1
|
Tanaka T, Motegi T, Sumikawa N, Mori M, Kurokawa S, Akiyoshi H. Early Enhancement in Contrast-Enhanced Computed Tomography Is an Index of DUSP9, SLPI, ALDH1L2, and SLC1A1 Expression in Canine Hepatocellular Carcinoma: A Preliminary Study. Vet Sci 2025; 12:137. [PMID: 40005897 PMCID: PMC11860268 DOI: 10.3390/vetsci12020137] [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: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Canine hepatocellular carcinoma (HCC) is characterized by distinct computed tomography (CT) findings. HCC exhibits tumor heterogeneity, with different genomic information and histopathological features within the same tumor. In human HCC, genetic alterations affect the prognosis and treatment, and research has begun to assess genetic alterations using minimally invasive and reproducible CT. However, the relationship between CT findings and the genomic information of canine HCC is unknown. Early contrast of HCC indicates increased intratumoral neovascular growth. In this study, we aimed to investigate the relationship between enhancement patterns in the arterial phase of CT imaging and gene expression in canine HCC using RNA sequencing. Based on the CT findings, three of the eight dogs studied were classified as having enhancement HCC and five as having non-enhancement HCC. RNA sequencing was performed using the mRNA extracted from the specimens. Eight differentially expressed genes met the cutoff criteria. Among these, DUSP9, SLPI, and ALDH1L2 were the most upregulated genes in enhancement HCC, whereas SLC1A1 was the most downregulated in non-enhancement HCC. Canine HCC may involve different angiogenesis mechanisms. CT findings can be used to assess the gene expression status in canine HCC and may add new value to CT imaging.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| | - Tomoki Motegi
- Section of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Nanami Sumikawa
- Laboratory of Veterinary Surgery, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (N.S.); (M.M.)
| | - Misaki Mori
- Laboratory of Veterinary Surgery, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (N.S.); (M.M.)
| | - Shohei Kurokawa
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| | - Hideo Akiyoshi
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| |
Collapse
|
2
|
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
|
3
|
Wang FA, Li Y, Zeng T. Deep Learning of radiology-genomics integration for computational oncology: A mini review. Comput Struct Biotechnol J 2024; 23:2708-2716. [PMID: 39035833 PMCID: PMC11260400 DOI: 10.1016/j.csbj.2024.06.019] [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: 03/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
In the field of computational oncology, patient status is often assessed using radiology-genomics, which includes two key technologies and data, such as radiology and genomics. Recent advances in deep learning have facilitated the integration of radiology-genomics data, and even new omics data, significantly improving the robustness and accuracy of clinical predictions. These factors are driving artificial intelligence (AI) closer to practical clinical applications. In particular, deep learning models are crucial in identifying new radiology-genomics biomarkers and therapeutic targets, supported by explainable AI (xAI) methods. This review focuses on recent developments in deep learning for radiology-genomics integration, highlights current challenges, and outlines some research directions for multimodal integration and biomarker discovery of radiology-genomics or radiology-omics that are urgently needed in computational oncology.
Collapse
Affiliation(s)
- Feng-ao Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yixue Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Murmu A, Győrffy B. Artificial intelligence methods available for cancer research. Front Med 2024; 18:778-797. [PMID: 39115792 DOI: 10.1007/s11684-024-1085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/17/2024] [Indexed: 11/01/2024]
Abstract
Cancer is a heterogeneous and multifaceted disease with a significant global footprint. Despite substantial technological advancements for battling cancer, early diagnosis and selection of effective treatment remains a challenge. With the convenience of large-scale datasets including multiple levels of data, new bioinformatic tools are needed to transform this wealth of information into clinically useful decision-support tools. In this field, artificial intelligence (AI) technologies with their highly diverse applications are rapidly gaining ground. Machine learning methods, such as Bayesian networks, support vector machines, decision trees, random forests, gradient boosting, and K-nearest neighbors, including neural network models like deep learning, have proven valuable in predictive, prognostic, and diagnostic studies. Researchers have recently employed large language models to tackle new dimensions of problems. However, leveraging the opportunity to utilize AI in clinical settings will require surpassing significant obstacles-a major issue is the lack of use of the available reporting guidelines obstructing the reproducibility of published studies. In this review, we discuss the applications of AI methods and explore their benefits and limitations. We summarize the available guidelines for AI in healthcare and highlight the potential role and impact of AI models on future directions in cancer research.
Collapse
Affiliation(s)
- Ankita Murmu
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- National Laboratory for Drug Research and Development, Budapest, 1117, Hungary
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary
| | - Balázs Győrffy
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary.
- Department of Biophysics, University of Pecs, Pecs, 7624, Hungary.
| |
Collapse
|
5
|
Hu Y, Geng Y, Wang H, Chen H, Wang Z, Fu L, Huang B, Jiang W. Improved Prediction of Epidermal Growth Factor Receptor Status by Combined Radiomics of Primary Nonsmall-Cell Lung Cancer and Distant Metastasis. J Comput Assist Tomogr 2024; 48:780-788. [PMID: 38498926 DOI: 10.1097/rct.0000000000001591] [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: 03/20/2024]
Abstract
OBJECTIVES This study aimed to investigate radiomics based on primary nonsmall-cell lung cancer (NSCLC) and distant metastases to predict epidermal growth factor receptor (EGFR) mutation status. METHODS A total of 290 patients (mean age, 58.21 ± 9.28) diagnosed with brain (BM, n = 150) or spinal bone metastasis (SM, n = 140) from primary NSCLC were enrolled as a primary cohort. An external validation cohort, consisting of 69 patients (mean age, 59.87 ± 7.23; BM, n = 36; SM, n = 33), was enrolled from another center. Thoracic computed tomography-based features were extracted from the primary tumor and peritumoral area and selected using the least absolute shrinkage and selection operator regression to build a radiomic signature (RS-primary). Contrast-enhanced magnetic resonance imaging-based features were calculated and selected from the BM and SM to build RS-BM and RS-SM, respectively. The RS-BM-Com and RS-SM-Com were developed by integrating the most important features from the primary tumor, BM, and SM. RESULTS Six computed tomography-based features showed high association with EGFR mutation status: 3 from intratumoral and 3 from peritumoral areas. By combination of features from primary tumor and metastases, the developed RS-BM-Com and RS-SM-Com performed well with areas under curve in the training (RS-BM-Com vs RS-BM, 0.936 vs 0.885, P = 0.177; RS-SM-Com vs RS-SM, 0.929 vs 0.843, P = 0.003), internal validation (RS-BM-Com vs RS-BM, 0.920 vs 0.858, P = 0.492; RS-SM-Com vs RS-SM, 0.896 vs 0.859, P = 0.379), and external validation (RS-BM-Com vs RS-BM, 0.882 vs 0.805, P = 0.263; RS-SM-Com vs RS-SM, 0.865 vs 0.816, P = 0.312) cohorts. CONCLUSIONS This study indicates that the accuracy of detecting EGFR mutations significantly enhanced in the presence of metastases in primary NSCLC. The established radiomic signatures from this approach may be useful as new predictors for patients with distant metastases.
Collapse
Affiliation(s)
- Yue Hu
- From the Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yikang Geng
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Liaoning
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang
| | - Zekun Wang
- Department of Medical Iconography, Liaoning Cancer Hospital & Institute, Liaoning
| | - Langyuan Fu
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Bo Huang
- Department of Pathology, Liaoning Cancer Hospital and Institute, Liaoning
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Liaoning Cancer Hospital and Institute, Liaoning, People's Republic of China
| |
Collapse
|
6
|
Chen LN, Keating C, Leb J, Saqi A, Shu CA. Unusual presentation of ROS1 rearranged metastatic non-small cell lung cancer. Respir Med Case Rep 2024; 51:102091. [PMID: 39257471 PMCID: PMC11386496 DOI: 10.1016/j.rmcr.2024.102091] [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: 05/08/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
The spectrum of clinical and radiographic presentations of lung adenocarcinoma is increasingly broad, including in the metastatic setting. Here, we report on a patient who initially presented with a mild chronic cough that remained stable over a decade, with serial CT scans showing gradual worsening of multifocal areas of consolidation and ground-glass opacities of the bilateral lungs. The patient was ultimately diagnosed with ROS1 rearranged lung adenocarcinoma and achieved a dramatic response with entrectinib. This case highlights the variable presentation of non-small cell lung cancer (NSCLC) and the importance of comprehensive molecular testing for newly diagnosed metastatic NSCLC.
Collapse
Affiliation(s)
- Lanyi Nora Chen
- Division of Hematology and Oncology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Claire Keating
- Division of Pulmonary Medicine, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Anjali Saqi
- Department of Pathology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Catherine A Shu
- Division of Hematology and Oncology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| |
Collapse
|
7
|
Chu Y, Zhang S, Wan W, Yang J, Zhang Y, Nie C, Xing W, Tong S, Liu J, Tian G, Wang B, Ji L. Pathological image profiling identifies onco-microbial, tumor immune microenvironment, and prognostic subtypes of colorectal cancer. APMIS 2024; 132:416-429. [PMID: 38403979 DOI: 10.1111/apm.13387] [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: 09/20/2023] [Accepted: 02/02/2024] [Indexed: 02/27/2024]
Abstract
Histology slide, tissue microbes, and the host gene expression can be independent prognostic factors of colorectal cancer (CRC), but the underlying associations and biological significance of these multimodal omics remain unknown. Here, we comprehensively profiled the matched pathological images, intratumoral microbes, and host gene expression characteristics in 527 patients with CRC. By clustering these patients based on histology slide features, we classified the patients into two histology slide subtypes (HSS). Onco-microbial community and tumor immune microenvironment (TIME) were also significantly different between the two subtypes (HSS1 and HSS2) of patients. Furthermore, variation in intratumoral microbes-host interaction was associated with the prognostic heterogeneity between HSS1 and HSS2. This study proposes a new CRC classification based on pathological image features and elucidates the process by which tumor microbes-host interactions are reflected in pathological images through the TIME.
Collapse
Affiliation(s)
- Yuwen Chu
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, China
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Shuo Zhang
- School of management, Harbin Institute of Technology, Harbin, China
| | - Wei Wan
- Department of Colorectal and Anal Surgery, Yidu Central Hospital of Weifang, Shandong, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Yumeng Zhang
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
- School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Chuanqi Nie
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, China
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Weipeng Xing
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, China
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Shanhe Tong
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, China
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Jinyang Liu
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Bing Wang
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| |
Collapse
|
8
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [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: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
9
|
Hou S, Wang H, Wang X, Chen H, Zhou B, Meng R, Sha X, Chang S, Wang H, Jiang W. Tumor-liver interface in MRI of liver metastasis enables prediction of EGFR mutation in patients with lung cancer: A proof-of-concept study. Med Phys 2024; 51:1083-1091. [PMID: 37408393 DOI: 10.1002/mp.16581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Preoperative prediction of the epidermal growth factor receptor (EGFR) status in non-small-cell lung cancer (NSCLC) patients with liver metastasis (LM) may have potential clinical values for assisting in treatment decision-making. PURPOSE To explore the value of tumor-liver interface (TLI)-based magnetic resonance imaging (MRI) radiomics for detecting the EGFR mutation in NSCLC patients with LM. METHODS This retrospective study included 123 and 44 patients from hospital 1 (between Feb. 2018 and Dec. 2021) and hospital 2 (between Nov. 2015 and Aug. 2022), respectively. The patients received contrast-enhanced T1-weighted (CET1) and T2-weighted (T2W) liver MRI scans before treatment. Radiomics features were extracted from MRI images of TLI and the whole tumor region, separately. The least absolute shrinkage and selection operator (LASSO) regression was used to screen the features and establish radiomics signatures (RSs) based on TLI (RS-TLI) and the whole tumor (RS-W). The RSs were evaluated by the receiver operating characteristic (ROC) curve analysis. RESULTS A total of 5 and 6 features were identified highly correlated with the EGFR mutation status from TLI and the whole tumor, respectively. The RS-TLI showed better prediction performance than RS-W in the training (AUCs, RS-TLI vs. RS-W, 0.842 vs. 0.797), internal validation (AUCs, RS-TLI vs. RS-W, 0.771 vs. 0.676) and external validation (AUCs, RS-TLI vs. RS-W, 0.733 vs. 0.679) cohort. CONCLUSION Our study demonstrated that TLI-based radiomics can improve prediction performance of the EGFR mutation in lung cancer patients with LM. The established multi-parametric MRI radiomics models may be used as new markers that can potentially assist in personalized treatment planning.
