radiol 2025; 183:111825. [PMID: 39657546 DOI: 10.1016/j.ejrad.2024.111825] [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: 07/07/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
This study investigates the predictive capability of radiomics in determining programmed cell death ligand 1 (PD-L1) expression (>=1%) status in non-small cell lung cancer (NSCLC) patients using a newly collected [18F]FDG PET/CT dataset. We aimed to replicate and validate the radiomics-based machine learning (ML) model proposed by Zhao et al. [1] predicting PD-L1 status from PET/CT-imaging. An independent cohort of 254 NSCLC patients underwent [18F]FDG PET/CT imaging, with primary tumor segmentation conducted using lung tissue window (LTW) and more conservative soft tissue window (STW) methods. Radiomics models ("Rad-score" and "complex model") and a clinical-stage model from Zhao et al. were evaluated via 10-fold cross-validation and AUC analysis, alongside a benchmark-study comparing different ML-model pipelines. Clinicopathological data were collected from medical records. On our data, the Rad-score model yielded mean AUCs of 0.593 (STW) and 0.573 (LTW), below Zhao et al.'s 0.761. The complex model achieved mean AUCs of 0.505 (STW) and 0.519 (LTW), lower than Zhao et al.'s 0.769. The clinical model showed a mean AUC of 0.555, below Zhao et al.'s 0.64. All models performed significantly lower than Zhao et al.'s findings. Our benchmark study on four ML pipelines revealed consistently low performance across all configurations. Our study failed to replicate original findings, suggesting poor model performance and questioning predictive value of radiomics features in classifying PD-L1 expression from PET/CT imaging. These results highlight challenges in replicating radiomics-based ML models and stress the need for rigorous validation.
Collapse
Affiliation(s)
- Anna Theresa Stüber
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; Department of Statistics, LMU Munich, Ludwigstr. 33, 80539 Munich, Germany; Munich Center for Machine Learning (MCML), Geschwister-Scholl-Platz 1, 80539 Munich, Germany.
| | - Maurice M Heimer
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Johanna Ta
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Matthias P Fabritius
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Boj F Hoppe
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Gabriel Sheikh
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany
| | - Lena Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; Bayerisches Zentrum für Krebsforschung (BZKF), partner site Munich, Einsteinstraße 1, 81675 Munich, Germany
| | - Philip Jurmeister
- Institute of Pathology, Faculty of Medicine, LMU Munich, Thalkirchnerstr. 36, 80337 Munich, Germany
| | - Amanda Tufman
- Department of Medicine V, LMU University Hospital, LMU Munich, Ziemssenstr. 1/5, 80336 Munich, Germany
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Michael Ingrisch
- Department of Radiology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; Munich Center for Machine Learning (MCML), Geschwister-Scholl-Platz 1, 80539 Munich, Germany
| |
Collapse
|
12
|
Zhu Y, Li H, Xiao S, Yu W, Shang H, Wang L, Liu Y, Wang Y, Yang J. CDKD-w+: A Keyframe Recognition Method for Coronary Digital Subtraction Angiography Video Sequence Based on w+ Space Encoding. SENSORS (BASEL, SWITZERLAND) 2025; 25:710. [PMID: 39943348 PMCID: PMC11821101 DOI: 10.3390/s25030710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025]
Abstract
Currently, various deep learning methods can assist in medical diagnosis. Coronary Digital Subtraction Angiography (DSA) is a medical imaging technology used in cardiac interventional procedures. By employing X-ray sensors to visualize the coronary arteries, it generates two-dimensional images from any angle. However, due to the complexity of the coronary structures, the 2D images may sometimes lack sufficient information, necessitating the construction of a 3D model. Camera-level 3D modeling can be realized based on deep learning. Nevertheless, the beating of the heart results in varying degrees of arterial vasoconstriction and vasodilation, leading to substantial discrepancies between DSA sequences, which introduce errors in 3D modeling of the coronary arteries, resulting in the inability of the 3D model to reflect the coronary arteries. We propose a coronary DSA video sequence keyframe recognition method, CDKD-w+, based on w+ space encoding. The method utilizes a pSp encoder to encode the coronary DSA images, converting them into latent codes in the w+ space. Differential analysis of inter-frame latent codes is employed for heartbeat keyframe localization, aiding in coronary 3D modeling. Experimental results on a self-constructed coronary DSA heartbeat keyframe recognition dataset demonstrate an accuracy of 97%, outperforming traditional metrics such as L1, SSIM, and PSNR.
Collapse
Affiliation(s)
- Yong Zhu
- School of Electrical and Information Engineering, Tianjin University, Tianjin 300072, China; (Y.Z.); (H.L.); (J.Y.)
| | - Haoyu Li
- School of Electrical and Information Engineering, Tianjin University, Tianjin 300072, China; (Y.Z.); (H.L.); (J.Y.)
| | - Shuai Xiao
- School of Electrical and Information Engineering, Tianjin University, Tianjin 300072, China; (Y.Z.); (H.L.); (J.Y.)
| | - Wei Yu
- Tianjin Institute of Software Engineering, Tianjin 300387, China; (W.Y.); (H.S.); (L.W.); (Y.L.); (Y.W.)
| | - Hongyu Shang
- Tianjin Institute of Software Engineering, Tianjin 300387, China; (W.Y.); (H.S.); (L.W.); (Y.L.); (Y.W.)
| | - Lin Wang
- Tianjin Institute of Software Engineering, Tianjin 300387, China; (W.Y.); (H.S.); (L.W.); (Y.L.); (Y.W.)
| | - Yang Liu
- Tianjin Institute of Software Engineering, Tianjin 300387, China; (W.Y.); (H.S.); (L.W.); (Y.L.); (Y.W.)
| | - Yin Wang
- Tianjin Institute of Software Engineering, Tianjin 300387, China; (W.Y.); (H.S.); (L.W.); (Y.L.); (Y.W.)
| | - Jiachen Yang
- School of Electrical and Information Engineering, Tianjin University, Tianjin 300072, China; (Y.Z.); (H.L.); (J.Y.)
| |
Collapse
|
13
|
Yin L, Liu R, Li W, Li S, Hou X. Deep learning-based CT radiomics predicts prognosis of unresectable hepatocellular carcinoma treated with TACE-HAIC combined with PD-1 inhibitors and tyrosine kinase inhibitors. BMC Gastroenterol 2025; 25:24. [PMID: 39838292 PMCID: PMC11748841 DOI: 10.1186/s12876-024-03555-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE To develop and validate a computed tomography (CT)-based deep learning radiomics model to predict treatment response and progression-free survival (PFS) in patients with unresectable hepatocellular carcinoma (uHCC) treated with transarterial chemoembolization (TACE)-hepatic arterial infusion chemotherapy (HAIC) combined with PD-1 inhibitors and tyrosine kinase inhibitors (TKIs). METHODS This retrospective study included 172 patients with uHCC who underwent combination therapy of TACE-HAIC with TKIs and PD-1 inhibitors. Among them, 122 were from the Interventional Department of the Harbin Medical University Cancer Hospital, with 92 randomly assigned to the training cohort and 30 cases randomly assigned to the testing cohort. The remaining 50 cases were from the Interventional Department of the Affiliated Fourth Hospital of Harbin Medical University and were used for external validation. All patients underwent liver enhanced CT examination before treatment. Residual convolutional neural network (ResNet) technology was used to extract image features. A predictive model for treatment response of combination therapy and PFS was established based on image features and clinical features. Model effectiveness was evaluated using metrics such as the area under the receiver operating characteristic (ROC) curve (AUC), concordance index (C-index), accuracy, precision, and F1-score. RESULTS All patients had a median follow-up of 25.2 months (95% CI 24.4-26.0), with a median PFS of 14.0 months (95% CI 8.5-19.4) and a median overall survival (OS) of 26.2 months (95% CI 15.9-36.4) achieved. Objective response rate (ORR) and disease control rate (DCR) was 41.0% and 55.7%, respectively. In the treatment response prediction model, the AUC for the training cohort reached 0.96, with an accuracy of 89.5%, precision of 85.6%, and F1-score of 0.896; the AUC for the testing cohort was 0.87, with an accuracy of 80.4%, precision of 74.5%, and F1-score of 0.802. The AUC of the external validation cohort was 0.85, with accuracy of 79.1%, precision of 73.6%, and f1-score of 0.784. In the PFS prediction model, the predicted AUC for 12 months, 18 months, and 24 months-PFS in the training cohort were 0.874, 0.809, 0.801, respectively. The AUC of testing cohort were 0.762, 0.804, 0.792. The AUC of external validation cohort were 0.764, 0.796, 0.773. The C-index of the combination model, radiomics model, and clinical model were 0.75, 0.591, and 0.655, respectively. The calibration curve demonstrated that the combination model was significantly superior to both the radiomics and clinical models. CONCLUSIONS The study provides a CT-based radiomics model that can predict PFS for patients with uHCC treated with TACE-HAIC combined with PD-1 and TKIs.
Collapse
Affiliation(s)
- Linan Yin
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Ruibao Liu
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China.
| | - Wei Li
- Department of Interventional Radiology, Affiliated Fourth Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Shijie Li
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Xunbo Hou
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| |
Collapse
|
14
|
Wu L, Wei D, Li S, Wu S, Lin Y, Chen L. The potential of MRI radiomics based on extrapulmonary metastases in predicting EGFR mutations: a systematic review and meta-analysis. Biomed Eng Online 2025; 24:4. [PMID: 39825348 PMCID: PMC11742221 DOI: 10.1186/s12938-025-01331-6] [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: 07/03/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) gene mutations can lead to distant metastasis in non-small cell lung cancer (NSCLC). When the primary NSCLC lesions are removed or cannot be sampled, the EGFR status of the metastatic lesions are the potential alternative method to reflect EGFR mutations in the primary NSCLC lesions. This review aimed to evaluate the potential of magnetic resonance imaging (MRI) radiomics based on extrapulmonary metastases in predicting EGFR mutations through a systematic reviews and meta-analysis. MATERIALS AND METHODS A systematic review of the studies on MRI radiomics based on extrapulmonary metastases in predicting EGFR mutations. The area under the curve (AUC), sensitivity (SNEC), and specificity (SPEC) of each study were separately extracted for comprehensive evaluation of MRI radiomics in predicting EGFR mutations in primary or metastatic NSCLC. RESULTS Thirteen studies were ultimately included, with 2369 cases of metastatic NSCLC, including five studies predicting EGFR mutations in primary NSCLC, eight studies predicting EGFR mutations in metastatic NSCL. In terms of EGFR mutations in the primary lesion of NSCLC, the pooled AUC was 0.90, with SENC and SPEC of 0.80 and 0.85, respectively, which seems superior to the radiomics meta-analysis based on NSCLC primary lesions. In terms of EGFR mutations in NSCLC metastases, the pooled AUC was 0.86, with SENC and SEPC of 0.79 and 0.79, respectively, indicating moderate evaluation performance. CONCLUSIONS MRI radiomics helps to predict the EGFR mutation status in the primary or metastatic lesions of NSCLC, serve as a high-precision supplement to current molecular detection methods.
