1
|
Noortman WA, Vriens D, Bussink J, Meijer TWH, Aarntzen EHJG, Deroose CM, Lhommel R, Aide N, Le Tourneau C, de Koster EJ, Oyen WJG, Triemstra L, Ruurda JP, Vegt E, de Geus-Oei LF, van Velden FHP. Multicollinearity and redundancy of the PET radiomic feature set. Eur Radiol 2025:10.1007/s00330-025-11637-7. [PMID: 40332568 DOI: 10.1007/s00330-025-11637-7] [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: 10/18/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION The aim of this study was to map multicollinearity of the radiomic feature set in five independent [18F]FDG-PET cohorts with different tumour types and identify generalizable non-redundant features. METHODS Five [18F]FDG-PET radiomic cohorts were analysed: non-small cell lung carcinomas (N = 35), pheochromocytomas and paragangliomas (N = 40), head and neck squamous cell carcinomas (N = 54), [18F]FDG-positive thyroid nodules with indeterminate cytology (N = 84), and gastric carcinomas (N = 206). Lesions were delineated, and 105 radiomic features were extracted using PyRradiomics. In every cohort, Spearman's rank correlation coefficient (ρ) matrices of features were calculated to determine which features showed (very) strong (ρ > 0.7 and ρ > 0.9) correlations with any other feature in all five cohorts. Cluster analysis of an averaged correlation matrix for all cohorts was performed at a threshold of ρ = 0.7 and ρ = 0.9. For each cluster, a representative, non-redundant feature was selected. RESULTS Seventy-two and 90 out of 105 features showed a (very) strong correlation with another feature in the correlation matrix in all five cohorts. Cluster analysis resulted in 35 and 15 non-redundant features at thresholds of ρ = 0.9 and ρ = 0.7, including 6 and 3 shape features, 4 and 2 intensity features, and 25 and 10 texture features, respectively. Seventy or 90 redundant features could be omitted at these thresholds, respectively. CONCLUSION At least two-thirds of the radiomic feature set could be omitted because of strong multicollinearity in multiple independent cohorts. More redundant features could be identified using a less conservative threshold. Future research should indicate whether multicollinearity of the radiomic feature set is similar for other radiopharmaceuticals and imaging modalities. KEY POINTS Question Radiomic feature sets contain many strongly correlating features, which results in statistical challenges. Findings Analysis of the correlation matrices showed that the same radiomic features were strongly correlated in five independent [18F]FDG-PET cohorts with different tumour types. Clinical relevance At least two-thirds of the radiomic feature set could be omitted, because of strong multicollinearity. More redundant features could be identified using a less conservative threshold.
Collapse
Affiliation(s)
- Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Dennis Vriens
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tineke W H Meijer
- Department of Radiation Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
- Department of Nuclear Medicine, Eberhard Karls University, Tuebingen, Germany
| | | | - Renaud Lhommel
- Division of Nuclear Medicine and Institut de Recherche Clinique, Cliniques Universitaires Saint Luc (UCLouvain), Brussels, Belgium
| | - Nicolas Aide
- INSERM ANTICIPE U1086, François Baclesse Cancer Centre, Caen, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation, Institut Curie, Paris-Saclay University, Paris, France
| | - Elizabeth J de Koster
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, The Netherlands
- Department of Biomedical Sciences and Humanitas Clinical and Research Centre, Department of Nuclear Medicine, Humanitas University, Milan, Italy
| | - Lianne Triemstra
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Erik Vegt
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Horne A, Abravan A, Fornacon-Wood I, O’Connor JPB, Price G, McWilliam A, Faivre-Finn C. Mastering CT-based radiomic research in lung cancer: a practical guide from study design to critical appraisal. Br J Radiol 2025; 98:653-668. [PMID: 40100283 PMCID: PMC12012345 DOI: 10.1093/bjr/tqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Radiomics is a health technology that has the potential to extract clinically meaningful biomarkers from standard of care imaging. Despite a wealth of exploratory analysis performed on scans acquired from patients with lung cancer and existing guidelines describing some of the key steps, no radiomic-based biomarker has been widely accepted. This is primarily due to limitations with methodology, data analysis, and interpretation of the available studies. There is currently a lack of guidance relating to the entire radiomic workflow from study design to critical appraisal. This guide, written with early career lung cancer researchers, describes a more complete radiomic workflow. Lung cancer image analysis is the focus due to some of the unique challenges encountered such as patient movement from breathing. The guide will focus on CT imaging as these are the most common scans performed on patients with lung cancer. The aim of this article is to support the production of high-quality research that has the potential to positively impact outcome of patients with lung cancer.