Collapse
Affiliation(s)
- Shaoping Hou
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Hongbo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiaoyu Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital, Shenyang, Liaoning, P.R. China
| | - Boyu Zhou
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Ruiqing Meng
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Xianzheng Sha
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Shijie Chang
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, P.R. China
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, P.R. China
| |
Collapse
|
10
|
Cellina M, De Padova G, Caldarelli N, Libri D, Cè M, Martinenghi C, Alì M, Papa S, Carrafiello G. Artificial Intelligence in Lung Cancer Imaging: From Data to Therapy. Crit Rev Oncog 2024; 29:1-13. [PMID: 38505877 DOI: 10.1615/critrevoncog.2023050439] [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: 03/21/2024]
Abstract
Lung cancer remains a global health challenge, leading to substantial morbidity and mortality. While prevention and early detection strategies have improved, the need for precise diagnosis, prognosis, and treatment remains crucial. In this comprehensive review article, we explore the role of artificial intelligence (AI) in reshaping the management of lung cancer. AI may have different potential applications in lung cancer characterization and outcome prediction. Manual segmentation is a time-consuming task, with high inter-observer variability, that can be replaced by AI-based approaches, including deep learning models such as U-Net, BCDU-Net, and others, to quantify lung nodules and cancers objectively and to extract radiomics features for the characterization of the tissue. AI models have also demonstrated their ability to predict treatment responses, such as immunotherapy and targeted therapy, by integrating radiomic features with clinical data. Additionally, AI-based prognostic models have been developed to identify patients at higher risk and personalize treatment strategies. In conclusion, this review article provides a comprehensive overview of the current state of AI applications in lung cancer management, spanning from segmentation and virtual biopsy to outcome prediction. The evolving role of AI in improving the precision and effectiveness of lung cancer diagnosis and treatment underscores its potential to significantly impact clinical practice and patient outcomes.
Collapse
Affiliation(s)
- Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| | - Giuseppe De Padova
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Nazarena Caldarelli
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Libri
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Maurizio Cè
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Carlo Martinenghi
- Radiology Department, Ospedale San Raffaele, Via Olgettina, 60 - 20132 Milan, Italy
| | - Marco Alì
- Radiology Unit, CDI, Centro Diagnostico Italiano, Via Simone Saint Bon, 20, 20147 Milan, Italy
| | - Sergio Papa
- Radiology Unit, CDI, Centro Diagnostico Italiano, Via Simone Saint Bon, 20, 20147 Milan, Italy
| | - Gianpaolo Carrafiello
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy; Radiology Department, Fondazione IRCCS Cà Granda, Policlinico di Milano Ospedale Maggiore, Università di Milano, 20122 Milan, Italy
| |
Collapse
|
11
|
Cheng Y, Wang H, Yuan W, Wang H, Zhu Y, Chen H, Jiang W. Combined radiomics of primary tumour and bone metastasis improve the prediction of EGFR mutation status and response to EGFR-TKI therapy for NSCLC. Phys Med 2023; 116:103177. [PMID: 38000098 DOI: 10.1016/j.ejmp.2023.103177] [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: 12/16/2022] [Revised: 10/08/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
PURPOSE To develop radiomics models of primary tumour and spinal metastases to predict epidermal growth factor receptor (EGFR) mutations and therapeutic response to EGFR-tyrosine kinase inhibitor (TKI) in patients with metastatic non-small-cell lung cancer (NSCLC). METHODS We enrolled 203 patients with spinal metastases between December 2017 and September 2021, classified as patients with the EGFR mutation or EGFR wild-type. All patients underwent thoracic CT and spinal MRI scans before any treatment. Radiomics analysis was performed to extract features from primary tumour and metastases images and identify predictive features with the least absolute shrinkage and selection operator. Radiomics signatures (RS) were constructed based on primary tumour (RS-Pri), metastases (RS-Met), and in combination (RS-Com) to predict EGFR mutation status and response to EGFR-TKI. Receiver operating characteristic (ROC) curve analysis with 10-fold cross-validation was applied to assess the performance of the models. RESULTS To predict the EGFR mutation status, the RS based on the combination of primary tumour and metastases improved the prediction AUCs compared to those based on the primary tumour or metastasis alone in the training (RS-Com-EGFR: 0.927) and validation (RS-Com-EGFR: 0.812) cohorts. To predict response to EGFR-TKI, the developed RS based on combined primary tumour and metastasis generated the highest AUCs in the training (RS-Com-TKI: 0.880) and validation (RS-Com-TKI: 0.798) cohort. CONCLUSIONS Primary NSCLC and spinal metastases can provide complementary information to predict the EGFR mutation status and response to EGFR-TKI. The developed models that integrate primary lesions and metastases may be potential imaging markers to guide individual treatment decisions.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Liaoning 110122, PR China
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning 110042, PR China
| | - Wendi Yuan
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Liaoning 110122, PR China
| | - Haotian Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning 110042, PR China
| | - Yuheng Zhu
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Liaoning 110122, PR China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 110004 Shenyang, PR China.
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning 110042, PR China.
| |
Collapse
|
12
|
Gao D, Li Y, Xiang S, Zhang J. Key Targets and Molecular Mechanisms of the Fat-soluble Components of Ginseng for Lung Cancer Treatment. Appl Biochem Biotechnol 2023; 195:6495-6515. [PMID: 36870024 PMCID: PMC10643425 DOI: 10.1007/s12010-023-04409-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] [Accepted: 02/17/2023] [Indexed: 03/05/2023]
Abstract
OBJECTIVE To analyze the regulatory effects and key targets of the fat-soluble components of ginseng in lung cancer. METHODS Gas chromatography-mass spectrometry and the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform were used to analyze and identify the fat-soluble components of ginseng. Network pharmacology was used to analyze the therapeutic targets of the fat-soluble components of ginseng in lung cancer and screen key proteins. In vitro assays were conducted to verify the effects of the active fat-soluble components of ginseng on proliferation and apoptosis in lung cancer cells and to verify the regulation of key proteins. RESULTS Ten active fat-soluble components of ginseng were screened for follow-up. Network pharmacology showed 33 overlapping targets between the active fat-soluble components of ginseng and lung cancer, and functional enrichment of the targets showed involvement of response to nitrogen, hormone response, membrane raft, and positive regulation of external stimulus. Pathway enrichment analysis showed vascular endothelial growth factor (VEGF) signaling, adipocyte lipolysis regulation, chronic myelogenous leukemia, endocrine resistance, and NSCLC-related pathways. A protein-protein interaction network was constructed, and the top 10 targets were selected in accordance with their scores. Ultimately, five target genes (EGFR, KDR, MAPK3, PTPN11, and CTNNB1) were selected in combination with literature mining for subsequent experimental verification. Proliferation assays showed that the growth of lung cancer cells was significantly decreased in a concentration-dependent manner in the fat-soluble components of ginseng intervention group compared with controls. Flow cytometry showed that active fat-soluble components of ginseng promoted apoptosis in a concentration-dependent manner in lung cancer cells. Western blot and quantitative real-time PCR showed that levels of the five key proteins and mRNAs were significantly decreased in the intervention group; furthermore, histone protein and mRNA levels were significantly higher in the high-concentration intervention group compared with the low-concentration group. CONCLUSION The active fat-soluble components of ginseng inhibited the growth of lung cancer cells and promoted apoptosis. The underlying regulatory mechanisms may be related to signaling pathways involving EGFR, KDR, MAPK3, PTPN11, and CTNNB1.
Collapse
Affiliation(s)
- Dongdong Gao
- Department of Oncology, Zhumadian Central Hospital, 463000, Zhumadian, China
| | - Yingyue Li
- Medical Engineering Technology and Data Mining Institute, Zhengzhou University, 450001, Zhengzhou, China
| | - Sen Xiang
- Department of Oncology, Zhumadian Central Hospital, 463000, Zhumadian, China
| | - Jing Zhang
- School of Medicine, Huanghuai University, 463000, Zhumadian, China.
| |
Collapse
|
13
|
Zhang T, Liu Z, Lin L, Han T, Long F, Guo H, Han L. Detection of the gene mutation of epidermal growth factor receptor in lung adenocarcinoma by radiomic features from a small amount of PET data. Nucl Med Commun 2023; 44:795-802. [PMID: 37334529 DOI: 10.1097/mnm.0000000000001718] [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: 06/20/2023]
Abstract
OBJECTIVE The purpose of this work was to identify the potential mutation of epidermal growth factor receptor in nonsmall cell adenocarcinoma by noninvasive method, and to explore whether the same or better effect can be achieved using a small amount of single-mode PET image data. METHOD A total of 115 patients were recruited and the results of their 18F-FDG PET images and gene detection after resection were obtained; 117 original radiation features and 744 wavelet transform features were extracted from PET images. Several methods were used to reduce the dimension of the data, and four classifier models were established to classify it. The above process was repeated to reduce the total amount of data and the area under the receiver operating characteristic curve (AUC) value that changed with the reduction of the data and the stability of the results was recorded. RESULTS The classifier with the best comprehensive performance under this dataset was logistic regression, whose AUC value is 0.843. And similar results can be obtained from only 30 cases of data. CONCLUSION A similar or better result could be achieved using a small number of single-mode PET images. In addition, significant results could be obtained using only the PET images of 30 patients.
Collapse
Affiliation(s)
- Tianyou Zhang
- Department of Radiology, Chinese Academy of Medical Sciences Institute of Hematology and Blood Diseases Hospital
| | - Zefeng Liu
- Department of Radiology, Tianjin Medical University General Hospital
| | - Liying Lin
- First Central Clinical College, Tianjin Medical University, Tianjin
| | | | - Fenghua Long
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Hongyu Guo
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Li Han
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
- Department of Radiology, University of Michigan, Ann Arbor Michigan, USA
| |
Collapse
|
14
|
Agyekum EA, Wang YG, Xu FJ, Akortia D, Ren YZ, Chambers KH, Wang X, Taupa JO, Qian XQ. Predicting BRAFV600E mutations in papillary thyroid carcinoma using six machine learning algorithms based on ultrasound elastography. Sci Rep 2023; 13:12604. [PMID: 37537230 PMCID: PMC10400539 DOI: 10.1038/s41598-023-39747-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023] Open
Abstract
The most common BRAF mutation is thymine (T) to adenine (A) missense mutation in nucleotide 1796 (T1796A, V600E). The BRAFV600E gene encodes a protein-dependent kinase (PDK), which is a key component of the mitogen-activated protein kinase pathway and essential for controlling cell proliferation, differentiation, and death. The BRAFV600E mutation causes PDK to be activated improperly and continuously, resulting in abnormal proliferation and differentiation in PTC. Based on elastography ultrasound (US) radiomic features, this study seeks to create and validate six distinct machine learning algorithms to predict BRAFV6OOE mutation in PTC patients prior to surgery. This study employed routine US strain elastography image data from 138 PTC patients. The patients were separated into two groups: those who did not have the BRAFV600E mutation (n = 75) and those who did have the mutation (n = 63). The patients were randomly assigned to one of two data sets: training (70%), or validation (30%). From strain elastography US images, a total of 479 radiomic features were retrieved. Pearson's Correlation Coefficient (PCC) and Recursive Feature Elimination (RFE) with stratified tenfold cross-validation were used to decrease the features. Based on selected radiomic features, six machine learning algorithms including support vector machine with the linear kernel (SVM_L), support vector machine with radial basis function kernel (SVM_RBF), logistic regression (LR), Naïve Bayes (NB), K-nearest neighbors (KNN), and linear discriminant analysis (LDA) were compared to predict the possibility of BRAFV600E. The accuracy (ACC), the area under the curve (AUC), sensitivity (SEN), specificity (SPEC), positive predictive value (PPV), negative predictive value (NPV), decision curve analysis (DCA), and calibration curves of the machine learning algorithms were used to evaluate their performance. ① The machine learning algorithms' diagnostic performance depended on 27 radiomic features. ② AUCs for NB, KNN, LDA, LR, SVM_L, and SVM_RBF were 0.80 (95% confidence interval [CI]: 0.65-0.91), 0.87 (95% CI 0.73-0.95), 0.91(95% CI 0.79-0.98), 0.92 (95% CI 0.80-0.98), 0.93 (95% CI 0.80-0.98), and 0.98 (95% CI 0.88-1.00), respectively. ③ There was a significant difference in echogenicity,vertical and horizontal diameter ratios, and elasticity between PTC patients with BRAFV600E and PTC patients without BRAFV600E. Machine learning algorithms based on US elastography radiomic features are capable of predicting the likelihood of BRAFV600E in PTC patients, which can assist physicians in identifying the risk of BRAFV600E in PTC patients. Among the six machine learning algorithms, the support vector machine with radial basis function (SVM_RBF) achieved the best ACC (0.93), AUC (0.98), SEN (0.95), SPEC (0.90), PPV (0.91), and NPV (0.95).