Collapse
Affiliation(s)
- Linyong Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China
| | - Dayou Wei
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China.
| | - Songhua Li
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China
| | - Shaofeng Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China
| | - Yan Lin
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China
| | - Lifei Chen
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525011, People's Republic of China
| |
Collapse
|
15
|
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
|
16
|
Wu L, Wei D, Chen W, Wu C, Lu Z, Li S, Liu W. Comprehensive Potential of Artificial Intelligence for Predicting PD-L1 Expression and EGFR Mutations in Lung Cancer: A Systematic Review and Meta-Analysis. J Comput Assist Tomogr 2025; 49:101-112. [PMID: 39143665 DOI: 10.1097/rct.0000000000001644] [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: 08/16/2024]
Abstract
OBJECTIVE To evaluate the methodological quality and the predictive performance of artificial intelligence (AI) for predicting programmed death ligand 1 (PD-L1) expression and epidermal growth factor receptors (EGFR) mutations in lung cancer (LC) based on systematic review and meta-analysis. METHODS AI studies based on PET/CT, CT, PET, and immunohistochemistry (IHC)-whole-slide image (WSI) were included to predict PD-L1 expression or EGFR mutations in LC. The modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was used to evaluate the methodological quality. A comprehensive meta-analysis was conducted to analyze the overall area under the curve (AUC). The Cochrane diagnostic test and I2 statistics were used to assess the heterogeneity of the meta-analysis. RESULTS A total of 45 AI studies were included, of which 10 were used to predict PD-L1 expression and 35 were used to predict EGFR mutations. Based on the analysis using the QUADAS-2 tool, 37 studies achieved a high-quality score of 7. In the meta-analysis of PD-L1 expression levels, the overall AUCs for PET/CT, CT, and IHC-WSI were 0.80 (95% confidence interval [CI], 0.77-0.84), 0.74 (95% CI, 0.69-0.77), and 0.95 (95% CI, 0.93-0.97), respectively. For EGFR mutation status, the overall AUCs for PET/CT, CT, and PET were 0.85 (95% CI, 0.81-0.88), 0.83 (95% CI, 0.80-0.86), and 0.75 (95% CI, 0.71-0.79), respectively. The Cochrane Diagnostic Test revealed an I2 value exceeding 50%, indicating substantial heterogeneity in the PD-L1 and EGFR meta-analyses. When AI was combined with clinicopathological features, the enhancement in predicting PD-L1 expression was not substantial, whereas the prediction of EGFR mutations showed improvement compared to the CT and PET models, albeit not significantly so compared to the PET/CT models. CONCLUSIONS The overall performance of AI in predicting PD-L1 expression and EGFR mutations in LC has promising clinical implications.
Collapse
Affiliation(s)
- Linyong Wu
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Dayou Wei
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Wubiao Chen
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| | - Chaojun Wu
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Zhendong Lu
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| | - Songhua Li
- From the Department of Medical Ultrasound, Maoming People's Hospital, Maoming
| | - Wenci Liu
- Radiology Imaging Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, PR China
| |
Collapse
|
17
|
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
|
18
|
Magnin CY, Lauer D, Ammeter M, Gote-Schniering J. From images to clinical insights: an educational review on radiomics in lung diseases. Breathe (Sheff) 2025; 21:230225. [PMID: 40104259 PMCID: PMC11915127 DOI: 10.1183/20734735.0225-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/20/2025] Open
Abstract
Radiological imaging is a cornerstone in the clinical workup of lung diseases. Radiomics represents a significant advancement in clinical lung imaging, offering a powerful tool to complement traditional qualitative image analysis. Radiomic features are quantitative and computationally describe shape, intensity, texture and wavelet characteristics from medical images that can uncover detailed and often subtle information that goes beyond the visual capabilities of radiological examiners. By extracting this quantitative information, radiomics can provide deep insights into the pathophysiology of lung diseases and support clinical decision-making as well as personalised medicine approaches. In this educational review, we provide a step-by-step guide to radiomics-based medical image analysis, discussing the technical challenges and pitfalls, and outline the potential clinical applications of radiomics in diagnosing, prognosticating and evaluating treatment responses in respiratory medicine.
Collapse
Affiliation(s)
- Cheryl Y Magnin
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - David Lauer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - Michael Ammeter
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Janine Gote-Schniering
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Meng X, Wang X, Zhang Z, Song L, Chen J. Recent Advancements of Nanomedicine in Breast Cancer Surgery. Int J Nanomedicine 2024; 19:14143-14169. [PMID: 39759962 PMCID: PMC11699852 DOI: 10.2147/ijn.s494364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer surgery plays a pivotal role in the multidisciplinary approaches. Surgical techniques and objectives are gradually shifting from tumor complete resection towards prolonging survival, improving cosmetic outcomes, and restoring the social and psychological well-being of patients. However, surgical treatment still faces challenges such as inadequate sensitivity in sentinel lymph node localization, the need to improve intraoperative tumor boundary localization imaging, postoperative scar healing, and the risk of recurrence, necessitating other adjunct measures for improvement. To address these challenges, specificity-optimized nanomedicines have been introduced into the surgical therapeutic landscape of breast cancer. In particular, this review involves starting with an overview of breast structure and the composition of the tumor microenvironment and then introducing the guiding principle and foundation for the design of nanomedicine. Moreover, we will take the order process of breast cancer surgery diagnosis and treatment as the starting point, and adaptively propose the roles and advantages of nanomedicine in addressing the corresponding issues. Furthermore, we also involved the prospects of utilizing advanced technological approaches. Overall, this review seeks to uncover the sophisticated design and strategies of nanomedicine from a clinical standpoint, address the challenges faced in surgical treatment, and provide insights into this subject matter.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xin Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhihao Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Linlin Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jie Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
20
|
Ter Maat LS, De Mooij RAJ, Van Duin IAJ, Verhoeff JJC, Elias SG, Leiner T, van Amsterdam WAC, Troenokarso MF, Arntz ERAN, Van den Berkmortel FWPJ, Boers-Sonderen MJ, Boomsma MF, Van den Eertwegh FJM, de Groot JW, Hospers GAP, Piersma D, Vreugdenhil A, Westgeest HM, Kapiteijn E, De Wit AA, Blokx WAM, Van Diest PJ, De Jong PA, Pluim JPW, Suijkerbuijk KPM, Veta M. Deep learning on CT scans to predict checkpoint inhibitor treatment outcomes in advanced melanoma. Sci Rep 2024; 14:31668. [PMID: 39738216 PMCID: PMC11686296 DOI: 10.1038/s41598-024-81188-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) treatment has proven successful for advanced melanoma, but is associated with potentially severe toxicity and high costs. Accurate biomarkers for response are lacking. The present work is the first to investigate the value of deep learning on CT imaging of metastatic lesions for predicting ICI treatment outcomes in advanced melanoma. Adult patients that were treated with ICI for advanced melanoma were retrospectively identified from ten participating centers. A deep learning model (DLM) was trained on volumes of lesions on baseline CT to predict clinical benefit. The DLM was compared to and combined with a model of known clinical predictors (presence of liver and brain metastasis, level of lactate dehydrogenase, performance status and number of affected organs). A total of 730 eligible patients with 2722 lesions were included. The DLM reached an area under the receiver operating characteristic (AUROC) of 0.607 [95%CI 0.565-0.648]. In comparison, a model of clinical predictors reached an AUROC of 0.635 [95%CI 0.59 -0.678]. The combination model reached an AUROC of 0.635 [95% CI 0.595-0.676]. Differences in AUROC were not statistically significant. The output of the DLM was significantly correlated with four of the five input variables of the clinical model. The DLM reached a statistically significant discriminative value, but was unable to improve over known clinical predictors. The present work shows that the assessment over known clinical predictors is an essential step for imaging-based prediction and brings important nuance to the almost exclusively positive findings in this field.
Collapse
Affiliation(s)
- Laurens S Ter Maat
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rob A J De Mooij
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Isabella A J Van Duin
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joost J C Verhoeff
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tim Leiner
- Department of Radiology, Mayo Clinical, Rochester, MN, USA
| | - Wouter A C van Amsterdam
- Department of Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | | | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboudumc, Radboud University, Nijmegen, The Netherlands
| | | | - Fons J M Van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
| | | | - Geke A P Hospers
- Department of Medical Oncology, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Djura Piersma
- Department of Medical Oncology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Art Vreugdenhil
- Department of Medical Oncology, Maxima Medical Center, Veldhoven, The Netherlands
| | - Hans M Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Ardine A De Wit
- Department of Public Health, Healthcare Innovation and Evaluation and Medical Humanities, Julius Center Research Program Methodology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Paul J Van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pim A De Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Josien P W Pluim
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Mitko Veta
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
21
|
Huang L, Kong W, Luo Y, Xie H, Liu J, Zhang X, Zhang G. Predicting epidermal growth factor receptor mutation status of lung adenocarcinoma based on PET/CT images using deep learning. Front Oncol 2024; 14:1458374. [PMID: 39735601 PMCID: PMC11671303 DOI: 10.3389/fonc.2024.1458374] [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: 07/02/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024] Open
Abstract
Background The aim of this study is to develop deep learning models based on 18F-fluorodeoxyglucose positron emission tomography/computed tomographic (18F-FDG PET/CT) images for predicting individual epidermal growth factor receptor (EGFR) mutation status in lung adenocarcinoma (LUAD). Methods We enrolled 430 patients with non-small-cell lung cancer from two institutions in this study. The advanced Inception V3 model to predict EGFR mutations based on PET/CT images and developed CT, PET, and PET + CT models was used. Additionally, each patient's clinical characteristics (age, sex, and smoking history) and 18 CT features were recorded and analyzed. Univariate and multivariate regression analyses identified the independent risk factors for EGFR mutations, and a clinical model was established. The performance using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, specificity, recall, and F1-value was evaluated. The DeLong test was used to compare the predictive performance across various models. Results Among these four models, deep learning models based on CT and PET + CT exhibit the same predictive performance, followed by PET and the clinical model. The AUC values for CT, PET, PET + CT, and clinical models in the training set are 0.933 (95% CI, 0.922-0.943), 0.895 (95% CI, 0.882-0.907), 0.931 (95% CI, 0.921-0.942), and 0.740 (95% CI, 0.685-0.796), respectively; whereas those in the testing set are:0.921 (95% CI, 0.904-0.938), 0.876 (95% CI, 0.855-0.897), 0.921 (95% CI, 0.904-0.937), and 0.721 (95% CI, 0.629-0.814), respectively. The DeLong test results confirm that all deep learning models are superior to clinical one. Conclusion The PET/CT images based on trained CNNs effectively predict EGFR and non-EGFR mutations in LUAD. The deep learning predictive models could guide treatment options.