Collapse
Affiliation(s)
- Ashley Horne
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| | - Azadeh Abravan
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Isabella Fornacon-Wood
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - James P B O’Connor
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Gareth Price
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| |
Collapse
|
3
|
Nakajo M, Hirahara D, Jinguji M, Idichi T, Hirahara M, Tani A, Takumi K, Kamimura K, Ohtsuka T, Yoshiura T. Machine learning-based prognostic modeling in gallbladder cancer using clinical data and pre-treatment [ 18F]-FDG-PET-radiomic features. Jpn J Radiol 2025; 43:864-874. [PMID: 39730929 PMCID: PMC12053127 DOI: 10.1007/s11604-024-01722-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/12/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVES This study evaluates the effectiveness of machine learning (ML) models that incorporate clinical and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]-FDG)-positron emission tomography (PET)-radiomic features for predicting outcomes in gallbladder cancer patients. MATERIALS AND METHODS The study analyzed 52 gallbladder cancer patients who underwent pre-treatment [18F]-FDG-PET/CT scans between January 2011 and December 2021. Twenty-seven patients were assigned to the training cohort between January 2011 and January 2018, and the data randomly split into training (70%) and validation (30%) sets. The independent test cohort consisted of 25 patients between February 2018 and December 2021. Eight clinical features (T stage, N stage, M stage, Union for International Cancer Control [UICC] stage, histology, tumor size, carcinoembryonic antigen level, and carbohydrate antigen 19-9 level) and 49 radiomic features were used to forecast progression-free survival (PFS). Three feature selection methods were applied including the univariate statistical feature selection test method, least absolute shrinkage and selection operator Cox regression method and recursive feature elimination method, and two ML algorithms (Cox proportional hazard and random survival forest [RSF]) were employed. Predictive performance was assessed using the concordance index (C-index). RESULTS Two clinical variables (UICC stage, N stage) and three radiomic features (total lesion glycolysis, grey-level size-zone matrix_grey level non-uniformity and grey-level run-length matrix_run-length non-uniformity) were identified by the statistical feature selection method as significant for PFS prediction. The RSF model incorporating these features demonstrated strong predictive performance, with C-indices above 0.80 in both training and testing sets (training 0.81, testing 0.89). This model almost closely matched the actual and predicted progression timelines with a low mean absolute error of 1.435, a median absolute error of 0.082, and a root mean square error of 2.359. CONCLUSION This study highlights the potential of using ML approaches with clinical and pre-treatment [18F]-FDG-PET radiomic data for predicting the prognosis of gallbladder cancer.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Daisuke Hirahara
- Department of Management Planning Division, Harada Academy, 2-54-4 Higashitaniyama, Kagoshima, 890-0113, Japan
| | - Megumi Jinguji
- Department of Radiology, Nanpuh Hospital, 14-3 Nagata, Kagoshima, 892-8512, Japan
| | - Tetsuya Idichi
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atsushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Koji Takumi
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kiyohisa Kamimura
- Department of Advanced Radiological Imaging, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takao Ohtsuka
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
4
|
Du D, Shiri I, Yousefirizi F, Salmanpour MR, Lv J, Wu H, Zhu W, Zaidi H, Lu L, Rahmim A. Impact of harmonization and oversampling methods on radiomics analysis of multi-center imbalanced datasets: application to PET-based prediction of lung cancer subtypes. EJNMMI Phys 2025; 12:34. [PMID: 40192981 PMCID: PMC11977052 DOI: 10.1186/s40658-025-00750-7] [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: 07/18/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Medical imaging data frequently encounter image-generation heterogeneity and class imbalance properties, challenging strong generalized predictive performances with data-driven machine-learning methods. The purpose of this study was to investigate the impact of harmonization and oversampling methods on multi-center imbalanced datasets, with specific application to PET-based radiomics modeling for histologic subtype prediction in non-small cell lung cancer (NSCLC). METHODS The retrospective study included 245 patients with adenocarcinoma (ADC) and 78 patients with squamous cell carcinoma (SCC) from 4 centers. Utilizing 1502 radiomics features per patient, we trained, validated, and tested 4 machine-learning classifiers, to investigate the effect of no harmonization (NoH) or 4 feature harmonization methods, paired with no oversampling (NoO) or 5 oversampling methods on subtype prediction. Model performance was evaluated using the average area under the ROC curve (AUROC) and G-mean via 5 times 5-fold cross-validations. Statistical comparisons of the combined models against baseline (NoH + NoO) were performed for each fold of cross-validation using the DeLong test. RESULTS The number of cross-combinations with both AUROC and G-mean outperforming baseline in validation and testing was 15, 4, 2, and 7 (out of 29) for random forest (RF), linear discriminant analysis (LDA), logistic regression (LR), and support vector machine (SVM), respectively. ComBat harmonization combined with oversampling (SMOTE) via RF yielded better performance than baseline (AUROC and G-mean of validation: 0.725 vs. 0.608 and 0.625 vs. 0.398; testing: 0.637 vs. 0.567 and 0.506 vs. 0.287), though statistical significances were not observed. CONCLUSIONS Applying harmonization and oversampling methods in multi-center imbalanced datasets can improve NSCLC-subtype prediction, but the effect varies widely across classifiers. We have created open-source comparisons of harmonization and oversampling on different classifiers for comprehensive evaluations in different studies.
Collapse
Affiliation(s)
- Dongyang Du
- College of Computer Science, Inner Mongolia University, Hohhot, Inner Mongolia, 010021, China
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, CH-1211, Switzerland
| | - Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
| | - Mohammad R Salmanpour
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
| | - Jieqin Lv
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Huiqin Wu
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wentao Zhu
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, Zhejiang, 311121, China
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, CH-1211, Switzerland
| | - Lijun Lu
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Pazhou Lab, Guangzhou, Guangdong, 510330, China.
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| |
Collapse
|
5
|
Li S, Hu Y, Tian C, Luan J, Zhang X, Wei Q, Li X, Bian Y. Prediction of EGFR-TP53 genes co-mutations in patients with lung adenocarcinoma (LUAD) by 18F-FDG PET/CT radiomics. Clin Transl Oncol 2025; 27:1506-1515. [PMID: 39251494 DOI: 10.1007/s12094-024-03685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE This retrospective study was undertaken to assess the predictive efficacy of 18F-FDG PET/CT -derived radiomic features concerning the co-mutation status of epidermal growth factor receptor (EGFR) and TP53 in LUAD. METHODS A cohort of 150 LUAD patients underwent pretreatment 18F-FDG PET/CT scans with known mutation status of EGFR and TP53 were collected. The feature extraction based on their PET/CT images utilized the Pyradiomics package based on the 3D Slicer. The optimal radiomic features were selected through correlation analysis and the Gradient Boosting Decision Tree (GBDT) algorithm, followed by the construction of the radiomic model. The clinical model incorporated meaningful clinical variables, whereas the complex model integrated both the radiomic and clinical models. The area under the receiver operating characteristic curve (AUC) facilitated the comparison of prediction performance across the three models. The DCA gauged the clinical utility of these models. RESULTS The patient cohort was randomly allocated into a training set (n = 105) and a validation set (n = 45) in a 7:3 ratio. Eleven PET and eleven CT optimal radiomic features were selected to construct the radiomic model. The model showed a good ability to discriminate the co-occurrence of EGFR and TP53, with AUC equal to 0.850 in the training set, and 0.748 in the validation set, compared with 0.750 and 0.626 for the clinical model. The complex model exhibited the highest AUC values, with 0.880 and 0.794 in both sets, but there were no significant differences compared to the radiomic model. The DCA revealed favorable clinical value. CONCLUSION
Collapse
Affiliation(s)
- Shuheng Li
- Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Congna Tian
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiusong Luan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanzhu Bian
- Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
6
|
Safarian A, Mirshahvalad SA, Nasrollahi H, Jung T, Pirich C, Arabi H, Beheshti M. Impact of [ 18F]FDG PET/CT Radiomics and Artificial Intelligence in Clinical Decision Making in Lung Cancer: Its Current Role. Semin Nucl Med 2025; 55:156-166. [PMID: 40050131 DOI: 10.1053/j.semnuclmed.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 03/17/2025]
Abstract
Lung cancer remains one of the most prevalent cancers globally and the leading cause of cancer-related deaths, accounting for nearly one-fifth of all cancer fatalities. Fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography ([18F]FDG PET/CT) plays a vital role in assessing lung cancer and managing disease progression. While traditional PET/CT imaging relies on qualitative analysis and basic quantitative parameters, radiomics offers a more advanced approach to analyzing tumor phenotypes. Recently, radiomics has gained attention for its potential to enhance the prognostic and diagnostic capabilities of [18F]FDG PET/CT in various cancers. This review explores the expanding role of [18F]FDG PET/CT-based radiomics, particularly when integrated with artificial intelligence (AI), in managing lung cancer, especially non-small cell lung cancer (NSCLC). We review how radiomics and AI improve diagnostics, staging, tumor subtype identification, and molecular marker detection, which influence treatment decisions. Additionally, we address challenges in clinical integration, such as imaging protocol standardization, feature reproducibility, and the need for extensive prospective studies. Ultimately, radiomics and AI hold great promise for enabling more personalized and effective lung cancer treatments, potentially transforming disease management.