Collapse
Affiliation(s)
- Enock Adjei Agyekum
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu-Guo Wang
- Department of Ultrasound, Traditional Chinese Medicine Hospital of Nanjing Lishui District, Nanjing, China
| | - Fei-Ju Xu
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
| | - Debora Akortia
- School of Public Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Yong-Zhen Ren
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | | | - Xian Wang
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
| | - Jenny Olalia Taupa
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiao-Qin Qian
- Ultrasound Medical Laboratory, Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China.
| |
Collapse
|
15
|
Chen W, Sá RC, Bai Y, Napel S, Gevaert O, Lauderdale DS, Giger ML. Machine learning with multimodal data for COVID-19. Heliyon 2023; 9:e17934. [PMID: 37483733 PMCID: PMC10362086 DOI: 10.1016/j.heliyon.2023.e17934] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
In response to the unprecedented global healthcare crisis of the COVID-19 pandemic, the scientific community has joined forces to tackle the challenges and prepare for future pandemics. Multiple modalities of data have been investigated to understand the nature of COVID-19. In this paper, MIDRC investigators present an overview of the state-of-the-art development of multimodal machine learning for COVID-19 and model assessment considerations for future studies. We begin with a discussion of the lessons learned from radiogenomic studies for cancer diagnosis. We then summarize the multi-modality COVID-19 data investigated in the literature including symptoms and other clinical data, laboratory tests, imaging, pathology, physiology, and other omics data. Publicly available multimodal COVID-19 data provided by MIDRC and other sources are summarized. After an overview of machine learning developments using multimodal data for COVID-19, we present our perspectives on the future development of multimodal machine learning models for COVID-19.
Collapse
Affiliation(s)
- Weijie Chen
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Rui C. Sá
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine, University of California, San Diego, USA
| | - Yuntong Bai
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Sandy Napel
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, Stanford University, USA
| | - Olivier Gevaert
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine and Department of Biomedical Data Science, Stanford University, USA
| | - Diane S. Lauderdale
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Public Health Sciences, University of Chicago, USA
| | - Maryellen L. Giger
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, University of Chicago, USA
| |
Collapse
|
16
|
Selby HM, Mukherjee P, Parham C, Malik SB, Gevaert O, Napel S, Shah RP. Performance of alternative manual and automated deep learning segmentation techniques for the prediction of benign and malignant lung nodules. J Med Imaging (Bellingham) 2023; 10:044006. [PMID: 37564098 PMCID: PMC10411216 DOI: 10.1117/1.jmi.10.4.044006] [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: 10/04/2022] [Revised: 05/02/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Purpose We aim to evaluate the performance of radiomic biopsy (RB), best-fit bounding box (BB), and a deep-learning-based segmentation method called no-new-U-Net (nnU-Net), compared to the standard full manual (FM) segmentation method for predicting benign and malignant lung nodules using a computed tomography (CT) radiomic machine learning model. Materials and Methods A total of 188 CT scans of lung nodules from 2 institutions were used for our study. One radiologist identified and delineated all 188 lung nodules, whereas a second radiologist segmented a subset (n = 20 ) of these nodules. Both radiologists employed FM and RB segmentation methods. BB segmentations were generated computationally from the FM segmentations. The nnU-Net, a deep-learning-based segmentation method, performed automatic nodule detection and segmentation. The time radiologists took to perform segmentations was recorded. Radiomic features were extracted from each segmentation method, and models to predict benign and malignant lung nodules were developed. The Kruskal-Wallis and DeLong tests were used to compare segmentation times and areas under the curve (AUC), respectively. Results For the delineation of the FM, RB, and BB segmentations, the two radiologists required a median time (IQR) of 113 (54 to 251.5), 21 (9.25 to 38), and 16 (12 to 64.25) s, respectively (p = 0.04 ). In dataset 1, the mean AUC (95% CI) of the FM, RB, BB, and nnU-Net model were 0.964 (0.96 to 0.968), 0.985 (0.983 to 0.987), 0.961 (0.956 to 0.965), and 0.878 (0.869 to 0.888). In dataset 2, the mean AUC (95% CI) of the FM, RB, BB, and nnU-Net model were 0.717 (0.705 to 0.729), 0.919 (0.913 to 0.924), 0.699 (0.687 to 0.711), and 0.644 (0.632 to 0.657). Conclusion Radiomic biopsy-based models outperformed FM and BB models in prediction of benign and malignant lung nodules in two independent datasets while deep-learning segmentation-based models performed similarly to FM and BB. RB could be a more efficient segmentation method, but further validation is needed.
Collapse
Affiliation(s)
- Heather M. Selby
- Stanford University School of Medicine, Stanford Center for Biomedical Informatics (BMIR), Stanford, California, United States
| | - Pritam Mukherjee
- National Institutes of Health Clinical Center, Bethesda, Maryland, United States
| | - Christopher Parham
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
| | - Sachin B. Malik
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
| | - Olivier Gevaert
- Stanford University School of Medicine, Stanford Center for Biomedical Informatics (BMIR), Stanford, California, United States
| | - Sandy Napel
- Stanford University School of Medicine, Department of Radiology, Stanford, California, United States
| | - Rajesh P. Shah
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
- Stanford University School of Medicine, Department of Radiology, Stanford, California, United States
| |
Collapse
|
17
|
Yan W, Quan C, Mourad WF, Yuan J, Shi Z, Yang J, Lu Q, Zhang J. Application of radiomics in lung immuno-oncology. PRECISION RADIATION ONCOLOGY 2023; 7:128-136. [PMID: 40337267 PMCID: PMC11935008 DOI: 10.1002/pro6.1191] [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: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 04/08/2023] Open
Abstract
Radiomics is a rapidly evolving field of research that extracts and analyzes quantitative features within medical images. Those features are termed as radiomic features that can characterize a tumor in a comprehensive and quantitative manner with regard to its internal structure and heterogeneity. Radiomic features can be used, alone or in combination with demographic, histological, genomic, or proteomic data, for predicting prognosis or treatment response. Immunotherapy, or immune-oncology, is the study of cancer treatment by taking advantage of the body's immune system to prevent, control, and eliminate cancer. In this review, we first provide a brief introduction to both radiomics and immune-oncology in lung cancer. Then, we discuss the need for developing immune-oncology biomarkers, and the advantages of radiomics in identifying biomarkers related to immunotherapy. We also discuss potential areas in and out of tumors, such as the intra-tumoral hypoxic region and tumor microenvironment, where radiomic markers might be extracted, as well as a potential application of radiomic biomarkers in clinical lung cancer management. Finally, we present radiation and immune modulation in non-small cell lung cancer, clinical trials and their design to incorporate radiomic biomarkers, and radiomics-guided precision radiation therapy.
Collapse
Affiliation(s)
- Weisi Yan
- Baptist Health SystemLexingtonKentuckyUSA
| | - Chen Quan
- City of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Waleed F. Mourad
- Department of Radiation MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Jianda Yuan
- Translational Oncology at Merck & CoKenilworthNew JerseyUSA
| | | | - Jun Yang
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Qiuxia Lu
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Jie Zhang
- Department of RadiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
18
|
Steyaert S, Pizurica M, Nagaraj D, Khandelwal P, Hernandez-Boussard T, Gentles AJ, Gevaert O. Multimodal data fusion for cancer biomarker discovery with deep learning. NAT MACH INTELL 2023; 5:351-362. [PMID: 37693852 PMCID: PMC10484010 DOI: 10.1038/s42256-023-00633-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/17/2023] [Indexed: 09/12/2023]
Abstract
Technological advances now make it possible to study a patient from multiple angles with high-dimensional, high-throughput multi-scale biomedical data. In oncology, massive amounts of data are being generated ranging from molecular, histopathology, radiology to clinical records. The introduction of deep learning has significantly advanced the analysis of biomedical data. However, most approaches focus on single data modalities leading to slow progress in methods to integrate complementary data types. Development of effective multimodal fusion approaches is becoming increasingly important as a single modality might not be consistent and sufficient to capture the heterogeneity of complex diseases to tailor medical care and improve personalised medicine. Many initiatives now focus on integrating these disparate modalities to unravel the biological processes involved in multifactorial diseases such as cancer. However, many obstacles remain, including lack of usable data as well as methods for clinical validation and interpretation. Here, we cover these current challenges and reflect on opportunities through deep learning to tackle data sparsity and scarcity, multimodal interpretability, and standardisation of datasets.
Collapse
Affiliation(s)
- Sandra Steyaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | | | | | - Tina Hernandez-Boussard
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Andrew J Gentles
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| |
Collapse
|
19
|
Jia L, Wu W, Hou G, Zhao J, Qiang Y, Zhang Y, Cai M. Residual neural network with mixed loss based on batch training technique for identification of EGFR mutation status in lung cancer. MULTIMEDIA TOOLS AND APPLICATIONS 2023; 82:1-21. [PMID: 37362735 PMCID: PMC10020767 DOI: 10.1007/s11042-023-14876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 11/11/2022] [Accepted: 02/06/2023] [Indexed: 06/28/2023]
Abstract
Epidermal growth factor receptor (EGFR) is the key to targeted therapy with tyrosine kinase inhibitors in lung cancer. Traditional identification of EGFR mutation status requires biopsy and sequence testing, which may not be suitable for certain groups who cannot perform biopsy. In this paper, using easily accessible and non-invasive CT images, the residual neural network (ResNet) with mixed loss based on batch training technique is proposed for identification of EGFR mutation status in lung cancer. In this model, the ResNet is regarded as the baseline for feature extraction to avoid the gradient disappearance. Besides, a new mixed loss based on the batch similarity and the cross entropy is proposed to guide the network to better learn the model parameters. The proposed mixed loss utilizes the similarity among batch samples to evaluate the distribution of training data, which can reduce the similarity of different classes and the difference of the same classes. In the experiments, VGG16Net, DenseNet, ResNet18, ResNet34 and ResNet50 models with the mixed loss are trained on the public CT dataset with 155 patients including EGFR mutation status from TCIA. The trained networks are employed to the collected preoperative CT dataset with 56 patients from the cooperative hospital for validating the efficiency of the proposed models. Experimental results show that the proposed models are more appropriate and effective on the lung cancer dataset for identifying the EGFR mutation status. In these models, the ResNet34 with mixed loss is optimal (accuracy = 81.58%, AUC = 0.8861, sensitivity = 80.02%, specificity = 82.90%).