Collapse
Affiliation(s)
- Lele Huang
- Department of Nuclear Medicine, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Weifang Kong
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yongjun Luo
- Department of Nuclear Medicine, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Hongjun Xie
- Department of Nuclear Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangyan Liu
- Department of Nuclear Medicine, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xin Zhang
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Beijing, China
| | - Guojin Zhang
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
22
|
Cheng H, Bai L, Zhang X, Chen W, He S, Liu Y, Wang J, Song S. 68Ga labeled Olmutinib: Design, synthesis, and evaluation of a novel PET EGFR probe. Bioorg Chem 2024; 153:107987. [PMID: 39579551 DOI: 10.1016/j.bioorg.2024.107987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Radiolabeled tyrosine kinase inhibitors (TKIs) offer a promising approach for molecular imaging of EGFR-positive cancers. Despite the development of various EGFR small-molecule probes, none of the 68Ga-labeled small-molecule probes based on the chelator DOTA have shown tumor-specific uptake. To address this challenge, we selected Olmutinib, a third-generation EGFR covalent inhibitor, as a PET imaging tracer for EGFR-positive tumors. We synthesized the precursor DOTA-Olmutinib through a five-step process and subsequently radiolabeled it with 68Ga to prepare 68Ga-DOTA-Olmutinib. 68Ga-DOTA-Olmutinib displayed moderate lipophilicity (log P = 0.85) and exhibited high stability in vitro and in vivo. Western blot analysis was used to detect the level of EGFR in multiple tumor cells. In cell uptake experiments, 68Ga-DOTA-Olmutinib exhibited enhanced uptake specifically in tumor cells with a higher level of EGFR supporting it as an EGFR-specific tracer. Additionally, PET/CT imaging with 68Ga-DOTA-Olmutinib showed significant tumor uptake at 60 min with 4 % ID/g post-injection, marking a breakthrough, though the uptake is not yet ideal. Overall, our results suggest that 68Ga-labeled Olmutinib holds promise as a potential PET tracer for detecting EGFR-positive cancers.
Collapse
Affiliation(s)
- Hua Cheng
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China; Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Liyan Bai
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xi Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Wenfei Chen
- The Fifth Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Xinjiang 830011, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yunqi Liu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Juan Wang
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Koyama J, Morise M, Furukawa T, Oyama S, Matsuzawa R, Tanaka I, Wakahara K, Yokota H, Kimura T, Shiratori Y, Kondoh Y, Hashimoto N, Ishii M. Artificial intelligence-based personalized survival prediction using clinical and radiomics features in patients with advanced non-small cell lung cancer. BMC Cancer 2024; 24:1417. [PMID: 39558311 PMCID: PMC11572056 DOI: 10.1186/s12885-024-13190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Multiple first-line treatment options have been developed for advanced non-small cell lung cancer (NSCLC) in each subgroup determined by predictive biomarkers, specifically driver oncogene and programmed cell death ligand-1 (PD-L1) status. However, the methodology for optimal treatment selection in individual patients is not established. This study aimed to develop artificial intelligence (AI)-based personalized survival prediction model according to treatment selection. METHODS The prediction model was built based on random survival forest (RSF) algorithm using patient characteristics, anticancer treatment histories, and radiomics features of the primary tumor. The predictive accuracy was validated with external test data and compared with that of cox proportional hazard (CPH) model. RESULTS A total of 459 patients (training, n = 299; test, n = 160) with advanced NSCLC were enrolled. The algorithm identified following features as significant factors associated with survival: age, sex, performance status, Brinkman index, comorbidity of chronic obstructive pulmonary disease, histology, stage, driver oncogene status, tumor PD-L1 expression, administered anticancer agent, six markers of blood test (sodium, lactate dehydrogenase, etc.), and three radiomics features associated with tumor texture, volume, and shape. The C-index of RSF model for test data was 0.841, which was higher than that of CPH model (0.775, P < 0.001). Furthermore, the RSF model enabled to identify poor survivor treated with pembrolizumab because of tumor PD-L1 high expression and those treated with driver oncogene targeted therapy according to driver oncogene status. CONCLUSIONS The proposed AI-based algorithm accurately predicted the survival of each patient with advanced NSCLC. The AI-based methodology will contribute to personalized medicine. TRIAL REGISTRATION The trial design was retrospectively registered study performed in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Nagoya University Graduate School of Medicine (approval: 2020 - 0287).
Collapse
Affiliation(s)
- Junji Koyama
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan.
| | - Taiki Furukawa
- Medical IT Center, Nagoya University Hospital, Nagoya, Japan
| | - Shintaro Oyama
- Innovative Research Center for Preventive Medical Engineering (PME), Nagoya University, Nagoya, Japan
- Image Processing Research Team, RIKEN Center for Advanced Photonics, Wako, Japan
| | - Reiko Matsuzawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan
| | - Ichidai Tanaka
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan
| | - Keiko Wakahara
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan
| | - Hideo Yokota
- Image Processing Research Team, RIKEN Center for Advanced Photonics, Wako, Japan
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, Wako, Japan
| | - Tomoki Kimura
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | | | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 4668550, Japan
| |
Collapse
|
24
|
Kumar A, Dixit S, Srinivasan K, M D, Vincent PMDR. Personalized cancer vaccine design using AI-powered technologies. Front Immunol 2024; 15:1357217. [PMID: 39582860 PMCID: PMC11581883 DOI: 10.3389/fimmu.2024.1357217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 09/24/2024] [Indexed: 11/26/2024] Open
Abstract
Immunotherapy has ushered in a new era of cancer treatment, yet cancer remains a leading cause of global mortality. Among various therapeutic strategies, cancer vaccines have shown promise by activating the immune system to specifically target cancer cells. While current cancer vaccines are primarily prophylactic, advancements in targeting tumor-associated antigens (TAAs) and neoantigens have paved the way for therapeutic vaccines. The integration of artificial intelligence (AI) into cancer vaccine development is revolutionizing the field by enhancing various aspect of design and delivery. This review explores how AI facilitates precise epitope design, optimizes mRNA and DNA vaccine instructions, and enables personalized vaccine strategies by predicting patient responses. By utilizing AI technologies, researchers can navigate complex biological datasets and uncover novel therapeutic targets, thereby improving the precision and efficacy of cancer vaccines. Despite the promise of AI-powered cancer vaccines, significant challenges remain, such as tumor heterogeneity and genetic variability, which can limit the effectiveness of neoantigen prediction. Moreover, ethical and regulatory concerns surrounding data privacy and algorithmic bias must be addressed to ensure responsible AI deployment. The future of cancer vaccine development lies in the seamless integration of AI to create personalized immunotherapies that offer targeted and effective cancer treatments. This review underscores the importance of interdisciplinary collaboration and innovation in overcoming these challenges and advancing cancer vaccine development.
Collapse
Affiliation(s)
- Anant Kumar
- School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Shriniket Dixit
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Kathiravan Srinivasan
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Dinakaran M
- School of Computer Science and Engineering, Vellore Institute of Technology, Chennai, India
| | - P. M. Durai Raj Vincent
- School of Computer Science Engineering and Information Systems, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
25
|
Ferro A, Bottosso M, Dieci MV, Scagliori E, Miglietta F, Aldegheri V, Bonanno L, Caumo F, Guarneri V, Griguolo G, Pasello G. Clinical applications of radiomics and deep learning in breast and lung cancer: A narrative literature review on current evidence and future perspectives. Crit Rev Oncol Hematol 2024; 203:104479. [PMID: 39151838 DOI: 10.1016/j.critrevonc.2024.104479] [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: 01/10/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024] Open
Abstract
Radiomics, analysing quantitative features from medical imaging, has rapidly become an emerging field in translational oncology. Radiomics has been investigated in several neoplastic malignancies as it might allow for a non-invasive tumour characterization and for the identification of predictive and prognostic biomarkers. Over the last few years, evidence has been accumulating regarding potential clinical applications of machine learning in many crucial moments of cancer patients' history. However, the incorporation of radiomics in clinical decision-making process is still limited by low data reproducibility and study variability. Moreover, the need for prospective validations and standardizations is emerging. In this narrative review, we summarize current evidence regarding radiomic applications in high-incidence cancers (breast and lung) for screening, diagnosis, staging, treatment choice, response, and clinical outcome evaluation. We also discuss pro and cons of the radiomic approach, suggesting possible solutions to critical issues which might invalidate radiomics studies and propose future perspectives.
Collapse
Affiliation(s)
- Alessandra Ferro
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Michele Bottosso
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Maria Vittoria Dieci
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy.
| | - Elena Scagliori
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Federica Miglietta
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Vittoria Aldegheri
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Laura Bonanno
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Francesca Caumo
- Unit of Breast Radiology, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Valentina Guarneri
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Gaia Griguolo
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Giulia Pasello
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
26
|
Jia Y, Wu Q, Yang Z, Sun R, Zhang K, Guo X, Xu R, Guo Y. Mechanisms of myocardial toxicity of antitumor drugs and potential therapeutic strategies: A review of the literature. Curr Probl Cardiol 2024; 49:102782. [PMID: 39134104 DOI: 10.1016/j.cpcardiol.2024.102782] [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: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
With the successive development of chemotherapy drugs, good results have been achieved in clinical application. However, myocardial toxicity is the biggest challenge. Anthracyclines, immune checkpoint inhibitors, and platinum drugs are widely used. Targeted drug delivery, nanomaterials and dynamic imaging evaluation are all emerging research directions. This article reviews the recent literature on the use of targeted nanodrug delivery and imaging techniques to evaluate the myocardial toxicity of antineoplastic drugs, and discusses the potential mechanisms.