Collapse
Affiliation(s)
- Alireza Safarian
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Rajaie Cardiovascular Medical and Research Center, Rajaie Cardiovascular Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirshahvalad
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Joint Department of Medical Imaging, University Medical Imaging Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Hadi Nasrollahi
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Theresa Jung
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Christian Pirich
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
7
|
Richlitzki C, Manapov F, Holzgreve A, Rabe M, Werner RA, Belka C, Unterrainer M, Eze C. Advances of PET/CT in Target Delineation of Lung Cancer Before Radiation Therapy. Semin Nucl Med 2025; 55:190-201. [PMID: 40064578 DOI: 10.1053/j.semnuclmed.2025.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
In the clinical management of lung cancer, radiotherapy remains a cornerstone of multimodal treatment strategies, often used alongside surgery or in combination with systemic therapies such as chemotherapy, tyrosine kinase inhibitors, and immune checkpoint inhibitors. While conventional imaging modalities like computed tomography (CT) and magnetic resonance imaging (MRI) continue to play a central role in staging, response assessment, and radiotherapy planning, advanced imaging techniques, particularly [18F]FDG PET/CT, are being increasingly integrated into routine clinical practice. These advanced techniques address the limitations of standard imaging by providing insight into molecular and metabolic tumor characteristics, enabling precise tumor visualization, accurate target volume delineation, and early treatment response assessment. This review examines the role of radiotherapy in the multidisciplinary management of lung cancer, detailing current concepts of morphological and functional imaging for staging and treatment planning. It also highlights the growing importance of PET-based radiotherapy planning, emphasizing its contributions to target volume definition and predictive value for treatment outcomes. Recent methodological advances, including the integration of artificial intelligence (AI), radiomics, technical innovations, and novel PET ligands, are discussed, highlighting their potential to improve the precision, efficacy, and personalization of lung cancer radiotherapy planning.
Collapse
Affiliation(s)
- Cedric Richlitzki
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Ahmanson Translational Theranostics Division, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Moritz Rabe
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rudolf Alexander Werner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany; Bavarian Cancer Research Center, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany; Die Radiologie, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
8
|
Chen YH, Lue KH, Chu SC, Lin CB, Liu SH. The value of 18F-fluorodeoxyglucose positron emission tomography-based radiomics in non-small cell lung cancer. Tzu Chi Med J 2025; 37:17-27. [PMID: 39850392 PMCID: PMC11753514 DOI: 10.4103/tcmj.tcmj_124_24] [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: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 01/25/2025] Open
Abstract
Currently, the second most commonly diagnosed cancer in the world is lung cancer, and 85% of cases are non-small cell lung cancer (NSCLC). With growing knowledge of oncogene drivers and cancer immunology, several novel therapeutics have emerged to improve the prognostic outcomes of NSCLC. However, treatment outcomes remain diverse, and an accurate tool to achieve precision medicine is an unmet need. Radiomics, a method of extracting medical imaging features, is promising for precision medicine. Among all radiomic tools, 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET)-based radiomics provides distinct information on glycolytic activity and heterogeneity. In this review, we collected relevant literature from PubMed and summarized the various applications of 18F-FDG PET-derived radiomics in improving the detection of metastasis, subtyping histopathologies, characterizing driver mutations, assessing treatment response, and evaluating survival outcomes of NSCLC. Furthermore, we reviewed the values of 18F-FDG PET-based deep learning. Finally, several challenges and caveats exist in the implementation of 18F-FDG PET-based radiomics for NSCLC. Implementing 18F-FDG PET-based radiomics in clinical practice is necessary to ensure reproducibility. Moreover, basic studies elucidating the underlying biological significance of 18F-FDG PET-based radiomics are lacking. Current inadequacies hamper immediate clinical adoption; however, radiomic studies are progressively addressing these issues. 18F-FDG PET-based radiomics remains an invaluable and indispensable aspect of precision medicine for NSCLC.