Collapse
Affiliation(s)
- Liye Jia
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Wei Wu
- Department of Physiology, Shanxi Medical University, Taiyuan, 030051 China
| | - Guojie Hou
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Juanjuan Zhao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Yan Qiang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Yanan Zhang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Meiling Cai
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| |
Collapse
|
20
|
Hochhegger B, Pasini R, Roncally Carvalho A, Rodrigues R, Altmayer S, Kayat Bittencourt L, Marchiori E, Forghani R. Artificial Intelligence for Cardiothoracic Imaging: Overview of Current and Emerging Applications. Semin Roentgenol 2023; 58:184-195. [PMID: 37087139 DOI: 10.1053/j.ro.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 03/07/2023]
Abstract
Artificial intelligence algorithms can learn by assimilating information from large datasets in order to decipher complex associations, identify previously undiscovered pathophysiological states, and construct prediction models. There has been tremendous interest and increased incorporation of artificial intelligence into various industries, including healthcare. As a result, there has been an exponential rise in the number of research articles and industry participants producing models intended for a variety of applications in medical imaging, which can be challenging to navigate for radiologists. In thoracic imaging, multiple applications are being evaluated for chest radiography and computed tomography and include applications for lung nodule evaluation and cancer imaging, quantifying diffuse lung disorders, and cardiac imaging, to name a few. This review aims to provide an overview of current clinical AI models, focusing on the most common clinical applications of AI in cardiothoracic imaging.
Collapse
|
21
|
Dovrou A, Bei E, Sfakianakis S, Marias K, Papanikolaou N, Zervakis M. Synergies of Radiomics and Transcriptomics in Lung Cancer Diagnosis: A Pilot Study. Diagnostics (Basel) 2023; 13:738. [PMID: 36832225 PMCID: PMC9955510 DOI: 10.3390/diagnostics13040738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Radiotranscriptomics is an emerging field that aims to investigate the relationships between the radiomic features extracted from medical images and gene expression profiles that contribute in the diagnosis, treatment planning, and prognosis of cancer. This study proposes a methodological framework for the investigation of these associations with application on non-small-cell lung cancer (NSCLC). Six publicly available NSCLC datasets with transcriptomics data were used to derive and validate a transcriptomic signature for its ability to differentiate between cancer and non-malignant lung tissue. A publicly available dataset of 24 NSCLC-diagnosed patients, with both transcriptomic and imaging data, was used for the joint radiotranscriptomic analysis. For each patient, 749 Computed Tomography (CT) radiomic features were extracted and the corresponding transcriptomics data were provided through DNA microarrays. The radiomic features were clustered using the iterative K-means algorithm resulting in 77 homogeneous clusters, represented by meta-radiomic features. The most significant differentially expressed genes (DEGs) were selected by performing Significance Analysis of Microarrays (SAM) and 2-fold change. The interactions among the CT imaging features and the selected DEGs were investigated using SAM and a Spearman rank correlation test with a False Discovery Rate (FDR) of 5%, leading to the extraction of 73 DEGs significantly correlated with radiomic features. These genes were used to produce predictive models of the meta-radiomics features, defined as p-metaomics features, by performing Lasso regression. Of the 77 meta-radiomic features, 51 can be modeled in terms of the transcriptomic signature. These significant radiotranscriptomics relationships form a reliable basis to biologically justify the radiomics features extracted from anatomic imaging modalities. Thus, the biological value of these radiomic features was justified via enrichment analysis on their transcriptomics-based regression models, revealing closely associated biological processes and pathways. Overall, the proposed methodological framework provides joint radiotranscriptomics markers and models to support the connection and complementarities between the transcriptome and the phenotype in cancer, as demonstrated in the case of NSCLC.
Collapse
Affiliation(s)
- Aikaterini Dovrou
- Digital Image and Signal Processing Laboratory, School of Electrical and Computer Engineering (ECE), Technical University of Crete, GR-73100 Chania, Greece
| | - Ekaterini Bei
- Digital Image and Signal Processing Laboratory, School of Electrical and Computer Engineering (ECE), Technical University of Crete, GR-73100 Chania, Greece
| | - Stelios Sfakianakis
- Computational BioMedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece
| | - Kostas Marias
- Computational BioMedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR-71410 Heraklion, Greece
| | - Nickolas Papanikolaou
- Computational Clinical Imaging Group, Champalimaud Clinical Centre, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisbon, Portugal
| | - Michalis Zervakis
- Digital Image and Signal Processing Laboratory, School of Electrical and Computer Engineering (ECE), Technical University of Crete, GR-73100 Chania, Greece
| |
Collapse
|
22
|
Hartmann K, Sadée CY, Satwah I, Carrillo-Perez F, Gevaert O. Imaging genomics: data fusion in uncovering disease heritability. Trends Mol Med 2023; 29:141-151. [PMID: 36470817 PMCID: PMC10507799 DOI: 10.1016/j.molmed.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 12/04/2022]
Abstract
Sequencing of the human genome in the early 2000s enabled probing of the genetic basis of disease on a scale previously unimaginable. Now, two decades later, after interrogating millions of markers in thousands of individuals, a significant portion of disease heritability still remains hidden. Recent efforts to unravel this 'missing heritability' have focused on garnering new insight from merging different data types, including medical imaging. Imaging offers promising intermediate phenotypes to bridge the gap between genetic variation and disease pathology. In this review we outline this fusion and provide examples of imaging genomics in a range of diseases, from oncology to cardiovascular and neurodegenerative disease. Finally, we discuss how ongoing revolutions in data science and sharing are primed to advance the field.
Collapse
Affiliation(s)
- Katherine Hartmann
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Christoph Y Sadée
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Ishan Satwah
- College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Computer Architecture and Technology, University of Granada. C.I.T.I.C., Granada, Spain
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Deep learning for preoperative prediction of the EGFR mutation and subtypes based on the MRI image of spinal metastasis from primary NSCLC. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Fan Y, Zhao Z, Wang X, Ai H, Yang C, Luo Y, Jiang X. Radiomics for prediction of response to EGFR-TKI based on metastasis/brain parenchyma (M/BP)-interface. LA RADIOLOGIA MEDICA 2022; 127:1342-1354. [PMID: 36284030 DOI: 10.1007/s11547-022-01569-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE To evaluate the potential of subregional radiomics as a novel tumor marker in predicting epidermal growth factor receptor (EGFR) mutation status and response to EGFR-tyrosine kinase inhibitor (TKI) therapy in NSCLC patients with brain metastasis (BM). MATERIALS AND METHODS We included 230 patients from center 1, and 80 patients were included from center 2 to form a primary and external validation cohort, respectively. Patients underwent contrast-enhanced T1-weighted and T2-weighted MRI scans before treatment. The individual- and population-level clustering was used to partition the peritumoral edema area (POA) into phenotypically consistent subregions. Radiomics features were calculated and selected from the tumor active area (TAA), POA and subregions, and used to develop models. Prediction values of each region were investigated and compared with receiver operating characteristic curves and Delong test. RESULTS For predicting EGFR mutations, a multi-region combined model (EGFR-Fusion) was developed based on joint of the partitioned metastasis/brain parenchyma (M/BP)-interface and TAA, and generated the highest prediction performance in the training (AUC = 0.945, SEN = 0.878, SPE = 0.937), internal validation (AUC = 0.880, SEN = 0.733, SPE = 0.969), and external validation (AUC = 0.895, SEN = 0.875, SPE = 0.800) cohorts. For predicting response to EGFR-TKI, the developed multi-region combined model (TKI-Fusion) yielded predictive AUCs of 0.869 (SEN = 0.717, SPE = 0.884), 0.786 (SEN = 0.708, SPE = 0.818), and 0.802 (SEN = 0.750, SPE = 0.800) in the training, internal validation and external validation cohort, respectively. CONCLUSION Our study revealed that complementary information regarding the EGFR status and response to EGFR-TKI can be provided by subregional radiomics. The proposed radiomics models may be new markers to guide treatment plans for NSCLC patients with BM.
Collapse
Affiliation(s)
- Ying Fan
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Zilong Zhao
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xingling Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Hua Ai
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Chunna Yang
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China.
| | - Xiran Jiang
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
25
|
Artificial Intelligence in Lung Cancer Imaging: Unfolding the Future. Diagnostics (Basel) 2022; 12:diagnostics12112644. [PMID: 36359485 PMCID: PMC9689810 DOI: 10.3390/diagnostics12112644] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is one of the malignancies with higher morbidity and mortality. Imaging plays an essential role in each phase of lung cancer management, from detection to assessment of response to treatment. The development of imaging-based artificial intelligence (AI) models has the potential to play a key role in early detection and customized treatment planning. Computer-aided detection of lung nodules in screening programs has revolutionized the early detection of the disease. Moreover, the possibility to use AI approaches to identify patients at risk of developing lung cancer during their life can help a more targeted screening program. The combination of imaging features and clinical and laboratory data through AI models is giving promising results in the prediction of patients’ outcomes, response to specific therapies, and risk for toxic reaction development. In this review, we provide an overview of the main imaging AI-based tools in lung cancer imaging, including automated lesion detection, characterization, segmentation, prediction of outcome, and treatment response to provide radiologists and clinicians with the foundation for these applications in a clinical scenario.
Collapse
|
26
|
Stahlberg EA, Abdel-Rahman M, Aguilar B, Asadpoure A, Beckman RA, Borkon LL, Bryan JN, Cebulla CM, Chang YH, Chatterjee A, Deng J, Dolatshahi S, Gevaert O, Greenspan EJ, Hao W, Hernandez-Boussard T, Jackson PR, Kuijjer M, Lee A, Macklin P, Madhavan S, McCoy MD, Mohammad Mirzaei N, Razzaghi T, Rocha HL, Shahriyari L, Shmulevich I, Stover DG, Sun Y, Syeda-Mahmood T, Wang J, Wang Q, Zervantonakis I. Exploring approaches for predictive cancer patient digital twins: Opportunities for collaboration and innovation. Front Digit Health 2022; 4:1007784. [PMID: 36274654 PMCID: PMC9586248 DOI: 10.3389/fdgth.2022.1007784] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/30/2022] [Indexed: 01/26/2023] Open
Abstract
We are rapidly approaching a future in which cancer patient digital twins will reach their potential to predict cancer prevention, diagnosis, and treatment in individual patients. This will be realized based on advances in high performance computing, computational modeling, and an expanding repertoire of observational data across multiple scales and modalities. In 2020, the US National Cancer Institute, and the US Department of Energy, through a trans-disciplinary research community at the intersection of advanced computing and cancer research, initiated team science collaborative projects to explore the development and implementation of predictive Cancer Patient Digital Twins. Several diverse pilot projects were launched to provide key insights into important features of this emerging landscape and to determine the requirements for the development and adoption of cancer patient digital twins. Projects included exploring approaches to using a large cohort of digital twins to perform deep phenotyping and plan treatments at the individual level, prototyping self-learning digital twin platforms, using adaptive digital twin approaches to monitor treatment response and resistance, developing methods to integrate and fuse data and observations across multiple scales, and personalizing treatment based on cancer type. Collectively these efforts have yielded increased insights into the opportunities and challenges facing cancer patient digital twin approaches and helped define a path forward. Given the rapidly growing interest in patient digital twins, this manuscript provides a valuable early progress report of several CPDT pilot projects commenced in common, their overall aims, early progress, lessons learned and future directions that will increasingly involve the broader research community.