Collapse
Affiliation(s)
- Yang Jia
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Qihong Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Zhigang Yang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu 610041, China
| | - Ran Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Kun Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Xia Guo
- Department of Hematology, West China Second University Hospital, Sichuan University; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Rong Xu
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| | - Yingkun Guo
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
27
|
Zuo Y, Liu Q, Li N, Li P, Fang Y, Bian L, Zhang J, Song S. Explainable 18F-FDG PET/CT radiomics model for predicting EGFR mutation status in lung adenocarcinoma: a two-center study. J Cancer Res Clin Oncol 2024; 150:469. [PMID: 39436414 PMCID: PMC11496337 DOI: 10.1007/s00432-024-05998-7] [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: 09/01/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
PURPOSE To establish an explainable 18F-FDG PET/CT-derived prediction model to identify EGFR mutation status and subtypes (EGFR wild, EGFR-E19, and EGFR-E21) in lung adenocarcinoma (LUAD). METHODS Baseline 18F-FDG PET/CT images of 478 patients with LUAD from 2 hospitals were collected. Data from hospital A (n = 390) was randomly split into a training group (n = 312) and an internal test group (n = 78), with data from hospital B (n = 88) utilized for external test. Further, a total of 4,760 handcrafted radiomics features (HRFs) were extracted from PET/CT scans. Candidates for the prediction model were constructed by cross-combinations of 11 feature selection methods and 7 classifiers. The optimal model was determined by combining the results of cross-center data validation and model visualization (Yellowbrick). The predictive performance was assessed via receiver operating characteristic curve, confusion matrix and classification report. Four explainable artificial intelligence technologies were used for optimal model interpretation. RESULTS Sex and SUVmax were selected as clinical risk factors, which were then combined with 8 robust PET/CT HRFs to establish the models. The optimal performance was obtained by combining a light gradient boosting machine classifier with random forest feature selection method achieving an optimal performance with a macro-average AUC of 0.75 in the internal test group and 0.81 in the external test group. CONCLUSION The explainable EGFR mutation status prediction model have certain clinical practicability and good generalization performance, which may help in the timely selection of treatment options and prognosis prediction in patients with LUAD.
Collapse
Affiliation(s)
- Yan Zuo
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Nan Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Panli Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Yichong Fang
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200233, P. R. China
| | - Linjie Bian
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, P. R. China.
- Center for Biomedical Imaging, Fudan University, shanghai, 200032, P. R. China.
- Shanghai Engineering Research Center of Molecular Imaging Probes, shanghai, 200032, P. R. China.
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, shanghai, 200433, P. R. China.
| |
Collapse
|
28
|
Lin A, Zhang H, Wang Y, Cui Q, Zhu K, Zhou D, Han S, Meng S, Han J, Li L, Zhou C, Ma X. Radiomics based on MRI to predict recurrent L4-5 disc herniation after percutaneous endoscopic lumbar discectomy. BMC Med Imaging 2024; 24:273. [PMID: 39390384 PMCID: PMC11468133 DOI: 10.1186/s12880-024-01450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND In recent years, radiomics has been shown to be an effective tool for the diagnosis and prediction of diseases. Existing evidence suggests that imaging features play a key role in predicting the recurrence of lumbar disk herniation (rLDH). Thus, this study aimed to evaluate the risk of rLDH in patients undergoing percutaneous endoscopic lumbar discectomy (PELD) using radiomics to facilitate the development of more rational surgical and perioperative management strategies. METHOD This was a retrospective case-control study involving 487 patients who underwent PELD at the L4/5 level. The rLDH and negative groups were matched using propensity score matching (PSM). A total of 1409 radiomic features were extracted from preoperative lumbar MRI images using intraclass correlation coefficient (ICC) analysis, t-test, and LASSO analysis. Afterward, 6 predictive models were constructed and evaluated using ROC curve analysis, AUC, specificity, sensitivity, confusion matrix, and 2 repeated 3-fold cross-validations. Lastly, the Shapley Additive Explanation (SHAP) analysis provided visual explanations for the models. RESULTS Following screening and matching, 128 patients were included in both the recurrence and control groups. Moreover, 18 of the extracted radiomic features were selected for generating six models, which achieved an AUC of 0.551-0.859 for predicting rLDH. Among these models, SVM, RF, and XG Boost exhibited superior performances. Finally, cross-validation revealed that their accuracy was 0.674-0.791, 0.647-0.729, and 0.674-0.718. CONCLUSION Radiomics based on MRI can be used to predict the risk of rLDH, offering more comprehensive guidance for perioperative treatment by extracting imaging information that cannot be visualized with the naked eye. Meanwhile, the accuracy and generalizability of the model can be improved in the future by incorporating more data and conducting multicenter studies.
Collapse
Affiliation(s)
- Antao Lin
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Hao Zhang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Yan Wang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Qian Cui
- Department of Medical Imaging, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Kai Zhu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Dan Zhou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Shuo Han
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Shengwei Meng
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Jialuo Han
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Lei Li
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China
| | - Chuanli Zhou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China.
| | - Xuexiao Ma
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, People's Republic of China.
| |
Collapse
|
29
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
30
|
Xiao Y, Li Y, Zhao H. Spatiotemporal metabolomic approaches to the cancer-immunity panorama: a methodological perspective. Mol Cancer 2024; 23:202. [PMID: 39294747 PMCID: PMC11409752 DOI: 10.1186/s12943-024-02113-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Metabolic reprogramming drives the development of an immunosuppressive tumor microenvironment (TME) through various pathways, contributing to cancer progression and reducing the effectiveness of anticancer immunotherapy. However, our understanding of the metabolic landscape within the tumor-immune context has been limited by conventional metabolic measurements, which have not provided comprehensive insights into the spatiotemporal heterogeneity of metabolism within TME. The emergence of single-cell, spatial, and in vivo metabolomic technologies has now enabled detailed and unbiased analysis, revealing unprecedented spatiotemporal heterogeneity that is particularly valuable in the field of cancer immunology. This review summarizes the methodologies of metabolomics and metabolic regulomics that can be applied to the study of cancer-immunity across single-cell, spatial, and in vivo dimensions, and systematically assesses their benefits and limitations.
Collapse
Affiliation(s)
- Yang Xiao
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400044, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Huakan Zhao
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400044, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
31
|
Wang N, Dai M, Jing F, Liu Y, Zhao Y, Zhang Z, Wang J, Zhang J, Wang Y, Zhao X. Value of 18F-FDG PET/CT-based radiomics features for differentiating primary lung cancer and solitary lung metastasis in patients with colorectal adenocarcinoma. Int J Radiat Biol 2024:1-9. [PMID: 39288285 DOI: 10.1080/09553002.2024.2404465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE To investigate the value and applicability of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics in differentiating primary lung cancer (PLC) from solitary lung metastasis (SLM) in patients with colorectal cancer (CRC). MATERIALS AND METHODS This retrospective study included 103 patients with CRC and solitary pulmonary nodules (SPNs). The least absolute shrinkage and selection operator (LASSO) was used to screen for optimal radiomics features and establish a PET/CT radiomics model. PET/CT Visual and complex models (combining radiomics with PET/CT visual features) were developed. The area under the receiver operating characteristic (ROC) curve (AUC) was used to determine the predictive value and diagnostic efficiency of the models. RESULTS The AUC of the PET/CT radiomics model for differentiating PLC from SLM was 0.872 (95% CI: 0.806-0.939), which was not different from that of the visual (0.829 [95% CI: 0.749-0.908; p = .352]). However, the AUC of the complex model (0.936 [95% CI:0.892-0.981]) was significantly higher than that of the PET/CT radiomics (p = .005) and visual model (p = .001). The sensitivity (SEN), specificity (SPE), accuracy (ACC), positive predictive value (PPV), and negative predictive value (NPV) of PET/CT radiomics for differentiating PLC from SLM were 0.720, 0.887, 0.806, 0.857, and 0.770, respectively. CONCLUSION PET/CT radiomics can effectively distinguish PLC and SLM in patients with CRC and SPNs and guide the implementation of personalized treatment.
Collapse
Affiliation(s)
- Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Yingchen Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| |
Collapse
|
32
|
He W, Huang W, Zhang L, Wu X, Zhang S, Zhang B. Radiogenomics: bridging the gap between imaging and genomics for precision oncology. MedComm (Beijing) 2024; 5:e722. [PMID: 39252824 PMCID: PMC11381657 DOI: 10.1002/mco2.722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/06/2024] [Accepted: 08/18/2024] [Indexed: 09/11/2024] Open
Abstract
Genomics allows the tracing of origin and evolution of cancer at molecular scale and underpin modern cancer diagnosis and treatment systems. Yet, molecular biomarker-guided clinical decision-making encounters major challenges in the realm of individualized medicine, consisting of the invasiveness of procedures and the sampling errors due to high tumor heterogeneity. By contrast, medical imaging enables noninvasive and global characterization of tumors at a low cost. In recent years, radiomics has overcomes the limitations of human visual evaluation by high-throughput quantitative analysis, enabling the comprehensive utilization of the vast amount of information underlying radiological images. The cross-scale integration of radiomics and genomics (hereafter radiogenomics) has the enormous potential to enhance cancer decoding and act as a catalyst for digital precision medicine. Herein, we provide a comprehensive overview of the current framework and potential clinical applications of radiogenomics in patient care. We also highlight recent research advances to illustrate how radiogenomics can address common clinical problems in solid tumors such as breast cancer, lung cancer, and glioma. Finally, we analyze existing literature to outline challenges and propose solutions, while also identifying future research pathways. We believe that the perspectives shared in this survey will provide a valuable guide for researchers in the realm of radiogenomics aiming to advance precision oncology.
Collapse
Affiliation(s)
- Wenle He
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Wenhui Huang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Lu Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Xuewei Wu
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Shuixing Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Bin Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| |
Collapse
|
33
|
HOU Y, ZHANG T, WANG H. [Advancements in Radiomics for Immunotherapy of Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:637-644. [PMID: 39318257 PMCID: PMC11425675 DOI: 10.3779/j.issn.1009-3419.2024.102.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Indexed: 09/26/2024]
Abstract
Lung cancer is the main cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) being the predominant subtype. At present, immunotherapy represented by immune checkpoint inhibitors (ICIs) of programmed cell death receptor 1 or its ligand has been widely used in the clinical diagnosis and treatment of patients with NSCLC. However, only a few patients can benefit from it, and reliable predictive markers for immunotherapy are lacking. Radiomics is a tool that uses computer software and algorithms to extract a large amount of quantitative information from biomedical images. A large number of studies have confirmed that the radiomic model that predicts the immune efficacy of NSCLC can be used as a new type of immune efficacy predictive marker, which is expected to guide the individualized diagnosis and treatment decisions for patients with lung cancer and has a bright application prospect. This article reviews the research progress of radiomics in predicting the immune therapy response of NSCLC, identifying pseudo-progression and hyperprogression, ICIs-related pneumonia, cachexia risk, and combining with other genomics.