Collapse
Affiliation(s)
- Yu-Hung Chen
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kun-Han Lue
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sung-Chao Chu
- School of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Bin Lin
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shu-Hsin Liu
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
9
|
Nakajo M, Hirahara D, Jinguji M, Hirahara M, Tani A, Nagano H, Takumi K, Kamimura K, Kanzaki F, Yamashita M, Yoshiura T. Applying deep learning-based ensemble model to [ 18F]-FDG-PET-radiomic features for differentiating benign from malignant parotid gland diseases. Jpn J Radiol 2025; 43:91-100. [PMID: 39254903 PMCID: PMC11717794 DOI: 10.1007/s11604-024-01649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024]
Abstract
OBJECTIVES To develop and identify machine learning (ML) models using pretreatment 2-deoxy-2-[18F]fluoro-D-glucose ([18F]-FDG)-positron emission tomography (PET)-based radiomic features to differentiate benign from malignant parotid gland diseases (PGDs). MATERIALS AND METHODS This retrospective study included 62 patients with 63 PGDs who underwent pretreatment [18F]-FDG-PET/computed tomography (CT). The lesions were assigned to the training (n = 44) and testing (n = 19) cohorts. In total, 49 [18F]-FDG-PET-based radiomic features were utilized to differentiate benign from malignant PGDs using five different conventional ML algorithmic models (random forest, neural network, k-nearest neighbors, logistic regression, and support vector machine) and the deep learning (DL)-based ensemble ML model. In the training cohort, each conventional ML model was constructed using the five most important features selected by the recursive feature elimination method with the tenfold cross-validation and synthetic minority oversampling technique. The DL-based ensemble ML model was constructed using the five most important features of the bagging and multilayer stacking methods. The area under the receiver operating characteristic curves (AUCs) and accuracies were used to compare predictive performances. RESULTS In total, 24 benign and 39 malignant PGDs were identified. Metabolic tumor volume and four GLSZM features (GLSZM_ZSE, GLSZM_SZE, GLSZM_GLNU, and GLSZM_ZSNU) were the five most important radiomic features. All five features except GLSZM_SZE were significantly higher in malignant PGDs than in benign ones (each p < 0.05). The DL-based ensemble ML model had the best performing classifier in the training and testing cohorts (AUC = 1.000, accuracy = 1.000 vs AUC = 0.976, accuracy = 0.947). CONCLUSIONS The DL-based ensemble ML model using [18F]-FDG-PET-based radiomic features can be useful for differentiating benign from malignant PGDs. The DL-based ensemble ML model using [18F]-FDG-PET-based radiomic features can overcome the previously reported limitation of [18F]-FDG-PET/CT scan for differentiating benign from malignant PGDs. The DL-based ensemble ML approach using [18F]-FDG-PET-based radiomic features can provide useful information for managing PGD.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Daisuke Hirahara
- Department of Management Planning Division, Harada Academy, 2-54-4 Higashitaniyama, Kagoshima, 890-0113, Japan
| | - Megumi Jinguji
- Department of Radiology, Nanpuh Hospital, 14-3 Nagata, Kagoshima, 892-8512, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atsushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Hiromi Nagano
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Koji Takumi
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kiyohisa Kamimura
- Department of Advanced Radiological Imaging, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Fumiko Kanzaki
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Masaru Yamashita
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
10
|
Erasmus LT, Strange CD, Ahuja J, Agrawal R, Shroff GS, Marom EM, Truong MT. Imaging of Lung Cancer Staging: TNM 9 Updates. Semin Ultrasound CT MR 2024; 45:410-419. [PMID: 39069273 DOI: 10.1053/j.sult.2024.07.005] [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: 07/30/2024]
Abstract
Imaging plays a key role in clinical staging of lung cancer and guiding therapy. A thorough understanding of the staging system including the nomenclature and updates is necessary to tailor treatment plans and optimize patient care. The 9th edition of the Tumor, Node, Metastasis staging system for lung cancer has no changes for T classification and subdivides N2 and M1c categories. In nodal staging, N2 splits into N2a, ipsilateral mediastinal single station involvement and N2b, ipsilateral mediastinal multiple stations involvement. In the staging of multiple extrathoracic metastases, M1c splits into M1c1, multiple extrathoracic metastases in one organ system and M1c2, multiple extrathoracic metastases in multiple organ systems. Awareness of the proposed changes in TNM-9 staging classification is essential to provide methodical and accurate imaging interpretation.
Collapse
Affiliation(s)
- Lauren T Erasmus
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX
| | - Chad D Strange
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jitesh Ahuja
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rishi Agrawal
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Girish S Shroff
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Edith M Marom
- Department of Radiology, Chaim Sheba Medical Center, Tel Aviv, Israel
| | - Mylene T Truong
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
11
|
Murchan P, Ó Broin P, Baird AM, Sheils O, P Finn S. Deep feature batch correction using ComBat for machine learning applications in computational pathology. J Pathol Inform 2024; 15:100396. [PMID: 39398947 PMCID: PMC11470259 DOI: 10.1016/j.jpi.2024.100396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Background Developing artificial intelligence (AI) models for digital pathology requires large datasets from multiple sources. However, without careful implementation, AI models risk learning confounding site-specific features in datasets instead of clinically relevant information, leading to overestimated performance, poor generalizability to real-world data, and potential misdiagnosis. Methods Whole-slide images (WSIs) from The Cancer Genome Atlas (TCGA) colon (COAD), and stomach adenocarcinoma datasets were selected for inclusion in this study. Patch embeddings were obtained using three feature extraction models, followed by ComBat harmonization. Attention-based multiple instance learning models were trained to predict tissue-source site (TSS), as well as clinical and genetic attributes, using raw, Macenko normalized, and Combat-harmonized patch embeddings. Results TSS prediction achieved high accuracy (AUROC > 0.95) with all three feature extraction models. ComBat harmonization significantly reduced the AUROC for TSS prediction, with mean AUROCs dropping to approximately 0.5 for most models, indicating successful mitigation of batch effects (e.g., CCL-ResNet50 in TCGA-COAD: Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, p < 0.001). Clinical attributes associated with TSS, such as race and treatment response, showed decreased predictability post-harmonization. Notably, the prediction of genetic features like MSI status remained robust after harmonization (e.g., MSI in TCGA-COAD: Pre-ComBat AUROC = 0.667, Post-ComBat AUROC = 0.669, p=0.952), indicating the preservation of true histological signals. Conclusion ComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates. This approach is promising for the integration of large-scale digital pathology datasets.