Collapse
Affiliation(s)
- Eric A. Stahlberg
- Cancer Data Science Initiatives, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Mohamed Abdel-Rahman
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Boris Aguilar
- Institute for Systems Biology, Seattle, WA, United States
| | - Alireza Asadpoure
- Department of Civil and Environmental Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Robert A. Beckman
- Innovation Center for Biomedical Informatics, Georgetown University, Washington DC, United States
| | - Lynn L. Borkon
- Cancer Data Science Initiatives, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jeffrey N. Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States
| | - Colleen M. Cebulla
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, OR, United States
| | - Ansu Chatterjee
- School of Statistics, University of Minnesota, Minneapolis, MN, United States
| | - Jun Deng
- Department of Therapeutic Radiology, Yale University School of Medicine, Yale University, New Haven, CT, United States
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, United States
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine and Department of Biomedical Data Science, Stanford University, Stanford, CA, United States
| | - Emily J. Greenspan
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wenrui Hao
- Department of Mathematics, The Pennsylvania State University, University Park, PA, United States
| | - Tina Hernandez-Boussard
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine and Department of Biomedical Data Science, Stanford University, Stanford, CA, United States
| | - Pamela R. Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Marieke Kuijjer
- Computational Biology and Systems Medicine Group, Centre for Molecular Medicine Norway University of Oslo, Oslo, Norway
| | - Adrian Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
| | - Subha Madhavan
- Innovation Center for Biomedical Informatics, Georgetown University, Washington DC, United States
| | - Matthew D. McCoy
- Innovation Center for Biomedical Informatics, Georgetown University, Washington DC, United States
| | - Navid Mohammad Mirzaei
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, United States
| | - Talayeh Razzaghi
- School of Industrial and Systems Engineering, The University of Oklahoma, Norman, OK, United States
| | - Heber L. Rocha
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, United States
| | | | - Daniel G. Stover
- Division of Medical Oncology and Department of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Yi Sun
- Department of Mathematics, University of South Carolina, Columbia, SC, United States
| | | | - Jinhua Wang
- Institute for Health Informatics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Qi Wang
- Department of Mathematics, University of South Carolina, Columbia, SC, United States
| | - Ioannis Zervantonakis
- Department of Bioengineering, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Deng K, Wang L, Liu Y, Li X, Hou Q, Cao M, Ng NN, Wang H, Chen H, Yeom KW, Zhao M, Wu N, Gao P, Shi J, Liu Z, Li W, Tian J, Song J. A deep learning-based system for survival benefit prediction of tyrosine kinase inhibitors and immune checkpoint inhibitors in stage IV non-small cell lung cancer patients: A multicenter, prognostic study. EClinicalMedicine 2022; 51:101541. [PMID: 35813093 PMCID: PMC9256845 DOI: 10.1016/j.eclinm.2022.101541] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND For clinical decision making, it is crucial to identify patients with stage IV non-small cell lung cancer (NSCLC) who may benefit from tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs). In this study, a deep learning-based system was designed and validated using pre-therapy computed tomography (CT) images to predict the survival benefits of EGFR-TKIs and ICIs in stage IV NSCLC patients. METHODS This retrospective study collected data from 570 patients with stage IV EGFR-mutant NSCLC treated with EGFR-TKIs at five institutions between 2010 and 2021 (data of 314 patients were from a previously registered study), and 129 patients with stage IV NSCLC treated with ICIs at three institutions between 2017 and 2021 to build the ICI test dataset. Five-fold cross-validation was applied to divide the EGFR-TKI-treated patients from four institutions into training and internal validation datasets randomly in a ratio of 80%:20%, and the data from another institution was used as an external test dataset. An EfficientNetV2-based survival benefit prognosis (ESBP) system was developed with pre-therapy CT images as the input and the probability score as the output to identify which patients would receive additional survival benefit longer than the median PFS. Its prognostic performance was validated on the ICI test dataset. For diagnosing which patient would receive additional survival benefit, the accuracy of ESBP was compared with the estimations of three radiologists and three oncologists with varying degrees of expertise (two, five, and ten years). Improvements in the clinicians' diagnostic accuracy with ESBP assistance were then quantified. FINDINGS ESBP achieved positive predictive values of 80·40%, 75·40%, and 77·43% for additional EGFR-TKI survival benefit prediction using the probability score of 0·2 as the threshold on the training, internal validation, and external test datasets, respectively. The higher ESBP score (>0·2) indicated a better prognosis for progression-free survival (hazard ratio: 0·36, 95% CI: 0·19-0·68, p<0·0001) in patients on the external test dataset. Patients with scores >0·2 in the ICI test dataset also showed better survival benefit (hazard ratio: 0·33, 95% CI: 0·18-0·55, p<0·0001). This suggests the potential of ESBP to identify the two subgroups of benefiting patients by decoding the commonalities from pre-therapy CT images (stage IV EGFR-mutant NSCLC patients receiving additional survival benefit from EGFR-TKIs and stage IV NSCLC patients receiving additional survival benefit from ICIs). ESBP assistance improved the diagnostic accuracy of the clinicians with two years of experience from 47·91% to 66·32%, and the clinicians with five years of experience from 53·12% to 61·41%. INTERPRETATION This study developed and externally validated a preoperative CT image-based deep learning model to predict the survival benefits of EGFR-TKI and ICI therapies in stage IV NSCLC patients, which will facilitate optimized and individualized treatment strategies. FUNDING This study received funding from the National Natural Science Foundation of China (82001904, 81930053, and 62027901), and Key-Area Research and Development Program of Guangdong Province (2021B0101420005).
Collapse
Affiliation(s)
- 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, China
| | - Lu Wang
- Library of Shengjing Hospital of China Medical University, Shenyang, China
- School of Health Management, China Medical University, Shenyang, Liaoning, China
| | - Yuchan Liu
- 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, China
| | - Xin Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qiuyang Hou
- 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, China
| | - Mulan Cao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Nathan Norton Ng
- Department of Radiology, School of Medicine Stanford University, Stanford CA 94305, United States
| | - Huan Wang
- Radiation oncology department of thoracic cancer, Liaoning Cancer Hospital and Institute, Liaoning, China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kristen W. Yeom
- Department of Radiology, School of Medicine Stanford University, Stanford CA 94305, United States
| | - Mingfang Zhao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ning Wu
- PET-CT center, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Gao
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingyun Shi
- Department of Radiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu, Sichuan, China
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Jiangdian Song
- School of Health Management, China Medical University, Shenyang, Liaoning, China
- Corresponding author at: School of Health Management, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
28
|
Vliegenthart R, Fouras A, Jacobs C, Papanikolaou N. Innovations in thoracic imaging: CT, radiomics, AI and x-ray velocimetry. Respirology 2022; 27:818-833. [PMID: 35965430 PMCID: PMC9546393 DOI: 10.1111/resp.14344] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/08/2022] [Indexed: 12/11/2022]
Abstract
In recent years, pulmonary imaging has seen enormous progress, with the introduction, validation and implementation of new hardware and software. There is a general trend from mere visual evaluation of radiological images to quantification of abnormalities and biomarkers, and assessment of ‘non visual’ markers that contribute to establishing diagnosis or prognosis. Important catalysts to these developments in thoracic imaging include new indications (like computed tomography [CT] lung cancer screening) and the COVID‐19 pandemic. This review focuses on developments in CT, radiomics, artificial intelligence (AI) and x‐ray velocimetry for imaging of the lungs. Recent developments in CT include the potential for ultra‐low‐dose CT imaging for lung nodules, and the advent of a new generation of CT systems based on photon‐counting detector technology. Radiomics has demonstrated potential towards predictive and prognostic tasks particularly in lung cancer, previously not achievable by visual inspection by radiologists, exploiting high dimensional patterns (mostly texture related) on medical imaging data. Deep learning technology has revolutionized the field of AI and as a result, performance of AI algorithms is approaching human performance for an increasing number of specific tasks. X‐ray velocimetry integrates x‐ray (fluoroscopic) imaging with unique image processing to produce quantitative four dimensional measurement of lung tissue motion, and accurate calculations of lung ventilation. See relatedEditorial
Collapse
Affiliation(s)
- Rozemarijn Vliegenthart
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Data Science in Health (DASH), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Colin Jacobs
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nickolas Papanikolaou
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal.,AI Hub, The Royal Marsden NHS Foundation Trust, London, UK.,The Institute of Cancer Research, London, UK
| |
Collapse
|
29
|
Shi J, Zhao Z, Jiang T, Ai H, Liu J, Chen X, Luo Y, Fan H, Jiang X. A deep learning approach with subregion partition in MRI image analysis for metastatic brain tumor. Front Neuroinform 2022; 16:973698. [PMID: 35991287 PMCID: PMC9382021 DOI: 10.3389/fninf.2022.973698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo propose a deep learning network with subregion partition for predicting metastatic origins and EGFR/HER2 status in patients with brain metastasis.MethodsWe retrospectively enrolled 140 patients with clinico-pathologically confirmed brain metastasis originated from primary NSCLC (n = 60), breast cancer (BC, n = 60) and other tumor types (n = 20). All patients underwent contrast-enhanced brain MRI scans. The brain metastasis was subdivided into phenotypically consistent subregions using patient-level and population-level clustering. A residual network with a global average pooling layer (RN-GAP) was proposed to calculate deep learning-based features. Features from each subregion were selected with least absolute shrinkage and selection operator (LASSO) to build logistic regression models (LRs) for predicting primary tumor types (LR-NSCLC for the NSCLC origin and LR-BC for the BC origin), EGFR mutation status (LR-EGFR) and HER2 status (LR-HER2).ResultsThe brain metastasis can be partitioned into a marginal subregion (S1) and an inner subregion (S2) in the MRI image. The developed models showed good predictive performance in the training (AUCs, LR-NSCLC vs. LR-BC vs. LR-EGFR vs. LR-HER2, 0.860 vs. 0.909 vs. 0.850 vs. 0.900) and validation (AUCs, LR-NSCLC vs. LR-BC vs. LR-EGFR vs. LR-HER2, 0.819 vs. 0.872 vs. 0.750 vs. 0.830) set.ConclusionOur proposed deep learning network with subregion partitions can accurately predict metastatic origins and EGFR/HER2 status of brain metastasis, and hence may have the potential to be non-invasive and preoperative new markers for guiding personalized treatment plans in patients with brain metastasis.
Collapse
Affiliation(s)
- Jiaxin Shi
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Zilong Zhao
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tao Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Hua Ai
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Jiani Liu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Xinpu Chen
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Huijie Fan
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- *Correspondence: Huijie Fan,
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
- Xiran Jiang,
| |
Collapse
|
30
|
Silva P, Pereira T, Teixeira M, Silva F, Oliveira HP. On the way for the best imaging features from CT images to predict EGFR Mutation Status in Lung Cancer. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:2659-2662. [PMID: 36085894 DOI: 10.1109/embc48229.2022.9871911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Artificial Intelligence-based tools have shown promising results to help clinicians in diagnosis tasks. Radio-genomics would aid in the genotype characterization using information from radiologic images. The prediction of the mutations status of main oncogenes associated with lung cancer will help the clinicians to have a more accurate diagnosis and a personalized treatment plan, decreasing the need to use the biopsy. In this work, novel and objective features were extracted from the lung that contained the nodule, and several machine learning methods were combined with feature selection techniques to select the best approach to predict the EGFR mutation status in lung cancer CT images. An AUC of 0.756 ± 0.055 was obtained using a logistic regression and independent component analysis as feature selector, supporting the hypothesis that CT images can capture pathophysiological information with great value for clinical assessment and personalized medicine of lung cancer. Clinical Relevance - Radiogenomic approaches could be an interesting help for lung cancer characterization. This work represents a preliminary study for the development of computer-aided decision systems to provide a more accurate and fast characterization of lung cancer which is fundamental for an adequate treatment plan for lung cancer patients.