.
Collapse
|
34
|
Akazawa M, Hashimoto K. Prediction of hemorrhage in placenta previa: Radiomics analysis of pelvic MRI images. Eur J Obstet Gynecol Reprod Biol 2024; 299:37-42. [PMID: 38830301 DOI: 10.1016/j.ejogrb.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Prediction of intraoperative massive hemorrhage is still challenging in placenta previa. Radiomics analysis has been investigated as a new evaluation method for analyzing medical images. We used radiomics analysis on placental magnetic resonance imaging (MRI) images to predict intraoperative hemorrhage in placenta previa. METHODS We used the sagittal MRI T2-weighted sequence in placenta previa. Using the rectangular region from the uterine os to the anterior wall, we extracted 97 radiomics features. We also collected patient demographics and blood test data as clinical variables. Combining these radiomics features and clinical variables, logistic regression models with a stepwise method were built to predict the risk of hemorrhage, defined as blood loss of > 2000 ml. We evaluated the prediction performance of the models using accuracy and area under the curve (AUC), also analyzing the important variables for the prediction by stepwise methods. RESULTS We enrolled a total of 63 placenta previa cases including 30 hemorrhage cases from two institutes. The model combining clinical variables and radiomics features showed the best prediction performance with an accuracy of 0.70 and an AUC of 0.69 in the internal validation data, and accuracy of 0.41 and an AUC of 0.70 in the external validation data, compared with human experts (accuracy of 0.62). Regarding variable selection, two radiomics features. 'original_glrlm_LowGrayLevelRunEmphasis,' and 'diagnostics_Image-original_Minimum,' were important predictors for hemorrhage by the stepwise method. DISCUSSION Radiomics features based on MRI could be used as effective predictive variables for hemorrhage in placenta previa. Radiomics analysis of placental imaging could lead to further analysis of quantitative variables related to obstetric diseases.
Collapse
Affiliation(s)
- Munetoshi Akazawa
- Department of Obstetrics and Gynecology, Tokyo Women's Medical University Adachi Medical Center, Adachi‑ku, Kohoku 2‑1‑10, Tokyo, Japan.
| | - Kazunori Hashimoto
- Department of Obstetrics and Gynecology, Tokyo Women's Medical University Adachi Medical Center, Adachi‑ku, Kohoku 2‑1‑10, Tokyo, Japan
| |
Collapse
|
35
|
Yang M, Li X, Cai C, Liu C, Ma M, Qu W, Zhong S, Zheng E, Zhu H, Jin F, Shi H. [ 18F]FDG PET-CT radiomics signature to predict pathological complete response to neoadjuvant chemoimmunotherapy in non-small cell lung cancer: a multicenter study. Eur Radiol 2024; 34:4352-4363. [PMID: 38127071 DOI: 10.1007/s00330-023-10503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES This study aims to develop and validate a radiomics model based on 18F-fluorodeoxyglucose positron emission tomography-computed tomography ([18F]FDG PET-CT) images to predict pathological complete response (pCR) to neoadjuvant chemoimmunotherapy in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS One hundred eighty-five patients receiving neoadjuvant chemoimmunotherapy for NSCLC at 5 centers from January 2019 to December 2022 were included and divided into a training cohort and a validation cohort. Radiomics models were constructed via the least absolute shrinkage and selection operator (LASSO) method. The performances of models were evaluated by the area under the receiver operating characteristic curve (AUC). In addition, genetic analyses were conducted to reveal the underlying biological basis of the radiomics score. RESULTS After the LASSO process, 9 PET-CT radiomics features were selected for pCR prediction. In the validation cohort, the ability of PET-CT radiomics model to predict pCR was shown to have an AUC of 0.818 (95% confidence interval [CI], 0.711, 0.925), which was better than the PET radiomics model (0.728 [95% CI, 0.610, 0.846]), CT radiomics model (0.732 [95% CI, 0.607, 0.857]), and maximum standard uptake value (0.603 [95% CI, 0.473, 0.733]) (p < 0.05). Moreover, a high radiomics score was related to the upregulation of pathways suppressing tumor proliferation and the infiltration of antitumor immune cell. CONCLUSION The proposed PET-CT radiomics model was capable of predicting pCR to neoadjuvant chemoimmunotherapy in NSCLC patients. CLINICAL RELEVANCE STATEMENT This study indicated that the generated 18F-fluorodeoxyglucose positron emission tomography-computed tomography radiomics model could predict pathological complete response to neoadjuvant chemoimmunotherapy, implying the potential of our radiomics model to personalize the neoadjuvant chemoimmunotherapy in lung cancer patients. KEY POINTS • Recognizing patients potentially benefiting neoadjuvant chemoimmunotherapy is critical for individualized therapy of lung cancer. • [18F]FDG PET-CT radiomics could predict pathological complete response to neoadjuvant immunotherapy in non-small cell lung cancer. • [18F]FDG PET-CT radiomics model could personalize neoadjuvant chemoimmunotherapy in lung cancer patients.
Collapse
Affiliation(s)
- Minglei Yang
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Xiaoxiao Li
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Chuang Cai
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chunli Liu
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Minjie Ma
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wendong Qu
- Department of Thoracic Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| | | | - Enkuo Zheng
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Huangkai Zhu
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Feng Jin
- Shandong Key Laboratory of Infectious Respiratory Diseases, Shandong Public Health Clinical Center, Shandong University, Shandong, China.
| | - Huazheng Shi
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China.
| |
Collapse
|
36
|
Wang Y, Zhang L, Jiang Y, Cheng X, He W, Yu H, Li X, Yang J, Yao G, Lu Z, Zhang Y, Yan S, Zhao F. Multiparametric magnetic resonance imaging (MRI)-based radiomics model explained by the Shapley Additive exPlanations (SHAP) method for predicting complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: a multicenter retrospective study. Quant Imaging Med Surg 2024; 14:4617-4634. [PMID: 39022292 PMCID: PMC11250347 DOI: 10.21037/qims-24-7] [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: 01/02/2024] [Accepted: 04/09/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Predicting the response to neoadjuvant chemoradiotherapy (nCRT) before initiating treatment is essential for tailoring therapeutic strategies and monitoring prognosis in locally advanced rectal cancer (LARC). In this study, we aimed to develop and validate radiomic-based models to predict clinical and pathological complete responses (cCR and pCR, respectively) by incorporating the Shapley Additive exPlanations (SHAP) method for model interpretation. METHODS A total of 285 patients with complete pretreatment clinical characteristics and T1-weighted (T1W) and T2-weighted (T2W) magnetic resonance imaging (MRI) at 3 centers were retrospectively recruited. The features of tumor lesions were extracted by PyRadiomics and selected using least absolute shrinkage and selection operator (LASSO) algorithm. The selected features were used to build multilayer perceptron (MLP) models alone or combined with clinical features. Area under the receiver operating characteristic curve (AUC), decision curve, and calibration curve were applied to evaluate performance of models. The SHAP method was adopted to explain the prediction models. RESULTS The radiomic-based models all showed better performances than clinical models. The clinical-radiomic models showed the best differentiation on cCR and pCR with mean AUCs of 0.718 and 0.810 in the validation set, respectively. The decision curves of the clinical-radiomic models showed its values in clinical application. The SHAP method powerfully interpreted the prediction models both at a holistic and individual levels. CONCLUSIONS Our study highlights that the radiomic-based prediction models have more excellent abilities than clinical models and can effectively predict treatment response and optimize therapeutic strategies for patients with LARCs.
Collapse
Affiliation(s)
- Yiqi Wang
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Graduate School, Zhejiang University School of Medicine, Hangzhou, China
| | - Luyuan Zhang
- Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanting Jiang
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Graduate School, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaofei Cheng
- Department of Colorectal Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenguang He
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haogang Yu
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xinke Li
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jing Yang
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Guorong Yao
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhongjie Lu
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yi Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Senxiang Yan
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Feng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| |
Collapse
|
37
|
Song J, Zhang J, Liu G, Guo Z, Liao H, Feng W, Lin W, Li L, Zhang Y, Yang Y, Liu B, Luo R, Chen H, Wang S, Liu JH. PET/CT deep learning prognosis for treatment decision support in esophageal squamous cell carcinoma. Insights Imaging 2024; 15:161. [PMID: 38913225 PMCID: PMC11196479 DOI: 10.1186/s13244-024-01737-1] [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: 01/06/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVES The clinical decision-making regarding choosing surgery alone (SA) or surgery followed by postoperative adjuvant chemotherapy (SPOCT) in esophageal squamous cell carcinoma (ESCC) remains controversial. We aim to propose a pre-therapy PET/CT image-based deep learning approach to improve the survival benefit and clinical management of ESCC patients. METHODS This retrospective multicenter study included 837 ESCC patients from three institutions. Prognostic biomarkers integrating six networks were developed to build an ESCC prognosis (ESCCPro) model and predict the survival probability of ESCC patients treated with SA and SPOCT. Patients who did not undergo surgical resection were in a control group. Overall survival (OS) was the primary end-point event. The expected improvement in survival prognosis with the application of ESCCPro to assign treatment protocols was estimated by comparing the survival of patients in each subgroup. Seven clinicians with varying experience evaluated how ESCCPro performed in assisting clinical decision-making. RESULTS In this retrospective multicenter study, patients receiving SA had a median OS 9.2 months longer than controls. No significant differences in survival were found between SA patients with predicted poor outcomes and the controls (p > 0.05). It was estimated that if ESCCPro was used to determine SA and SPOCT eligibility, the median OS in the ESCCPro-recommended SA group and SPOCT group would have been 15.3 months and 24.9 months longer, respectively. In addition, ESCCPro also significantly improved prognosis accuracy, certainty, and the efficiency of clinical experts. CONCLUSION ESCCPro assistance improved the survival benefit of ESCC patients and the clinical decision-making among the two treatment approaches. CRITICAL RELEVANCE STATEMENT The ESCCPro model for treatment decision-making is promising to improve overall survival in ESCC patients undergoing surgical resection and patients undergoing surgery followed by postoperative adjuvant chemotherapy. KEY POINTS ESCC is associated with a poor prognosis and unclear ideal treatments. ESCCPro predicts the survival of patients with ESCC and the expected benefit from SA. ESCCPro improves clinicians' stratification of patients' prognoses.