Collapse
Affiliation(s)
- Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
| | - Pilib Ó Broin
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
- School of Mathematical & Statistical Sciences, University of Galway, Galway H91 TK33, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- Department of Histopathology, St. James's Hospital, James's Street, Dublin D08 X4RX, Ireland
| |
Collapse
|
12
|
Ni J, Chen H, Yu L, Guo T, Zhou Y, Jiang S, Ye R, Yang X, Chu L, Chu X, Li H, Liu W, Gu Y, Yuan Z, Gong J, Zhu Z. Predicting Regional Recurrence and Prognosis in Stereotactic Body Radiation Therapy-Treated Clinical Stage I Non-small Cell Lung Cancer Using a Radiomics Model Constructed With Surgical Data. Int J Radiat Oncol Biol Phys 2024; 120:1096-1106. [PMID: 38936632 DOI: 10.1016/j.ijrobp.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/15/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Risk stratification of regional recurrence (RR) is clinically important in the design of adjuvant treatment and surveillance strategies in patients with clinical stage I non-small cell lung cancer (NSCLC) treated with stereotactic body radiation therapy (SBRT). This study aimed to develop a radiomics model predicting occult lymph node metastasis (OLNM) using surgical data and apply it to the prediction of RR in SBRT-treated early-stage NSCLC patients. METHODS AND MATERIALS Patients with clinical stage I NSCLC who underwent curative surgery with systematic lymph node dissection from January 2013 to December 2018 (the training cohort) and from January 2019 to December 2020 (the validation cohort) were included. A preoperative computed tomography-based radiomics model, a clinical feature model, and a fusion model predicting OLNM were constructed. The performance of the 3 models was quantified and compared in the training and validation cohorts. Subsequently, the radiomics model was used to predict RR in a cohort of consecutive SBRT-treated early-stage NSCLC patients from 2 academic medical centers. RESULTS A total of 769 patients were included. Eight computed tomography features were identified in the radiomics model, achieving areas under the curves of 0.85 (95% CI, 0.81-0.89) and 0.83 (95% CI, 0.80-0.88) in the training and validation cohorts, respectively. Nevertheless, adding clinical features did not improve the performance of the radiomics model. With a median follow-up of 40.0 (95% CI, 35.2-44.8) months, 32 of the 213 patients in the SBRT cohort developed RR and those in the high-risk group based on the radiomics model had a higher cumulative incidence of RR (P < .001) and shorter regional recurrence-free survival (P = .02), progression-free survival (P = .004) and overall survival (P = .006) than those in the low-risk group. CONCLUSIONS The radiomics model based on pathologically confirmed data effectively identified patients with OLNM, which may be useful in the risk stratification among SBRT-treated patients with clinical stage I NSCLC.
Collapse
Affiliation(s)
- Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Hongru Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Lu Yu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shanshan Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ruiting Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Xiao Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Haiming Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Liu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Jing Gong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| |
Collapse
|
13
|
Zhou Y, Zhou J, Cai X, Ge S, Sang S, Yang Y, Zhang B, Deng S. Integrating 18F-FDG PET/CT radiomics and body composition for enhanced prognostic assessment in patients with esophageal cancer. BMC Cancer 2024; 24:1402. [PMID: 39543534 PMCID: PMC11566154 DOI: 10.1186/s12885-024-13157-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND This study aimed to develop a predictive model utilizing radiomics and body composition features derived from 18F-FDG PET/CT scans to forecast progression-free survival (PFS) and overall survival (OS) outcomes in patients with esophageal squamous cell carcinoma (ESCC). METHODS We analyzed data from 91 patients who underwent baseline 18F-FDG PET/CT imaging. Radiomic features extracted from PET and CT images and subsequent radiomics scores (Rad-scores) were calculated. Body composition metrics were also quantified, including muscle and fat distribution at the L3 level from CT scans. Multiparametric survival models were constructed using Cox regression analysis, and their performance was assessed using the area under the time-dependent receiver operating characteristic (ROC) curve (AUC) and concordance index (C-index). RESULTS Multivariate analysis identified Rad-scorePFS (P = 0.003), sarcopenia (P < 0.001), and visceral adipose tissue index (VATI) (P < 0.001) as independent predictors of PFS. For OS, Rad-scoreOS (P = 0.001), sarcopenia (P = 0.002), VATI (P = 0.037), stage (P = 0.042), and body mass index (BMI) (P = 0.008) were confirmed as independent prognostic factors. Integration of the Rad-score with clinical variables and body composition parameters enhanced predictive accuracy, yielding C-indices of 0.810 (95% CI: 0.737-0.884) for PFS and 0.806 (95% CI: 0.720-0.891) for OS. CONCLUSIONS This study underscored the potential of combining Rad-score with clinical and body composition data to refine prognostic assessment in ESCC patients.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jin Zhou
- Department of Nuclear Medicine, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Suqian, China
| | - Xiaowei Cai
- Department of Nuclear Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Shushan Ge
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yi Yang
- Department of Nuclear Medicine, Affiliated Hospital of Medical School, Suzhou Hospital, Nanjing University, Suzhou, China.
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China.
| |
Collapse
|
14
|
Lu X, Li C, Wang S, Yin Y, Fu H, Wang H, Cheng W, Chen S. The prognostic role of 18F-FDG PET/CT-based response evaluation in children with stage 4 neuroblastoma. Eur Radiol 2024; 34:7125-7135. [PMID: 38758254 DOI: 10.1007/s00330-024-10781-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVES To evaluate the association between metabolic response on 18F-FDG PET/CT and long-term survival in children with neuroblastoma (NB). METHODS A total of 39 consecutive children with newly diagnosed stage 4 NB undergoing both 18F-FDG PET/CT imaging at baseline and after chemotherapy were retrospectively analyzed. The associations between metabolic parameters, including SUVmax of the lesion with the most intense 18F-FDG uptake at baseline (SUVb), after chemotherapy (SUVe), and the percentage change between SUVb and SUVe, and long-term survival were evaluated. RESULTS With a median follow-up of 56 months, 22 patients who had achieved complete resolution on PET (no residual 18F-FDG uptake higher than the surrounding backgrounds) after chemotherapy had superior 5-year overall survival (OS) (73.6% vs. 39.0%, p = 0.044). SUVb > 6.9 indicated significantly poorer 5-year event-free survival (EFS) (12.5% vs. 59.3%, p = 0.005), as did SUVe > 1.2 (18.8% vs. 41.7%, p = 0.041). Children with SUVe > 1.2 had shorter 5-year OS (33.9% vs. 75.0%, p = 0.018). Multivariate analysis identified SUVe > 1.2 as an independent predictor for both EFS [hazard ratio (HR), 3.479, 95% CI, 1.381-8.761, p = 0.008] and OS (HR, 6.948, 95% CI, 1.663-29.025, p = 0.008), while SUVb > 6.9 was a predictor for EFS (HR, 2.889, 95% CI, 1.064-7.842, p = 0.037). Among 11 children with both SUVb > 6.9 and SUVe > 1.2, all experienced disease progression or relapse within 2 years since diagnosis. CONCLUSION 18F-FDG PET/CT could be of useful to evaluate treatment response in children with stage 4 NB. CLINICAL RELEVANCE STATEMENT 18F-FDG PET/CT after chemotherapy exhibits prognostic significance in neuroblastoma and holds potential as an alternative imaging modality for response evaluation, especially in cases with metaiodobenzylguanidine-nonavid or persistent avid disease. KEY POINTS The prognostic value of chemotherapy response on 18F-FDG PET/CT in advanced neuroblastoma is unknown. Higher 18F-FDG uptake after chemotherapy was associated with worse long-term event-free survival and overall survival. 18F-FDG PET/CT after chemotherapy holds prognostic significance in children with stage 4 neuroblastoma.