Collapse
|
31
|
Ferreira A, Pereira T, Silva F, Vilares AT, Silva MC, Cunha A, Oliveira HP. Synthesizing 3D Lung CT scans with Generative Adversarial Networks. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:2033-2036. [PMID: 36085795 DOI: 10.1109/embc48229.2022.9871481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In the healthcare domain, datasets are often private and lack large amounts of samples, making it difficult to cope with the inherent patient data heterogeneity. As an attempt to mitigate data scarcity, generative models are being used due to their ability to produce new data, using a dataset as a reference. However, synthesis studies often rely on a 2D representation of data, a seriously limited form of information when it comes to lung computed tomography scans where, for example, pathologies like nodules can manifest anywhere in the organ. Here, we develop a 3D Progressive Growing Generative Adversarial Network capable of generating thoracic CT volumes at a resolution of 1283, and analyze the model outputs through a quantitative metric (3D Muli-Scale Structural Similarity) and a Visual Turing Test. Clinical relevance - This paper is a novel application of the 3D PGGAN model to synthesize CT lung scans. This preliminary study focuses on synthesizing the entire volume of the lung rather than just the lung nodules. The synthesized data represent an attempt to mitigate data scarcity which is one of the major limitations to create learning models with good generalization in healthcare.
Collapse
|
32
|
Feng Y, Song F, Zhang P, Fan G, Zhang T, Zhao X, Ma C, Sun Y, Song X, Pu H, Liu F, Zhang G. Prediction of EGFR Mutation Status in Non–Small Cell Lung Cancer Based on Ensemble Learning. Front Pharmacol 2022; 13:897597. [PMID: 35833032 PMCID: PMC9271946 DOI: 10.3389/fphar.2022.897597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/19/2022] [Indexed: 11/29/2022] Open
Abstract
Objectives: We aimed to identify whether ensemble learning can improve the performance of the epidermal growth factor receptor (EGFR) mutation status predicting model. Methods: We retrospectively collected 168 patients with non–small cell lung cancer (NSCLC), who underwent both computed tomography (CT) examination and EGFR test. Using the radiomics features extracted from the CT images, an ensemble model was established with four individual classifiers: logistic regression (LR), support vector machine (SVM), random forest (RF), and extreme gradient boosting (XGBoost). The synthetic minority oversampling technique (SMOTE) was also used to decrease the influence of data imbalance. The performances of the predicting model were evaluated using the area under the curve (AUC). Results: Based on the 26 radiomics features after feature selection, the SVM performed best (AUCs of 0.8634 and 0.7885 on the training and test sets, respectively) among four individual classifiers. The ensemble model of RF, XGBoost, and LR achieved the best performance (AUCs of 0.8465 and 0.8654 on the training and test sets, respectively). Conclusion: Ensemble learning can improve the model performance in predicting the EGFR mutation status of patients with NSCLC, showing potential value in clinical practice.
Collapse
Affiliation(s)
- Youdan Feng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Fan Song
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Peng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Guangda Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Tianyi Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiangyu Zhao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Chenbin Ma
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yangyang Sun
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiao Song
- School of Medical Imaging, Shanxi Medical University, Taiyuan, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies, National University of Defense Technology, Changsha, China
| | - Fei Liu
- Beijing Advanced Information and Industrial Technology Research Institute, Beijing Information Science and Technology University, Beijing, China
| | - Guanglei Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Guanglei Zhang,
| |
Collapse
|
33
|
Fan Y, Dong Y, Wang H, Wang H, Sun X, Wang X, Zhao P, Luo Y, Jiang X. Development and externally validate MRI-based nomogram to assess EGFR and T790M mutations in patients with metastatic lung adenocarcinoma. Eur Radiol 2022; 32:6739-6751. [PMID: 35729427 DOI: 10.1007/s00330-022-08955-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES This study aims to explore values of multi-parametric MRI-based radiomics for detecting the epidermal growth factor receptor (EGFR) mutation and resistance (T790M) mutation in lung adenocarcinoma (LA) patients with spinal metastasis. METHODS This study enrolled a group of 160 LA patients from our hospital (between Jan. 2017 and Feb. 2021) to build a primary cohort. An external cohort was developed with 32 patients from another hospital (between Jan. 2017 and Jan. 2021). All patients underwent spinal MRI (including T1-weighted (T1W) and T2-weighted fat-suppressed (T2FS)) scans. Radiomics features were extracted from the metastasis for each patient and selected to develop radiomics signatures (RSs) for detecting the EGFR and T790M mutations. The clinical-radiomics nomogram models were constructed with RSs and important clinical parameters. The receiver operating characteristics (ROC) curve was used to evaluate the predication capabilities of each model. Calibration and decision curve analyses (DCA) were constructed to verify the performance of the models. RESULTS For detecting the EGFR and T790M mutation, the developed RSs comprised 9 and 4 most important features, respectively. The constructed nomogram models incorporating RSs and smoking status showed favorite prediction efficacy, with AUCs of 0.849 (Sen = 0.685, Spe = 0.885), 0.828 (Sen = 0.964, Spe = 0.692), and 0.778 (Sen = 0.611, Spe = 0.929) in the training, internal validation, and external validation sets for detecting the EGFR mutation, respectively, and with AUCs of 0.0.842 (Sen = 0.750, Spe = 0.867), 0.823 (Sen = 0.667, Spe = 0.938), and 0.800 (Sen = 0.875, Spe = 0.800) in the training, internal validation, and external validation sets for detecting the T790M mutation, respectively. CONCLUSIONS Radiomics features from the spinal metastasis were predictive on both EGFR and T790M mutations. The constructed nomogram models can be potentially considered as new markers to guild treatment management in LA patients with spinal metastasis. KEY POINTS • To our knowledge, this study was the first approach to detect the EGFR T790M mutation based on spinal metastasis in patients with lung adenocarcinoma. • We identified 13 MRI features that were strongly associated with the EGFR T790M mutation. • The proposed nomogram models can be considered as potential new markers for detecting EGFR and T790M mutations based on spinal metastasis.
Collapse
Affiliation(s)
- Ying Fan
- School of Intelligent Medicine, China Medical University, Liaoning, 110122, People's Republic of China
| | - Yue Dong
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Hongbo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Xinyan Sun
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Xiaoyu Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Peng Zhao
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, 110042, People's Republic of China
| | - Xiran Jiang
- School of Intelligent Medicine, China Medical University, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
34
|
Cao R, Pang Z, Wang X, Du Z, Chen H, Liu J, Yue Z, Wang H, Luo Y, Jiang X. Radiomics evaluates the EGFR mutation status from the brain metastasis: a multi-center study. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac7192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/19/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Objective. To develop and externally validate habitat-based MRI radiomics for preoperative prediction of the EGFR mutation status based on brain metastasis (BM) from primary lung adenocarcinoma (LA). Approach. We retrospectively reviewed 150 and 38 patients from hospital 1 and hospital 2 between January 2017 and December 2021 to form a primary and an external validation cohort, respectively. Radiomics features were calculated from the whole tumor (W), tumor active area (TAA) and peritumoral oedema area (POA) in the contrast-enhanced T1-weighted (T1CE) and T2-weighted (T2W) MRI image. The least absolute shrinkage and selection operator was applied to select the most important features and to develop radiomics signatures (RSs) based on W (RS-W), TAA (RS-TAA), POA (RS-POA) and in combination (RS-Com). The area under receiver operating characteristic curve (AUC) and accuracy analysis were performed to assess the performance of radiomics models. Main results. RS-TAA and RS-POA outperformed RS-W in terms of AUC, ACC and sensitivity. The multi-region combined RS-Com showed the best prediction performance in the primary validation (AUCs, RS-Com versus RS-W versus RS-TAA versus RS-POA, 0.901 versus 0.699 versus 0.812 versus 0.883) and external validation (AUCs, RS-Com versus RS-W versus RS-TAA versus RS-POA, 0.900 versus 0.637 versus 0.814 versus 0.842) cohort. Significance. The developed habitat-based radiomics models can accurately detect the EGFR mutation in patients with BM from primary LA, and may provide a preoperative basis for personal treatment planning.
Collapse
|
35
|
Multiple instance learning for lung pathophysiological findings detection using CT scans. Med Biol Eng Comput 2022; 60:1569-1584. [PMID: 35386027 DOI: 10.1007/s11517-022-02526-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/17/2022] [Indexed: 10/18/2022]
Abstract
Lung diseases affect the lives of billions of people worldwide, and 4 million people, each year, die prematurely due to this condition. These pathologies are characterized by specific imagiological findings in CT scans. The traditional Computer-Aided Diagnosis (CAD) approaches have been showing promising results to help clinicians; however, CADs normally consider a small part of the medical image for analysis, excluding possible relevant information for clinical evaluation. Multiple Instance Learning (MIL) approach takes into consideration different small pieces that are relevant for the final classification and creates a comprehensive analysis of pathophysiological changes. This study uses MIL-based approaches to identify the presence of lung pathophysiological findings in CT scans for the characterization of lung disease development. This work was focus on the detection of the following: Fibrosis, Emphysema, Satellite Nodules in Primary Lesion Lobe, Nodules in Contralateral Lung and Ground Glass, being Fibrosis and Emphysema the ones with more outstanding results, reaching an Area Under the Curve (AUC) of 0.89 and 0.72, respectively. Additionally, the MIL-based approach was used for EGFR mutation status prediction - the most relevant oncogene on lung cancer, with an AUC of 0.69. The results showed that this comprehensive approach can be a useful tool for lung pathophysiological characterization.
Collapse
|
36
|
Machine Learning-Based Radiomics for Prediction of Epidermal Growth Factor Receptor Mutations in Lung Adenocarcinoma. DISEASE MARKERS 2022; 2022:2056837. [PMID: 35578691 PMCID: PMC9107363 DOI: 10.1155/2022/2056837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 12/20/2022]
Abstract
Identifying an epidermal growth factor receptor (EGFR) mutation is important because EGFR tyrosine kinase inhibitors are the first-line treatment of choice for patients with EGFR mutation-positive lung adenocarcinomas (LUAC). This study is aimed at developing and validating a radiomics-based machine learning (ML) approach to identify EGFR mutations in patients with LUAC. We retrospectively collected data from 201 patients with positive EGFR mutation LUAC (140 in the training cohort and 61 in the validation cohort). We extracted 1316 radiomics features from preprocessed CT images and selected 14 radiomics features and 1 clinical feature which were most relevant to mutations through filter method. Subsequently, we built models using 7 ML approaches and established the receiver operating characteristic (ROC) curve to assess the discriminating performance of these models. In terms of predicting EGFR mutation, the model derived from radiomics features and combined models (radiomics features and relevant clinical factors) had an AUC of 0.79 (95% confidence interval (CI): 0.77-0.82), 0.86 (0.87-0.88), respectively. Our study offers a radiomics-based ML model using filter methods to detect the EGFR mutation in patients with LUAC. This convenient and low-cost method may be of help to noninvasively identify patients before obtaining tumor sample for molecule testing.