Collapse
Affiliation(s)
- Jiangdian Song
- School of Health Management, China Medical University, Shenyang, China.
| | - Jie Zhang
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Guichao Liu
- The Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
| | - Zhexu Guo
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors China Medical University, Ministry of Education, Shenyang, China
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, China
| | - Hongxian Liao
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Wenhui Feng
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Wenxiang Lin
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Lei Li
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yi Zhang
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yuxiang Yang
- Department of Radiology, The Second People's Hospital of Xiangzhou, Zhuhai, China
| | - Bin Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, Hong Kong
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Siyun Wang
- Department of Oncology, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Jian-Hua Liu
- Department of Oncology, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Xu N, Wang J, Dai G, Lu T, Li S, Deng K, Song J. EfficientNet-Based System for Detecting EGFR-Mutant Status and Predicting Prognosis of Tyrosine Kinase Inhibitors in Patients with NSCLC. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1086-1099. [PMID: 38361006 PMCID: PMC11169294 DOI: 10.1007/s10278-024-01022-z] [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: 09/29/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
We aimed to develop and validate a deep learning-based system using pre-therapy computed tomography (CT) images to detect epidermal growth factor receptor (EGFR)-mutant status in patients with non-small cell lung cancer (NSCLC) and predict the prognosis of advanced-stage patients with EGFR mutations treated with EGFR tyrosine kinase inhibitors (TKI). This retrospective, multicenter study included 485 patients with NSCLC from four hospitals. Of them, 339 patients from three centers were included in the training dataset to develop an EfficientNetV2-L-based model (EME) for predicting EGFR-mutant status, and the remaining patients were assigned to an independent test dataset. EME semantic features were extracted to construct an EME-prognostic model to stratify the prognosis of EGFR-mutant NSCLC patients receiving EGFR-TKI. A comparison of EME and radiomics was conducted. Additionally, we included patients from The Cancer Genome Atlas lung adenocarcinoma dataset with both CT images and RNA sequencing data to explore the biological associations between EME score and EGFR-related biological processes. EME obtained an area under the curve (AUC) of 0.907 (95% CI 0.840-0.926) on the test dataset, superior to the radiomics model (P = 0.007). The EME and radiomics fusion model showed better (AUC, 0.941) but not significantly increased performance (P = 0.895) compared with EME. In prognostic stratification, the EME-prognostic model achieved the best performance (C-index, 0.711). Moreover, the EME-prognostic score showed strong associations with biological pathways related to EGFR expression and EGFR-TKI efficacy. EME demonstrated a non-invasive and biologically interpretable approach to predict EGFR status, stratify survival prognosis, and correlate biological pathways in patients with NSCLC.
Collapse
Affiliation(s)
- Nan Xu
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jiajun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Gang Dai
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, Anhui, 230036, China
| | - Tao Lu
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Shu Li
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China
| | - Kexue Deng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, Anhui, 230036, China
| | - Jiangdian Song
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
39
|
Zhang H, Xu R, Guo X, Zhou D, Xu T, Zhong X, Kong M, Zhang Z, Wang Y, Ma X. Deep learning-based automated high-accuracy location and identification of fresh vertebral compression fractures from spinal radiographs: a multicenter cohort study. Front Bioeng Biotechnol 2024; 12:1397003. [PMID: 38812917 PMCID: PMC11135169 DOI: 10.3389/fbioe.2024.1397003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Background Digital radiography (DR) is a common and widely available examination. However, spinal DR cannot detect bone marrow edema, therefore, determining vertebral compression fractures (VCFs), especially fresh VCFs, remains challenging for clinicians. Methods We trained, validated, and externally tested the deep residual network (DRN) model that automated the detection and identification of fresh VCFs from spinal DR images. A total of 1,747 participants from five institutions were enrolled in this study and divided into the training cohort, validation cohort and external test cohorts (YHDH and BMUH cohorts). We evaluated the performance of DRN model based on the area under the receiver operating characteristic curve (AUC), feature attention maps, sensitivity, specificity, and accuracy. We compared it with five other deep learning models and validated and tested the model internally and externally and explored whether it remains highly accurate for an external test cohort. In addition, the influence of old VCFs on the performance of the DRN model was assessed. Results The AUC was 0.99, 0.89, and 0.88 in the validation, YHDH, and BMUH cohorts, respectively, for the DRN model for detecting and discriminating fresh VCFs. The accuracies were 81.45% and 72.90%, sensitivities were 84.75% and 91.43%, and specificities were 80.25% and 63.89% in the YHDH and BMUH cohorts, respectively. The DRN model generated correct activation on the fresh VCFs and accurate peak responses on the area of the target vertebral body parts and demonstrated better feature representation learning and classification performance. The AUC was 0.90 (95% confidence interval [CI] 0.84-0.95) and 0.84 (95% CI 0.72-0.93) in the non-old VCFs and old VCFs groups, respectively, in the YHDH cohort (p = 0.067). The AUC was 0.89 (95% CI 0.84-0.94) and 0.85 (95% CI 0.72-0.95) in the non-old VCFs and old VCFs groups, respectively, in the BMUH cohort (p = 0.051). Conclusion In present study, we developed the DRN model for automated diagnosis and identification of fresh VCFs from spinal DR images. The DRN model can provide interpretable attention maps to support the excellent prediction results, which is the key that most clinicians care about when using the model to assist decision-making.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ruixiang Xu
- Department of Pain, YanTai YuHuangDing Hospital, Yantai, Shandong, China
| | - Xiang Guo
- Department of Spinal Surgery, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Dan Zhou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tongshuai Xu
- Department of Spinal Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xin Zhong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Meng Kong
- Department of Spinal Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zhimin Zhang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Wang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xuexiao Ma
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
40
|
Batra U, Nathany S, Nath SK, Jose JT, Sharma T, P P, Pasricha S, Sharma M, Arambam N, Khanna V, Bansal A, Mehta A, Rawal K. AI-based pipeline for early screening of lung cancer: integrating radiology, clinical, and genomics data. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2024; 24:100352. [PMID: 38756151 PMCID: PMC11096686 DOI: 10.1016/j.lansea.2024.100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 12/11/2023] [Accepted: 01/04/2024] [Indexed: 05/18/2024]
Abstract
Background The prognosis of lung carcinoma has changed since the discovery of molecular targets and their specific drugs. Somatic Epidermal Growth Factor Receptor (EGFR) mutations have been reported in lung carcinoma, and these mutant proteins act as substrates for targeted therapies. However, in a resource-constrained country like India, panel-based next-generation sequencing cannot be made available to the population at large. Additional challenges such as adequacy of tissue in case of lung core biopsies and locating suitable tumour tissues as a result of innate intratumoral heterogeneity indicate the necessity of an AI-based end-to-end pipeline capable of automatically detecting and learning more effective lung nodule features from CT images and predicting the probability of the EGFR-mutant. This will help the oncologists and patients in resource-limited settings to achieve near-optimal care and appropriate therapy. Methods The EGFR gene sequencing and CT imaging data of 2277 patients with lung carcinoma were included from three cohorts in India and a White population cohort collected from TCIA. Another cohort LIDC-IDRI was used to train the AIPS-Nodule (AIPS-N) model for automatic detection and characterisation of lung nodules. We explored the value of combining the results of the AIPS-N with the clinical factors in the AIPS-Mutation (AIPS-M) model for predicting EGFR genotype, and it was evaluated by area under the curve (AUC). Findings AIPS-N achieved an average AP50 of 70.19% in detecting the location of nodules within the lung region of interest during validation and predicted the score of five lung nodule properties. The AIPS-M machine learning (ML) and deep learning (DL) models achieved AUCs ranging from 0.587 to 0.910. Interpretation The AIPS suggests that CT imaging combined with a fully automated lung-nodule analysis AI system can predict EGFR genotype and identify patients with an EGFR mutation in a cost-effective and non-invasive manner. Funding This work was supported by a grant provided by Conquer Cancer Foundation of ASCO [2021IIG-5555960128] and Pfizer Products India Pvt. Ltd.
Collapse
Affiliation(s)
- Ullas Batra
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | | | | | - Joslia T. Jose
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Trapti Sharma
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Preeti P
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Sunil Pasricha
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Mansi Sharma
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Nevidita Arambam
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Vrinda Khanna
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Abhishek Bansal
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Anurag Mehta
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Kamal Rawal
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
41
|
Li F, Qi L, Cheng S, Liu J, Chen J, Cui S, Dong S, Wang J. Predicting epidermal growth factor receptor mutations in non-small cell lung cancer through dual-layer spectral CT: a prospective study. Insights Imaging 2024; 15:109. [PMID: 38679659 PMCID: PMC11056350 DOI: 10.1186/s13244-024-01678-9] [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: 09/25/2023] [Accepted: 03/22/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVE To determine whether quantitative parameters of detector-derived dual-layer spectral computed tomography (DLCT) can reliably identify epidermal growth factor receptor (EGFR) mutation status in patients with non-small cell lung cancer (NSCLC). METHODS Patients with NSCLC who underwent arterial phase (AP) and venous phase (VP) DLCT between December 2021 and November 2022 were subdivided into the mutated and wild-type EGFR groups following EGFR mutation testing. Their baseline clinical data, conventional CT images, and spectral images were obtained. Iodine concentration (IC), iodine no water (INW), effective atomic number (Zeff), virtual monoenergetic images, the slope of the spectral attenuation curve (λHU), enhancement degree (ED), arterial enhancement fraction (AEF), and normalized AEF (NAEF) were measured for each lesion. RESULTS Ninety-two patients (median age, 61 years, interquartile range [51, 67]; 33 men) were evaluated. The univariate analysis indicated that IC, normalized IC (NIC), INW and ED for the AP and VP, as well as Zeff and λHU for the VP were significantly associated with EGFR mutation status (all p < 0.05). INW(VP) showed the best diagnostic performance (AUC, 0.892 [95% confidence interval {CI}: 0.823, 0.960]). However, neither AEF (p = 0.156) nor NAEF (p = 0.567) showed significant differences between the two groups. The multivariate analysis showed that INW(AP) and NIC(VP) were significant predictors of EGFR mutation status, with the latter showing better performance (p = 0.029; AUC, 0.897 [95% CI: 0.816, 0.951] vs. 0.774 [95% CI: 0.675, 0.855]). CONCLUSION Quantitative parameters of DLCT can help predict EGFR mutation status in patients with NSCLC. CRITICAL RELEVANCE STATEMENT Quantitative parameters of DLCT, especially NIC(VP), can help predict EGFR mutation status in patients with NSCLC, facilitating appropriate and individualized treatment for them. KEY POINTS Determining EGFR mutation status in patients with NSCLC before starting therapy is essential. Quantitative parameters of DLCT can predict EGFR mutation status in NSCLC patients. NIC in venous phase is an important parameter to guide individualized treatment selection for NSCLC patients.