Collapse
Affiliation(s)
- Xueyuan Lu
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoyan Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongliang Fu
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Miao C, Chen Y, Zhang H, Zhao W, Wang C, Ma Z, Zhu S, Hu X. Heterogeneity in peripheral blood immune lymphocyte subsets predicts the response of immunotherapy or chemoradiotherapy in advanced lung cancer: an analysis across different pathological types, treatment modalities and age. Front Immunol 2024; 15:1464728. [PMID: 39483483 PMCID: PMC11524866 DOI: 10.3389/fimmu.2024.1464728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background The shaping of the tumor immune microenvironment does not only rely on tumor-infiltrating lymphocytes but on the recruitment of lymphocytes in peripheral blood. Monitoring peripheral blood lymphocyte subsets level (PBLSL) can predict treatment response and prognosis with immune checkpoint inhibitors. This study investigated the heterogeneity of PBLSL in response to chemoradiotherapy (CRT) or combined with immunotherapy (CRIT) in advanced lung cancer patients. Methods 77 patients with advanced lung cancer receiving CRT or CRIT were divided into treatment-responsive and non-responsive groups based on efficacy. The study analyzed short-term efficacy and progression-free survival (PFS) according to baseline PBLSL and explored the impact under different stratifications, including treatment modality, pathology type, and age. Results In all patients, higher levels of B cells, higher CD4+/CD8+ T cell ratios, and lower CD8+ T cell levels were associated with better short-term outcomes (P = 0.0035, P = 0.044, P = 0.022). Subgroup analysis revealed that in the CRT group, higher B cell levels correlated with improved efficacy (P = 0.011) and superior PFS (P = 0.048, HR = 0.3886, 95% CI = 0.1696 to 0.8902). In the CRIT group, higher CD4+ T cell levels, lower CD8+ T cell levels, and higher CD4+/CD8+ T cell ratios were linked to better efficacy (P = 0.038, P = 0.047, P = 0.017). For adenocarcinoma patients, higher CD4+/CD8+ T cell ratios and lower CD8+ T cell levels predicted better efficacy (P = 0.0155, P = 0.0119). B cell levels were significant in squamous cell carcinoma (P = 0.0291), while no PBLSL was predictive for small cell lung cancer. Among patients under 65, higher B cell levels were linked to improved efficacy and prolonged PFS (P = 0.0036, P = 0.0332, HR = 0.4111, 95% CI = 0.1973 to 0.8563). For patients over 65, differences in CD4+ T cell levels and CD4+/CD8+ T cell ratios were significant (P = 0.0433, P = 0.0338). Conclusion PBLSL predicted efficacy and prognosis in various patient stratifications, suggesting PBLSL is a reliable predictor for CRT and CRIT in advanced lung cancer. Detecting different cellular subpopulations helps identify patients with significant treatment responses across different stratifications.
Collapse
Affiliation(s)
- Chuanwang Miao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuanji Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Clinical Medicine, Shandong First Medical University, Jinan, Shandong, China
| | - Hao Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cunliang Wang
- Department of Radiotherapy, Linyi Cancer Hospital, Linyi, Shandong, China
| | - Zeliang Ma
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Shan Zhu
- Department of Radiation Oncology, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
| | - Xudong Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
16
|
Coppes RP, van Dijk LV. Future of Team-based Basic and Translational Science in Radiation Oncology. Semin Radiat Oncol 2024; 34:370-378. [PMID: 39271272 DOI: 10.1016/j.semradonc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
To further optimize radiotherapy, a more personalized treatment towards individual patient's risk profiles, dissecting both patient-specific tumor and normal tissue response to multimodality treatments is needed. Novel developments in radiobiology, using in vitro patient-specific complex tissue resembling 3D models and multiomics approaches at a spatial single-cell level, may provide unprecedented insight into the radiation responses of tumors and normal tissue. Here, we describe the necessary team effort, including all disciplines in radiation oncology, to integrate such data into clinical prediction models and link the relatively "big data" from the clinical practice, allowing accurate patient stratification for personalized treatment approaches.
Collapse
Affiliation(s)
- R P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.; Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands..
| | - L V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
17
|
Li C, Lu X, Zhang F, Huang S, Ding L, Wang H, Chen S. Neuroblastoma with high ASPM reveals pronounced heterogeneity and poor prognosis. BMC Cancer 2024; 24:1151. [PMID: 39289658 PMCID: PMC11406734 DOI: 10.1186/s12885-024-12912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE We explored the preliminary value of abnormal spindle-like microcephaly- associated (ASPM) protein in aiding precise risk sub-stratification, prediction of metabolic heterogeneity, and prognosis of neuroblastoma (NB). METHODS This retrospective study enrolled newly diagnosed patients with NB who underwent positron emission tomography/computed tomography (PET/CT) before therapy, and tumor tissue was collected after surgery. Regression analysis was used to evaluate ASPM expression and risk stratification in patients with NB. The expression levels of ASPM, clinical information, and PET/CT text features were analyzed using univariate and multivariate survival analyses. Finally, a correlation analysis was used to explore the relationship between ASPM and tumor metabolic heterogeneity. RESULTS There were 48 patients with NB in this study (35 boys and 13 girls); 22 patients progressed and 16 died. We found that the level of ASPM was highly associated with risk stratification (OR = 5.295, 95%IC: 1.348-41.722, p = 0.021). Patients with NB and high-risk stratification with high ASPM level had a lower 3-year progression-free survival (PFS) rate (14.28%) and 1-year PFS rate (57.14%) than those with low ASPM level (57.14% and 93.75%, respectively). Using univariate and multivariate survival analyses, this study revealed that ASPM and LDH were independent risk factors for both PFS and overall survival (OS), whales GLZLM_ZLNU was only a risk factor for PFS. CONCLUSION ASPM holds promise as a novel biomarker for refining current risk stratification and predicting prognosis in neuroblastoma. Elevated levels of ASPM, LDH, and GLZLM_ZLNU may be associated with poorer survival outcomes in neuroblastoma patients.