Collapse
|
37
|
Wang J, Falkson SR, Guo HH. Radiopaque Recreations of Lung Pathologies From Clinical Computed Tomography Images Using Potassium Iodide Inkjet 3-dimensional Printing: Proof of Concept. J Thorac Imaging 2022; 37:146-153. [PMID: 34334783 DOI: 10.1097/rti.0000000000000607] [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: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to develop a 3-dimensional (3D) printing method to create computed tomography (CT) realistic phantoms of lung cancer nodules and lung parenchymal disease from clinical CT images. MATERIALS AND METHODS Low-density paper was used as substrate material for inkjet printing with potassium iodide solution to reproduce phantoms that mimic the CT attenuation of lung parenchyma. The relationship between grayscale values and the corresponding CT numbers of prints was first established through the derivation of exponential fitted equation from scanning data. Next, chest CTs from patients with early-stage lung cancer and coronavirus disease 2019 (COVID-19) pneumonia were chosen for 3D printing. CT images of original lung nodule and the 3D-printed nodule phantom were compared based on pixel-to-pixel correlation and radiomic features. RESULTS CT images of part-solid lung cancer and 3D-printed nodule phantom showed both high visual similarity and quantitative correlation. R2 values from linear regressions of pixel-to-pixel correlations between 5 sets of patient and 3D-printed image pairs were 0.92, 0.94, 0.86, 0.85, and 0.83, respectively. Comparison of radiomic measures between clinical CT and printed models demonstrated 6.1% median difference, with 25th and 75th percentile range at 2.4% and 15.2% absolute difference, respectively. The densities and parenchymal morphologies from COVID-19 pneumonia CT images were well reproduced in the 3D-printed phantom scans. CONCLUSION The 3D printing method presented in this work facilitates creation of CT-realistic reproductions of lung cancer and parenchymal disease from individual patient scans with microbiological and pathology confirmation.
Collapse
Affiliation(s)
- Jia Wang
- Environmental Health and Safety, Stanford University
| | | | - H Henry Guo
- Department of Radiology, Stanford Medical Center, Palo Alto, CA
| |
Collapse
|
38
|
Wang Q, Dong Y, Xiao T, Zhang S, Yu J, Li L, Zhang Q, Wang Y, Xiao Y, Wang W. Prediction of programmed cell death protein 1 in hepatocellular carcinoma patients using radiomics analysis with radiofrequency-based ultrasound multifeature maps. Biomed Eng Online 2022; 21:24. [PMID: 35413926 PMCID: PMC9006564 DOI: 10.1186/s12938-021-00927-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: 02/01/2021] [Accepted: 08/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study explored the feasibility of radiofrequency (RF)-based radiomics analysis techniques for the preoperative prediction of programmed cell death protein 1 (PD-1) in patients with hepatocellular carcinoma (HCC). METHODS The RF-based radiomics analysis method used ultrasound multifeature maps calculated from the RF signals of HCC patients, including direct energy attenuation (DEA) feature map, skewness of spectrum difference (SSD) feature map, and noncentrality parameter S of the Rician distribution (NRD) feature map. From each of the above ultrasound maps, 345 high-throughput radiomics features were extracted. Then, the useful radiomics features were selected by the sparse representation method and input into support vector machine (SVM) classifier for PD-1 prediction. RESULTS AND CONCLUSION Among all the RF-based prediction models and the ultrasound grayscale comparative model, the RF-based model using all of the three ultrasound feature maps had the highest prediction accuracy (ACC) and area under the curve (AUC), which were 92.5% and 94.23%, respectively. The method proposed in this paper is effective for the meaningful feature extraction of RF signals and can effectively predict PD-1 in patients with HCC.
Collapse
Affiliation(s)
- Qingmin Wang
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Yi Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Tianlei Xiao
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Shiquan Zhang
- Institute of Biomedical and Health Engineering Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, University Town, Shenzhen, 518055, China
| | - Jinhua Yu
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Leyin Li
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Qi Zhang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Yang Xiao
- Institute of Biomedical and Health Engineering Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, University Town, Shenzhen, 518055, China.
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
39
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
40
|
Silva F, Pereira T, Neves I, Morgado J, Freitas C, Malafaia M, Sousa J, Fonseca J, Negrão E, Flor de Lima B, Correia da Silva M, Madureira AJ, Ramos I, Costa JL, Hespanhol V, Cunha A, Oliveira HP. Towards Machine Learning-Aided Lung Cancer Clinical Routines: Approaches and Open Challenges. J Pers Med 2022; 12:480. [PMID: 35330479 PMCID: PMC8950137 DOI: 10.3390/jpm12030480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 12/15/2022] Open
Abstract
Advancements in the development of computer-aided decision (CAD) systems for clinical routines provide unquestionable benefits in connecting human medical expertise with machine intelligence, to achieve better quality healthcare. Considering the large number of incidences and mortality numbers associated with lung cancer, there is a need for the most accurate clinical procedures; thus, the possibility of using artificial intelligence (AI) tools for decision support is becoming a closer reality. At any stage of the lung cancer clinical pathway, specific obstacles are identified and "motivate" the application of innovative AI solutions. This work provides a comprehensive review of the most recent research dedicated toward the development of CAD tools using computed tomography images for lung cancer-related tasks. We discuss the major challenges and provide critical perspectives on future directions. Although we focus on lung cancer in this review, we also provide a more clear definition of the path used to integrate AI in healthcare, emphasizing fundamental research points that are crucial for overcoming current barriers.
Collapse
Affiliation(s)
- Francisco Silva
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- FCUP—Faculty of Science, University of Porto, 4169-007 Porto, Portugal
| | - Tania Pereira
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
| | - Inês Neves
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- ICBAS—Abel Salazar Biomedical Sciences Institute, University of Porto, 4050-313 Porto, Portugal
| | - Joana Morgado
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
| | - Cláudia Freitas
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
- FMUP—Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Mafalda Malafaia
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- FEUP—Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Joana Sousa
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
| | - João Fonseca
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- FEUP—Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Eduardo Negrão
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
| | - Beatriz Flor de Lima
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
| | - Miguel Correia da Silva
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
| | - António J. Madureira
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
- FMUP—Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Isabel Ramos
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
- FMUP—Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - José Luis Costa
- FMUP—Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Venceslau Hespanhol
- CHUSJ—Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (C.F.); (E.N.); (B.F.d.L.); (M.C.d.S.); (A.J.M.); (I.R.); (V.H.)
- FMUP—Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - António Cunha
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- UTAD—University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Hélder P. Oliveira
- INESC TEC—Institute for Systems and Computer Engineering, Technology and Science, 4200-465 Porto, Portugal; (I.N.); (J.M.); (M.M.); (J.S.); (J.F.); (A.C.); (H.P.O.)
- FCUP—Faculty of Science, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
41
|
Cao R, Dong Y, Wang X, Ren M, Wang X, Zhao N, Yu T, Zhang L, Luo Y, Cui EN, Jiang X. MRI-Based Radiomics Nomogram as a Potential Biomarker to Predict the EGFR Mutations in Exon 19 and 21 Based on Thoracic Spinal Metastases in Lung Adenocarcinoma. Acad Radiol 2022; 29:e9-e17. [PMID: 34332860 DOI: 10.1016/j.acra.2021.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 01/17/2023]
Abstract
RATIONALE AND OBJECTIVES Preoperative identifications of epidermal growth factor receptor (EGFR) mutation subtypes based on the MRI image of spinal metastases are needed to provide individualized therapy, but has not been previously investigated. This study aims to develop and evaluate an MRI-based radiomics nomogram for differentiating the exon 19 and 21 in EGFR mutation from spinal bone metastases in patients with primary lung adenocarcinoma. MATERIALS AND METHODS A total of 76 patients underwent T1-weighted and T2-weighted fat-suppressed MRI scans were enrolled in this study, 38 were positive for EGFR mutation in exon 19 and 38 were in exon 21.MRI imaging features were extracted and selected from each MRI pulse sequence, and used to form the radiomics signature. A radiomics nomogram was developed integrating the radiomics signature and important clinical factors with receiver operating characteristic, calibration and decision curve analysis to assess the nomogram. Clinical characteristics were analyzed with Mann-Whitney U and Chi-Square tests to identify the most important factors. RESULTS A total of 6 features were selected as the most discriminative predictors from the two MRI pulse sequences. The nomogram integrating the combined radiomics signature, age and CEA level generated good prediction performance in the training (AUCs, nomogram vs. combined radiomics signature vs. clinical model, 0.90 vs. 0.87 vs. 0.59) and validation (AUCs, nomogram vs. combined radiomics signature vs. clinical model, 0.88 vs. 0.86 vs. 0.72) cohort. DCA analysis confirmed the potential clinical utility of the nomogram. CONCLUSION This study demonstrated that MRI features from spinal bone metastases can be used to prognosticate EGFR mutation subtypes in exon 19 and 21. The developed pre-treatment nomogram can potentially guide treatments for lung adenocarcinoma patients.
Collapse
|
42
|
Zhang X, Zhang Y, Zhang G, Qiu X, Tan W, Yin X, Liao L. Deep Learning With Radiomics for Disease Diagnosis and Treatment: Challenges and Potential. Front Oncol 2022; 12:773840. [PMID: 35251962 PMCID: PMC8891653 DOI: 10.3389/fonc.2022.773840] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
The high-throughput extraction of quantitative imaging features from medical images for the purpose of radiomic analysis, i.e., radiomics in a broad sense, is a rapidly developing and emerging research field that has been attracting increasing interest, particularly in multimodality and multi-omics studies. In this context, the quantitative analysis of multidimensional data plays an essential role in assessing the spatio-temporal characteristics of different tissues and organs and their microenvironment. Herein, recent developments in this method, including manually defined features, data acquisition and preprocessing, lesion segmentation, feature extraction, feature selection and dimension reduction, statistical analysis, and model construction, are reviewed. In addition, deep learning-based techniques for automatic segmentation and radiomic analysis are being analyzed to address limitations such as rigorous workflow, manual/semi-automatic lesion annotation, and inadequate feature criteria, and multicenter validation. Furthermore, a summary of the current state-of-the-art applications of this technology in disease diagnosis, treatment response, and prognosis prediction from the perspective of radiology images, multimodality images, histopathology images, and three-dimensional dose distribution data, particularly in oncology, is presented. The potential and value of radiomics in diagnostic and therapeutic strategies are also further analyzed, and for the first time, the advances and challenges associated with dosiomics in radiotherapy are summarized, highlighting the latest progress in radiomics. Finally, a robust framework for radiomic analysis is presented and challenges and recommendations for future development are discussed, including but not limited to the factors that affect model stability (medical big data and multitype data and expert knowledge in medical), limitations of data-driven processes (reproducibility and interpretability of studies, different treatment alternatives for various institutions, and prospective researches and clinical trials), and thoughts on future directions (the capability to achieve clinical applications and open platform for radiomics analysis).
Collapse
Affiliation(s)
- Xingping Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Yanchun Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Guijuan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, China
| | - Xiaoxia Yin
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| |
Collapse
|
43
|
Xia T, Kumar A, Fulham M, Feng D, Wang Y, Kim EY, Jung Y, Kim J. Fused feature signatures to probe tumour radiogenomics relationships. Sci Rep 2022; 12:2173. [PMID: 35140267 PMCID: PMC8828715 DOI: 10.1038/s41598-022-06085-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: 06/18/2021] [Accepted: 01/14/2022] [Indexed: 11/09/2022] Open
Abstract
Radiogenomics relationships (RRs) aims to identify statistically significant correlations between medical image features and molecular characteristics from analysing tissue samples. Previous radiogenomics studies mainly relied on a single category of image feature extraction techniques (ETs); these are (i) handcrafted ETs that encompass visual imaging characteristics, curated from knowledge of human experts and, (ii) deep ETs that quantify abstract-level imaging characteristics from large data. Prior studies therefore failed to leverage the complementary information that are accessible from fusing the ETs. In this study, we propose a fused feature signature (FFSig): a selection of image features from handcrafted and deep ETs (e.g., transfer learning and fine-tuning of deep learning models). We evaluated the FFSig's ability to better represent RRs compared to individual ET approaches with two public datasets: the first dataset was used to build the FFSig using 89 patients with non-small cell lung cancer (NSCLC) comprising of gene expression data and CT images of the thorax and the upper abdomen for each patient; the second NSCLC dataset comprising of 117 patients with CT images and RNA-Seq data and was used as the validation set. Our results show that our FFSig encoded complementary imaging characteristics of tumours and identified more RRs with a broader range of genes that are related to important biological functions such as tumourigenesis. We suggest that the FFSig has the potential to identify important RRs that may assist cancer diagnosis and treatment in the future.