Collapse
Affiliation(s)
- Fenglan Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Linlin Qi
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Sainan Cheng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Jianing Liu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Jiaqi Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Shulei Cui
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Shushan Dong
- Clinical Science, Philips Healthcare, Beijing, China
| | - Jianwei Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China.
| |
Collapse
|
42
|
Lou N, Cui X, Lin X, Gao R, Xu C, Qiao N, Jiang J, Wang L, Wang W, Wang S, Shen W, Zheng X, Han X. Development and validation of a deep learning-based model to predict response and survival of T790M mutant non-small cell lung cancer patients in early clinical phase trials using electronic medical record and pharmacokinetic data. Transl Lung Cancer Res 2024; 13:706-720. [PMID: 38736496 PMCID: PMC11082707 DOI: 10.21037/tlcr-23-737] [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: 11/12/2023] [Accepted: 03/15/2024] [Indexed: 05/14/2024]
Abstract
Background Epidermal growth factor receptor (EGFR) T790M mutation is the standard predictive biomarker for third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) treatment. While not all T790M-positive patients respond to third-generation EGFR-TKIs and have a good prognosis, it necessitates novel tools to supplement EGFR genotype detection for predicting efficacy and stratifying EGFR-mutant patients with various prognoses. Mixture-of-experts (MoE) is designed to disassemble a large model into many small models. Meanwhile, it is also a model ensembling method that can better capture multiple patterns of intrinsic subgroups of enrolled patients. Therefore, the combination of MoE and Cox algorithm has the potential to predict efficacy and stratify survival in non-small cell lung cancer (NSCLC) patients with EGFR mutations. Methods We utilized the electronic medical record (EMR) and pharmacokinetic parameters of 326 T790M-mutated NSCLC patients, including 283 patients treated with Abivertinib in phase I (n=177, for training) and II (n=106, for validation) clinical trials and an additional validation cohort 2 comprising 43 patients treated with BPI-7711. Furthermore, 18 patients underwent whole-exome sequencing for biological interpretation of CoxMoE. We evaluated the predictive performance for therapeutic response using the area under the curve (AUC) and the Concordance index (C-index) for progression-free survival (PFS). Results CoxMoE exhibited AUCs of 0.73-0.83 for predicting efficacy defined by best overall response (BoR) and achieved C-index values of 0.64-0.65 for PFS prediction in training and validating cohorts. The PFS of 198 patients with a low risk [median, 6.0 (range, 1.0-23.3) months in the abivertinib treated cohort; median 16.5 (range, 1.4-27.4) months in BPI-7711 treated cohort] of being non-responder increased by 43% [hazard ratio (HR), 0.56; 95% confidence interval (CI), 0.40-0.78; P=0.0013] and 50% (HR, 0; 95% CI, 0-0; P=0.01) compared to those at high-risk [median, 4.2 (range, 1.0-35) months in the abivertinib treated cohort; median, 11.0 (range, 1.4-25.1) months in BPI-7711 treated cohort]. Additionally, activated partial thromboplastin time (APTT), creatinine clearance (Ccr), monocyte, and steady-state plasma trough concentration utilited to construct model were found significantly associated with drug resistance and aggressive tumor pathways. A robust correlation was observed between APTT and Ccr with PFS (log-rank test; P<0.01) and treatment response (Wilcoxon test; P<0.05), respectively. Conclusions CoxMoE offers a valuable approach for patient selection by forecasting therapeutic response and PFS utilizing laboratory tests and pharmacokinetic parameters in the setting of early-phase clinical trials. Simultaneously, CoxMoE could predict the efficacy of third-generation EGFR-TKI non-invasively for T790M-positive NSCLC patients, thereby complementing existing EGFR genotype detection.
Collapse
Affiliation(s)
- Ning Lou
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinge Cui
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyuan Lin
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, China
| | - Ruyun Gao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Chi Xu
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, China
| | - Nan Qiao
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, China
| | - Ji Jiang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lu Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Weicong Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shanbo Wang
- Hangzhou ACEA Pharmaceutical Research Co., Ltd., Hangzhou, China
| | - Wei Shen
- Hangzhou ACEA Pharmaceutical Research Co., Ltd., Hangzhou, China
| | - Xin Zheng
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
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
|
44
|
Louis T, Lucia F, Cousin F, Mievis C, Jansen N, Duysinx B, Le Pennec R, Visvikis D, Nebbache M, Rehn M, Hamya M, Geier M, Salaun PY, Schick U, Hatt M, Coucke P, Lovinfosse P, Hustinx R. Identification of CT radiomic features robust to acquisition and segmentation variations for improved prediction of radiotherapy-treated lung cancer patient recurrence. Sci Rep 2024; 14:9028. [PMID: 38641673 PMCID: PMC11031577 DOI: 10.1038/s41598-024-58551-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 04/21/2024] Open
Abstract
The primary objective of the present study was to identify a subset of radiomic features extracted from primary tumor imaged by computed tomography of early-stage non-small cell lung cancer patients, which remain unaffected by variations in segmentation quality and in computed tomography image acquisition protocol. The robustness of these features to segmentation variations was assessed by analyzing the correlation of feature values extracted from lesion volumes delineated by two annotators. The robustness to variations in acquisition protocol was evaluated by examining the correlation of features extracted from high-dose and low-dose computed tomography scans, both of which were acquired for each patient as part of the stereotactic body radiotherapy planning process. Among 106 radiomic features considered, 21 were identified as robust. An analysis including univariate and multivariate assessments was subsequently conducted to estimate the predictive performance of these robust features on the outcome of early-stage non-small cell lung cancer patients treated with stereotactic body radiation therapy. The univariate predictive analysis revealed that robust features demonstrated superior predictive potential compared to non-robust features. The multivariate analysis indicated that linear regression models built with robust features displayed greater generalization capabilities by outperforming other models in predicting the outcomes of an external validation dataset.
Collapse
Affiliation(s)
- Thomas Louis
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
| | - François Lucia
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
- Radiation Oncology Department, University Hospital of Brest, Brest, France.
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France.
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Carole Mievis
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Nicolas Jansen
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Bernard Duysinx
- Division of Pulmonology, University Hospital of Liège, Liège, Belgium
| | - Romain Le Pennec
- Nuclear Medicine Department, University Hospital of Brest, Brest, France
- GETBO INSERM UMR 1304, University of Brest, UBO, Brest, France
| | | | - Malik Nebbache
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Martin Rehn
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Mohamed Hamya
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Margaux Geier
- Medical Oncology Department, University Hospital of Brest, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospital of Brest, Brest, France
- GETBO INSERM UMR 1304, University of Brest, UBO, Brest, France
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital of Brest, Brest, France
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France
| | - Philippe Coucke
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
45
|
Sujit SJ, Aminu M, Karpinets TV, Chen P, Saad MB, Salehjahromi M, Boom JD, Qayati M, George JM, Allen H, Antonoff MB, Hong L, Hu X, Heeke S, Tran HT, Le X, Elamin YY, Altan M, Vokes NI, Sheshadri A, Lin J, Zhang J, Lu Y, Behrens C, Godoy MCB, Wu CC, Chang JY, Chung C, Jaffray DA, Wistuba II, Lee JJ, Vaporciyan AA, Gibbons DL, Heymach J, Zhang J, Cascone T, Wu J. Enhancing NSCLC recurrence prediction with PET/CT habitat imaging, ctDNA, and integrative radiogenomics-blood insights. Nat Commun 2024; 15:3152. [PMID: 38605064 PMCID: PMC11009351 DOI: 10.1038/s41467-024-47512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
While we recognize the prognostic importance of clinicopathological measures and circulating tumor DNA (ctDNA), the independent contribution of quantitative image markers to prognosis in non-small cell lung cancer (NSCLC) remains underexplored. In our multi-institutional study of 394 NSCLC patients, we utilize pre-treatment computed tomography (CT) and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) to establish a habitat imaging framework for assessing regional heterogeneity within individual tumors. This framework identifies three PET/CT subtypes, which maintain prognostic value after adjusting for clinicopathologic risk factors including tumor volume. Additionally, these subtypes complement ctDNA in predicting disease recurrence. Radiogenomics analysis unveil the molecular underpinnings of these imaging subtypes, highlighting downregulation in interferon alpha and gamma pathways in the high-risk subtype. In summary, our study demonstrates that these habitat imaging subtypes effectively stratify NSCLC patients based on their risk levels for disease recurrence after initial curative surgery or radiotherapy, providing valuable insights for personalized treatment approaches.
Collapse
Affiliation(s)
- Sheeba J Sujit
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tatiana V Karpinets
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maliazurina B Saad
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Morteza Salehjahromi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John D Boom
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mohamed Qayati
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M George
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Haley Allen
- Natural Sciences, Rice University, Houston, TX, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingzhi Hong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Hu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hai T Tran
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie Lin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Jaffray
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Genomics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Interception Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
46
|
Shao X, Ge X, Gao J, Niu R, Shi Y, Shao X, Jiang Z, Li R, Wang Y. Transfer learning-based PET/CT three-dimensional convolutional neural network fusion of image and clinical information for prediction of EGFR mutation in lung adenocarcinoma. BMC Med Imaging 2024; 24:54. [PMID: 38438844 PMCID: PMC10913633 DOI: 10.1186/s12880-024-01232-5] [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: 11/14/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND To introduce a three-dimensional convolutional neural network (3D CNN) leveraging transfer learning for fusing PET/CT images and clinical data to predict EGFR mutation status in lung adenocarcinoma (LADC). METHODS Retrospective data from 516 LADC patients, encompassing preoperative PET/CT images, clinical information, and EGFR mutation status, were divided into training (n = 404) and test sets (n = 112). Several deep learning models were developed utilizing transfer learning, involving CT-only and PET-only models. A dual-stream model fusing PET and CT and a three-stream transfer learning model (TS_TL) integrating clinical data were also developed. Image preprocessing includes semi-automatic segmentation, resampling, and image cropping. Considering the impact of class imbalance, the performance of the model was evaluated using ROC curves and AUC values. RESULTS TS_TL model demonstrated promising performance in predicting the EGFR mutation status, with an AUC of 0.883 (95%CI = 0.849-0.917) in the training set and 0.730 (95%CI = 0.629-0.830) in the independent test set. Particularly in advanced LADC, the model achieved an AUC of 0.871 (95%CI = 0.823-0.919) in the training set and 0.760 (95%CI = 0.638-0.881) in the test set. The model identified distinct activation areas in solid or subsolid lesions associated with wild and mutant types. Additionally, the patterns captured by the model were significantly altered by effective tyrosine kinase inhibitors treatment, leading to notable changes in predicted mutation probabilities. CONCLUSION PET/CT deep learning model can act as a tool for predicting EGFR mutation in LADC. Additionally, it offers clinicians insights for treatment decisions through evaluations both before and after treatment.