Collapse
Affiliation(s)
- Chao Li
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xueyuan Lu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fengxian Zhang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Shuo Huang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lin Ding
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
18
|
Zhang Y, Cui Y, Liu H, Chang C, Yin Y, Wang R. Prognostic nomogram combining 18F-FDG PET/CT radiomics and clinical data for stage III NSCLC survival prediction. Sci Rep 2024; 14:20557. [PMID: 39231973 PMCID: PMC11374974 DOI: 10.1038/s41598-024-71003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
The aim of this study was to establish and validate the precision of a novel radiomics approach that integrates 18Fluorine-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)-computed tomography (CT) scan data with clinical information to improve the prognostication of survival rates in patients diagnosed with stage III Non-Small Cell Lung Cancer (NSCLC) who are not candidates for surgery. We evaluated pretreatment 18F-FDG PET-CT scans from 156 individuals diagnosed with stage III inoperable NSCLC at Shandong Cancer Hospital. These individuals were divided into two groups: a training set comprising 110 patients and an internal validation set consisting of 46 patients. By employing random forest classifier and cox proportional hazards model , we identified and utilized relevant features to create predictive models and a nomogram. The effectiveness of these models was assessed through the use of the receiver operating characteristics(ROC) curves, Kaplan-Meier (KM) curves, and the application of the nomogram. Our findings showed that the combined model, which integrates both clinical and radiomic data, outperformed those based solely on clinical or radiomic features in predicting 3-year overall survival(OS). Furthermore, calibration plots revealed a high level of agreement between predicted and actual survival times. The research successfully established a predictive radiomics model that integrates 18F-FDG PET/CT imaging with clinical indicators to enhance survival predictions for patients with stage III inoperable NSCLC.
Collapse
Affiliation(s)
- Yalin Zhang
- Department of Radiation Oncology, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China
| | - Yongbin Cui
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Huiling Liu
- Department of Radiation Oncology, Binzhou People's Hospital, Binzhou, China
| | - Cheng Chang
- Department of Nuclear Medicine, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Ruozheng Wang
- Department of Radiation Oncology, The Third Affiliated Teaching Hospital of Xinjiang Medical University, Affiliated Cancer Hospital, Urumuqi, China.
- Xinjiang Key Laboratory of Oncology, Urumqi, China.
| |
Collapse
|
19
|
Tang X, Li Y, Qian WL, Han PL, Yan WF, Yang ZG. Enhancing intracranial efficacy prediction of osimertinib in non-small cell lung cancer: a novel approach through brain MRI radiomics. Front Neurol 2024; 15:1399983. [PMID: 39281414 PMCID: PMC11395019 DOI: 10.3389/fneur.2024.1399983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Osimertinib, a third-generation EGFR-TKI, is known for its high efficacy against brain metastases (BM) in non-small cell lung cancer (NSCLC) due to its ability to penetrate the blood-brain barrier. This study aims to evaluate the use of brain MRI radiomics in predicting the intracranial efficacy to osimertinib in NSCLC patients with BM. Materials and methods This study analyzed 115 brain metastases from NSCLC patients with the EGFR-T790M mutation treated with second-line osimertinib. The primary endpoint was intracranial response, and the secondary endpoint was intracranial progression-free survival (iPFS). We performed tumor delineation, image preprocessing, and radiomics feature extraction. Using a 5-fold cross-validation strategy, we built radiomic models with eight feature selectors and eight machine learning classifiers. The models' performance was evaluated by the area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis. Results The dataset of 115 brain metastases was divided into training and validation sets in a 7:3 ratio. The radiomic model utilizing the mRMR feature selector and stepwise logistic regression classifier showed the highest predictive accuracy, with AUCs of 0.879 for the training cohort and 0.786 for the validation cohort. This model outperformed a clinical-MRI morphological model, which included age, ring enhancement, and peritumoral edema (AUC: 0.794 for the training cohort and 0.697 for the validation cohort). The radiomic model also showed strong performance in calibration and decision curve analyses. Using a radiomic-score threshold of 199, patients were classified into two groups with significantly different median iPFS (3.0 months vs. 15.4 months, p < 0.001). Conclusion This study demonstrates that MRI radiomics can effectively predict the intracranial efficacy of osimertinib in NSCLC patients with brain metastases. This approach holds promise for assisting clinicians in personalizing treatment strategies.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wen-Lei Qian
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Pei-Lun Han
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Feng Yan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Gang Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Nemoto H, Saito M, Satoh Y, Komiyama T, Marino K, Aoki S, Suzuki H, Sano N, Nonaka H, Watanabe H, Funayama S, Onishi H. Evaluation of the performance of both machine learning models using PET and CT radiomics for predicting recurrence following lung stereotactic body radiation therapy: A single-institutional study. J Appl Clin Med Phys 2024; 25:e14322. [PMID: 38436611 PMCID: PMC11244675 DOI: 10.1002/acm2.14322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/14/2024] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
PURPOSE Predicting recurrence following stereotactic body radiotherapy (SBRT) for non-small cell lung cancer provides important information for the feasibility of the individualized radiotherapy and allows to select the appropriate treatment strategy based on the risk of recurrence. In this study, we evaluated the performance of both machine learning models using positron emission tomography (PET) and computed tomography (CT) radiomic features for predicting recurrence after SBRT. METHODS Planning CT and PET images of 82 non-small cell lung cancer patients who performed SBRT at our hospital were used. First, tumors were delineated on each CT and PET of each patient, and 111 unique radiomic features were extracted, respectively. Next, the 10 features were selected using three different feature selection algorithms, respectively. Recurrence prediction models based on the selected features and four different machine learning algorithms were developed, respectively. Finally, we compared the predictive performance of each model for each recurrence pattern using the mean area under the curve (AUC) calculated following the 0.632+ bootstrap method. RESULTS The highest performance for local recurrence, regional lymph node metastasis, and distant metastasis were observed in models using Support vector machine with PET features (mean AUC = 0.646), Naive Bayes with PET features (mean AUC = 0.611), and Support vector machine with CT features (mean AUC = 0.645), respectively. CONCLUSIONS We comprehensively evaluated the performance of prediction model developed for recurrence following SBRT. The model in this study would provide information to predict the recurrence pattern and assist in making treatment strategies.