Collapse
Affiliation(s)
- Tian Xia
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Ashnil Kumar
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael Fulham
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Dagan Feng
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Eun Young Kim
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Younhyun Jung
- School of Computing, Gachon University, Seongnam, Republic of Korea
| | - Jinman Kim
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
44
|
Liu Q, Hu P. Extendable and explainable deep learning for pan-cancer radiogenomics research. Curr Opin Chem Biol 2022; 66:102111. [PMID: 34999476 DOI: 10.1016/j.cbpa.2021.102111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022]
Abstract
Radiogenomics is a field where medical images and genomic profiles are jointly analyzed to answer critical clinical questions. Specifically, people want to identify non-invasive imaging biomarkers that are associated with both genomic features and clinical outcomes. Deep learning is an advanced computer science technique that has been applied in many fields, including medical image and genomic data analysis. This review summarizes the current state of deep learning in pan-cancer radiogenomic research, discusses its limitations, and indicates the potential future directions. Traditional machine learning in radiomics, genomics, and radiogenomics have also been briefly discussed. We also summarize the main pan-cancer radiogenomic research resources. Two characteristics of deep learning are emphasized when discussing its application to pan-cancer radiogenomics, which are extendibility and explainability.
Collapse
Affiliation(s)
- Qian Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada; Department of Computer Science, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada; Department of Statistics, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada.
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada; Department of Computer Science, University of Manitoba, Winnipeg, Manitoba, R3E 0W3, Canada.
| |
Collapse
|
45
|
Tang J, Jiang S, Ma J, Xi X, Li H, Wang L, Zhang B. Nomogram based on radiomics analysis of ultrasound images can improve preoperative BRAF mutation diagnosis for papillary thyroid microcarcinoma. Front Endocrinol (Lausanne) 2022; 13:915135. [PMID: 36060960 PMCID: PMC9437521 DOI: 10.3389/fendo.2022.915135] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The preoperative identification of BRAF mutation could assist to make appropriate treatment strategies for patients with papillary thyroid microcarcinoma (PTMC). This study aimed to establish an ultrasound (US) radiomics nomogram for the assessment of BRAF status. METHODS A total of 328 PTMC patients at the China-Japan Friendship Hospital between February 2019 and November 2021 were enrolled in this study. They were randomly divided into training (n = 232) and validation (n = 96) cohorts. Radiomics features were extracted from the US images. The least absolute shrinkage and selection operator (LASSO) regression was applied to select the BRAF status-related features and calculate the radiomics score (Rad-score). Univariate and multivariate logistic regression analyses were subsequently performed to identify the independent factors among Rad-score and conventional US features. The US radiomics nomogram was established and its predictive performance was evaluated via discrimination, calibration, and clinical usefulness in the training and validation sets. RESULTS Multivariate analysis indicated that the Rad-score, composition, and aspect ratio were independent predictive factors of BRAF status. The US radiomics nomogram which incorporated the three variables showed good calibration. The discrimination of the US radiomics nomogram showed better discriminative ability than the conventional US model both in the training set (AUC 0.685 vs. 0.592) and validation set (AUC 0.651 vs. 0.622). Decision curve analysis indicated the superior clinical applicability of the nomogram compared to the conventional US model. CONCLUSIONS The US radiomics nomogram displayed better performance than the conventional US model in predicting BRAF mutation in patients with PTMC.
Collapse
Affiliation(s)
- Jiajia Tang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
| | - Shitao Jiang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaojiao Ma
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
| | - Xuehua Xi
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
| | - Huilin Li
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
| | - Liangkai Wang
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Bo Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Ultrasound, China-Japan Friendship Hospital, National Center for Respiratory Medicine, National Clinical Research Centerfor Respiratory Diseases, Institute of Respiratory Medicine of Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Bo Zhang,
| |
Collapse
|
46
|
Tsai YM, Huang TW, Lin KH, Kuo YS, Lin YC, Chien YH, Chou HP, Chen YY, Huang HK, Wu TH, Chang H, Lee SC. Clinical significance of epidermal growth factor receptor mutations in resected stage IA non-small cell lung cancer. FORMOSAN JOURNAL OF SURGERY 2022. [DOI: 10.4103/fjs.fjs_104_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
47
|
Fan Y, Dong Y, Yang H, Chen H, Yu Y, Wang X, Wang X, Yu T, Luo Y, Jiang X. Subregional radiomics analysis for the detection of the EGFR mutation on thoracic spinal metastases from lung cancer. Phys Med Biol 2021; 66. [PMID: 34633298 DOI: 10.1088/1361-6560/ac2ea7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/11/2021] [Indexed: 01/20/2023]
Abstract
The present study intended to use radiomic analysis of spinal metastasis subregions to detect epidermal growth factor receptor (EGFR) mutation. In total, 94 patients with thoracic spinal metastasis originated from primary lung adenocarcinoma (2017-2020) were studied. All patients underwent T1-weighted (T1W) and T2 fat-suppressed (T2FS) MRI scans. The spinal metastases (tumor region) were subdivided into phenotypically consistent subregions based on patient- and population-level clustering: Three subregions, S1, S2 and S3, and the total tumor region. Radiomics features were extracted from each subregion and from the whole tumor region as well. Least shrinkage and selection operator (LASSO) regression were used for feature selection and radiomics signature definition. Detection performance of S3 was better than all other regions using T1W (AUCs, S1 versus S2 versus S3 versus whole tumor, 0.720 versus 0.764 versus 0.786 versus 0.758) and T2FS (AUCs, S1 versus S2 versus S3 versus whole tumor, 0.791 versus 0.708 versus 0.838 versus 0.797) MRI. The multi-regional radiomics signature derived from the joint of inner subregion S3 from T1W and T2FS MRI achieved the best detection capabilities with AUCs of 0.879 (ACC = 0.774, SEN = 0.838, SPE = 0.840) and 0.777 (ACC = 0.688, SEN = 0.947, SPE = 0.615) in the training and test sets, respectively. Our study revealed that MRI-based radiomic analysis of spinal metastasis subregions has the potential to detect the EGFR mutation in patients with primary lung adenocarcinoma.
Collapse
Affiliation(s)
- Ying Fan
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yue Dong
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Huazhe Yang
- Department of Biophysics, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yalian Yu
- Department of Otorhinolaryngology, the First Affiliated Hospital of China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaoyu Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xinling Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Tao Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| |
Collapse
|
48
|
Le NQK, Kha QH, Nguyen VH, Chen YC, Cheng SJ, Chen CY. Machine Learning-Based Radiomics Signatures for EGFR and KRAS Mutations Prediction in Non-Small-Cell Lung Cancer. Int J Mol Sci 2021; 22:ijms22179254. [PMID: 34502160 PMCID: PMC8431041 DOI: 10.3390/ijms22179254] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022] Open
Abstract
Early identification of epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations is crucial for selecting a therapeutic strategy for patients with non-small-cell lung cancer (NSCLC). We proposed a machine learning-based model for feature selection and prediction of EGFR and KRAS mutations in patients with NSCLC by including the least number of the most semantic radiomics features. We included a cohort of 161 patients from 211 patients with NSCLC from The Cancer Imaging Archive (TCIA) and analyzed 161 low-dose computed tomography (LDCT) images for detecting EGFR and KRAS mutations. A total of 851 radiomics features, which were classified into 9 categories, were obtained through manual segmentation and radiomics feature extraction from LDCT. We evaluated our models using a validation set consisting of 18 patients derived from the same TCIA dataset. The results showed that the genetic algorithm plus XGBoost classifier exhibited the most favorable performance, with an accuracy of 0.836 and 0.86 for detecting EGFR and KRAS mutations, respectively. We demonstrated that a noninvasive machine learning-based model including the least number of the most semantic radiomics signatures could robustly predict EGFR and KRAS mutations in patients with NSCLC.
Collapse
Affiliation(s)
- Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan;
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 106, Taiwan
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (N.Q.K.L.); (S.-J.C.); Tel.: +886-02-66382736 (ext. 1992) (N.Q.K.L.)
| | - Quang Hien Kha
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (Q.H.K.); (V.H.N.)
| | - Van Hiep Nguyen
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (Q.H.K.); (V.H.N.)
- Oncology Center, Bai Chay Hospital, Quang Ninh 20000, Vietnam
| | - Yung-Chieh Chen
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 11031, Taiwan;
| | - Sho-Jen Cheng
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 11031, Taiwan;
- Correspondence: (N.Q.K.L.); (S.-J.C.); Tel.: +886-02-66382736 (ext. 1992) (N.Q.K.L.)
| | - Cheng-Yu Chen
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan;
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 106, Taiwan
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 11031, Taiwan;
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
49
|
Tunali I, Gillies RJ, Schabath MB. Application of Radiomics and Artificial Intelligence for Lung Cancer Precision Medicine. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039537. [PMID: 33431509 PMCID: PMC8288444 DOI: 10.1101/cshperspect.a039537] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Medical imaging is the standard-of-care for early detection, diagnosis, treatment planning, monitoring, and image-guided interventions of lung cancer patients. Most medical images are stored digitally in a standardized Digital Imaging and Communications in Medicine format that can be readily accessed and used for qualitative and quantitative analysis. Over the several last decades, medical images have been shown to contain complementary and interchangeable data orthogonal to other sources such as pathology, hematology, genomics, and/or proteomics. As such, "radiomics" has emerged as a field of research that involves the process of converting standard-of-care images into quantitative image-based data that can be merged with other data sources and subsequently analyzed using conventional biostatistics or artificial intelligence (AI) methods. As radiomic features capture biological and pathophysiological information, these quantitative radiomic features have shown to provide rapid and accurate noninvasive biomarkers for lung cancer risk prediction, diagnostics, prognosis, treatment response monitoring, and tumor biology. In this review, radiomics and emerging AI methods in lung cancer research are highlighted and discussed including advantages, challenges, and pitfalls.
Collapse
Affiliation(s)
- Ilke Tunali
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
50
|
A radiomic signature model to predict the chemoradiation-induced alteration in tumor-infiltrating CD8 + cells in locally advanced rectal cancer. Radiother Oncol 2021; 162:124-131. [PMID: 34265357 DOI: 10.1016/j.radonc.2021.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND PURPOSE Regarding the altered tumor immune status following cytotoxic treatment, this study aims to develop a radiomic signature to predict CD8+ tumor-infiltrating lymphocyte (TIL) density changes in chemoradiotherapy (CRT) of rectal cancer. MATERIALS AND METHODS We used the magnetic resonance imaging (MRI) and immunohistochemistry data before and after neoadjuvant CRT. The discovery datasets consisted of pre-CRT dataset A1 (n = 113), post-CRT datasets A2 (n = 32; predominance of tumor) and A3 (n = 20; pure fibrosis). The developed model was validated in dataset B (n = 28). Thirty-eight radiomic features from T2-weighted MRI scans were incorporated into the least absolute shrinkage and selection operator method. RESULTS In pre-CRT dataset A1, the area under the receiver operating characteristic curve (AUC) values of radiomic score for predicting CD8+ TILs were 0.760 and 0.729 for training and validation subsets, respectively. A significant correlation was observed between the signature and CD8+ TIL density in the post-CRT dataset A2 (Pearson's R = -0.372, P = 0.036), whereas no association was found in dataset A3 (Pearson's R = -0.069, P = 0.77). The association was also observed in the validation dataset B (Pearson's R = -0.374, P = 0.049). In dataset A2, the radiomic score difference predicted changes in CD8+ TIL density (AUC = 0.824). CONCLUSION We established the MRI-derived radiomic signature for predicting CRT-induced alterations in CD8+ TILs. This study suggests the clinical utility of radiomics-immunophenotype modeling to evaluate tumor immune status following neoadjuvant chemoradiation in rectal cancer.
Collapse
|