Collapse
Affiliation(s)
- Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China.
| | - Xinyu Ge
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Jianxiong Gao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Rong Niu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Yunmei Shi
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Zhenxing Jiang
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Renyuan Li
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China.
| |
Collapse
|
47
|
Ruan D, Fang J, Teng X. Efficient 18F-fluorodeoxyglucose positron emission tomography/computed tomography-based machine learning model for predicting epidermal growth factor receptor mutations in non-small cell lung cancer. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2024; 68:70-83. [PMID: 35420272 DOI: 10.23736/s1824-4785.22.03441-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Beyond the human eye's limitations, radiomics provides more information that can be used for diagnosis. We develop a personalized and efficient model based on 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) to predict epidermal growth factor receptor (EGFR) mutations to help identify which non-small cell cancer (NSCLC) patients are candidates for EGFR-tyrosine kinase inhibitors (TKIs) therapy. METHODS We retrospectively included 100 patients with NSCLC and randomized them according to 70 patients in the training group and 30 patients in the validation group. The least absolute shrinkage and selection operator logistic regression (LLR) algorithm and support vector machine (SVM) classifier were used to build the models and predict whether EGFR is mutated or not. The predictive efficacy of the LLR algorithm-based model and the SVM classifier-based model was evaluated by plotting the receiver operating characteristic (ROC) curves and calculating the area under the curve (AUC). RESULTS The AUC, sensitivity and specificity of our radiomics model by LLR algorithm were 0.792, 0.967, and 0.600 for the training group and 0.643, 1.00, and 0.378 for the validation group, respectively, in predicting EGFR mutations. The AUC was 0.838 for the training group and 0.696 for the validation group after combining radiomics features with clinical features. The prediction results based on the SVM classifier showed that the validation group had the best performance when based on radial kernel function with AUC, sensitivity, and specificity of 0.741, 0.667, and 0.825, respectively. CONCLUSIONS Radiomics models based on 18F-FDG PET/CT modeled with different machine learning algorithms can improve the predictive efficacy of the models. Models that combine clinical features are more clinically valuable.
Collapse
Affiliation(s)
- Dan Ruan
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China -
| | - Janyao Fang
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| | - Xinyu Teng
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| |
Collapse
|
48
|
Abbaker N, Minervini F, Guttadauro A, Solli P, Cioffi U, Scarci M. The future of artificial intelligence in thoracic surgery for non-small cell lung cancer treatment a narrative review. Front Oncol 2024; 14:1347464. [PMID: 38414748 PMCID: PMC10897973 DOI: 10.3389/fonc.2024.1347464] [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: 11/30/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECTIVES To present a comprehensive review of the current state of artificial intelligence (AI) applications in lung cancer management, spanning the preoperative, intraoperative, and postoperative phases. METHODS A review of the literature was conducted using PubMed, EMBASE and Cochrane, including relevant studies between 2002 and 2023 to identify the latest research on artificial intelligence and lung cancer. CONCLUSION While AI holds promise in managing lung cancer, challenges exist. In the preoperative phase, AI can improve diagnostics and predict biomarkers, particularly in cases with limited biopsy materials. During surgery, AI provides real-time guidance. Postoperatively, AI assists in pathology assessment and predictive modeling. Challenges include interpretability issues, training limitations affecting model use and AI's ineffectiveness beyond classification. Overfitting and global generalization, along with high computational costs and ethical frameworks, pose hurdles. Addressing these challenges requires a careful approach, considering ethical, technical, and regulatory factors. Rigorous analysis, external validation, and a robust regulatory framework are crucial for responsible AI implementation in lung surgery, reflecting the evolving synergy between human expertise and technology.
Collapse
Affiliation(s)
- Namariq Abbaker
- Division of Thoracic Surgery, Imperial College NHS Healthcare Trust and National Heart and Lung Institute, London, United Kingdom
| | - Fabrizio Minervini
- Division of Thoracic Surgery, Luzerner Kantonsspital, Lucern, Switzerland
| | - Angelo Guttadauro
- Division of Surgery, Università Milano-Bicocca and Istituti Clinici Zucchi, Monza, Italy
| | - Piergiorgio Solli
- Division of Thoracic Surgery, Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Ugo Cioffi
- Department of Surgery, University of Milan, Milan, Italy
| | - Marco Scarci
- Division of Thoracic Surgery, Imperial College NHS Healthcare Trust and National Heart and Lung Institute, London, United Kingdom
| |
Collapse
|
49
|
Ma N, Yang W, Wang Q, Cui C, Hu Y, Wu Z. Predictive value of 18F-FDG PET/CT radiomics for EGFR mutation status in non-small cell lung cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1281572. [PMID: 38361781 PMCID: PMC10867100 DOI: 10.3389/fonc.2024.1281572] [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: 08/22/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Objective This study aimed to evaluate the value of 18F-FDG PET/CT radiomics in predicting EGFR gene mutations in non-small cell lung cancer by meta-analysis. Methods The PubMed, Embase, Cochrane Library, Web of Science, and CNKI databases were searched from the earliest available date to June 30, 2023. The meta-analysis was performed using the Stata 15.0 software. The methodological quality and risk of bias of included studies were assessed using the Quality Assessment of Diagnostic Accuracy Studies 2 and Radiomics Quality Score criteria. The possible causes of heterogeneity were analyzed by meta-regression. Results A total of 17 studies involving 3763 non-small cell lung cancer patients were finally included. We analyzed 17 training cohorts and 10 validation cohorts independently. Within the training cohort, the application of 18F-FDG PET/CT radiomics in predicting EGFR mutations in NSCLC demonstrated a sensitivity of 0.76 (95% CI: 0.70-0.81) and a specificity of 0.78 (95% CI: 0.74-0.82), accompanied by a positive likelihood ratio of 3.5 (95% CI:3.0-4.2), a negative likelihood ratio of 0.31 (95% CI: 0.24-0.39), a diagnostic odds ratio of 11.0 (95% CI: 8.0-16.0), and an area under the curve (AUC) of 0.84 (95% CI: 0.80-0.87). In the validation cohort, the values included a sensitivity of 0.76 (95% CI: 0.67-0.83), a specificity of 0.75 (95% CI: 0.68-0.80), a positive likelihood ratio of 3.0 (95% CI:2.4-3.8), a negative likelihood ratio of 0.32 (95% CI: 0.24-0.44), a diagnostic odds ratio of 9 (95% CI: 6-15), and an AUC of 0.82 (95% CI: 0.78-0.85). The average Radiomics Quality Score (RQS) across studies was 10.47 ± 4.72. Meta-regression analysis identifies the application of deep learning and regions as sources of heterogeneity. Conclusion 18F-FDG PET/CT radiomics may be useful in predicting mutation status of the EGFR gene in non-small cell lung cancer. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42022385364.
Collapse
Affiliation(s)
- Ning Ma
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Weihua Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiannan Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Caozhe Cui
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yiyi Hu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
50
|
Nguyen HS, Ho DKN, Nguyen NN, Tran HM, Tam KW, Le NQK. Predicting EGFR Mutation Status in Non-Small Cell Lung Cancer Using Artificial Intelligence: A Systematic Review and Meta-Analysis. Acad Radiol 2024; 31:660-683. [PMID: 37120403 DOI: 10.1016/j.acra.2023.03.040] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023]
Abstract
RATIONALE AND OBJECTIVES Recent advancements in artificial intelligence (AI) render a substantial promise for epidermal growth factor receptor (EGFR) mutation status prediction in non-small cell lung cancer (NSCLC). We aimed to evaluate the performance and quality of AI algorithms that use radiomics features in predicting EGFR mutation status in patient with NSCLC. MATERIALS AND METHODS We searched PubMed (Medline), EMBASE, Web of Science, and IEEExplore for studies published up to February 28, 2022. Studies utilizing an AI algorithm (either conventional machine learning [cML] and deep learning [DL]) for predicting EGFR mutations in patients with NSLCL were included. We extracted binary diagnostic accuracy data and constructed a bivariate random-effects model to obtain pooled sensitivity, specificity, and 95% confidence interval. This study is registered with PROSPERO, CRD42021278738. RESULTS Our search identified 460 studies, of which 42 were included. Thirty-five studies were included in the meta-analysis. The AI algorithms exhibited an overall area under the curve (AUC) value of 0.789 and pooled sensitivity and specificity levels of 72.2% and 73.3%, respectively. The DL algorithms outperformed cML in terms of AUC (0.822 vs. 0.775) and sensitivity (80.1% vs. 71.1%), but had lower specificity (70.0% vs. 73.8%, p-value < 0.001) compared to cML. Subgroup analysis revealed that the use of positron-emission tomography/computed tomography, additional clinical information, deep feature extraction, and manual segmentation can improve diagnostic performance. CONCLUSION DL algorithms can serve as a novel method for increasing predictive accuracy and thus have considerable potential for use in predicting EGFR mutation status in patient with NSCLC. We also suggest that guidelines on using AI algorithms in medical image analysis should be developed with a focus on oncologic radiomics.
Collapse
Affiliation(s)
- Hung Song Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (H.S.N., N.N.N.); Department of Pediatrics, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Viet Nam (H.S.N.); Intensive Care Unit Department, Children's Hospital 1, Ho Chi Minh City, Viet Nam (H.S.N.)
| | - Dang Khanh Ngan Ho
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan (D.K.N.H.)
| | - Nam Nhat Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (H.S.N., N.N.N.)
| | - Huy Minh Tran
- Department of Neurosurgery, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam (H.M.T.)
| | - Ka-Wai Tam
- Center for Evidence-based Health Care, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (K.-W.T.); Cochrane Taiwan, Taipei Medical University, Taipei City, Taiwan (K.-W.T.); Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (K.-W.T.); Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (K.-W.T.)
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan (N.Q.K.L.).
| |
Collapse
|