Collapse
Affiliation(s)
- Hikaru Nemoto
- Department of Advanced Biomedical ImagingUniversity of YamanashiChuoYamanashiJapan
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Masahide Saito
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Yoko Satoh
- Imaging CenterFujita Medical Innovation Center TokyoTokyoJapan
| | | | - Kan Marino
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Shinichi Aoki
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Hidekazu Suzuki
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Naoki Sano
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| | - Hotaka Nonaka
- Department of RadiologyFuji City General HospitalFujiShizuokaJapan
| | - Hiroaki Watanabe
- Department of RadiologyYamanashi Central HospitalKofuYamanashiJapan
| | - Satoshi Funayama
- Department of RadiologyHamamatsu University school of medicineHamamatsuShizuokaJapan
| | - Hiroshi Onishi
- Department of RadiologyUniversity of YamanashiChuoYamanashiJapan
| |
Collapse
|
21
|
Woodworth CF, Frota Lima LM, Bartholmai BJ, Koo CW. Imaging of Solid Pulmonary Nodules. Clin Chest Med 2024; 45:249-261. [PMID: 38816086 DOI: 10.1016/j.ccm.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Early detection with accurate classification of solid pulmonary nodules is critical in reducing lung cancer morbidity and mortality. Computed tomography (CT) remains the most widely used imaging examination for pulmonary nodule evaluation; however, other imaging modalities, such as PET/CT and MRI, are increasingly used for nodule characterization. Current advances in solid nodule imaging are largely due to developments in machine learning, including automated nodule segmentation and computer-aided detection. This review explores current multi-modality solid pulmonary nodule detection and characterization with discussion of radiomics and risk prediction models.
Collapse
Affiliation(s)
- Claire F Woodworth
- Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Livia Maria Frota Lima
- Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Brian J Bartholmai
- Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Chi Wan Koo
- Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
22
|
Lucia F, Louis T, Cousin F, Bourbonne V, Visvikis D, Mievis C, Jansen N, Duysinx B, Le Pennec R, Nebbache M, Rehn M, Hamya M, Geier M, Salaun PY, Schick U, Hatt M, Coucke P, Hustinx R, Lovinfosse P. Multicentric development and evaluation of [ 18F]FDG PET/CT and CT radiomic models to predict regional and/or distant recurrence in early-stage non-small cell lung cancer treated by stereotactic body radiation therapy. Eur J Nucl Med Mol Imaging 2024; 51:1097-1108. [PMID: 37987783 DOI: 10.1007/s00259-023-06510-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE To develop machine learning models to predict regional and/or distant recurrence in patients with early-stage non-small cell lung cancer (ES-NSCLC) after stereotactic body radiation therapy (SBRT) using [18F]FDG PET/CT and CT radiomics combined with clinical and dosimetric parameters. METHODS We retrospectively collected 464 patients (60% for training and 40% for testing) from University Hospital of Liège and 63 patients from University Hospital of Brest (external testing set) with ES-NSCLC treated with SBRT between 2010 and 2020 and who had undergone pretreatment [18F]FDG PET/CT and planning CT. Radiomic features were extracted using the PyRadiomics toolbox®. The ComBat harmonization method was applied to reduce the batch effect between centers. Clinical, radiomic, and combined models were trained and tested using a neural network approach to predict regional and/or distant recurrence. RESULTS In the training (n = 273) and testing sets (n = 191 and n = 63), the clinical model achieved moderate performances to predict regional and/or distant recurrence with C-statistics from 0.53 to 0.59 (95% CI, 0.41, 0.67). The radiomic (original_firstorder_Entropy, original_gldm_LowGrayLevelEmphasis and original_glcm_DifferenceAverage) model achieved higher predictive ability in the training set and kept the same performance in the testing sets, with C-statistics from 0.70 to 0.78 (95% CI, 0.63, 0.88) while the combined model performs moderately well with C-statistics from 0.50 to 0.62 (95% CI, 0.37, 0.69). CONCLUSION Radiomic features extracted from pre-SBRT analog and digital [18F]FDG PET/CT outperform clinical parameters in the prediction of regional and/or distant recurrence and to discuss an adjuvant systemic treatment in ES-NSCLC. Prospective validation of our models should now be carried out.
Collapse
Affiliation(s)
- François Lucia
- Radiation Oncology Department, University Hospital, Brest, France.
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France.
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
- Service de Radiothérapie, CHRU Morvan, 2 Avenue Foch, 29609 Cedex, Brest, France.
| | - Thomas Louis
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | | | - Carole Mievis
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Nicolas Jansen
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | | | - Romain Le Pennec
- Nuclear Medicine Department, University Hospital, Brest, France
- GETBO, INSERM, UMR 1304, University of Brest, UBO, Brest, France
| | - Malik Nebbache
- Radiation Oncology Department, University Hospital, Brest, France
| | - Martin Rehn
- Radiation Oncology Department, University Hospital, Brest, France
| | - Mohamed Hamya
- Radiation Oncology Department, University Hospital, Brest, France
| | - Margaux Geier
- Medical Oncology Department, University Hospital, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospital, Brest, France
- GETBO, INSERM, UMR 1304, University of Brest, UBO, Brest, France
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Philippe Coucke
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
- GIGA-CRC In Vivo Imaging, University of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
23
|
Liu Z, Luo C, Chen X, Feng Y, Feng J, Zhang R, Ouyang F, Li X, Tan Z, Deng L, Chen Y, Cai Z, Zhang X, Liu J, Liu W, Guo B, Hu Q. Noninvasive prediction of perineural invasion in intrahepatic cholangiocarcinoma by clinicoradiological features and computed tomography radiomics based on interpretable machine learning: a multicenter cohort study. Int J Surg 2024; 110:1039-1051. [PMID: 37924497 PMCID: PMC10871628 DOI: 10.1097/js9.0000000000000881] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/22/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Perineural invasion (PNI) of intrahepatic cholangiocarcinoma (ICC) is a strong independent risk factor for tumour recurrence and long-term patient survival. However, there is a lack of noninvasive tools for accurately predicting the PNI status. The authors develop and validate a combined model incorporating radiomics signature and clinicoradiological features based on machine learning for predicting PNI in ICC, and used the Shapley Additive explanation (SHAP) to visualize the prediction process for clinical application. METHODS This retrospective and prospective study included 243 patients with pathologically diagnosed ICC (training, n =136; external validation, n =81; prospective, n =26, respectively) who underwent preoperative contrast-enhanced computed tomography between January 2012 and May 2023 at three institutions (three tertiary referral centres in Guangdong Province, China). The ElasticNet was applied to select radiomics features and construct signature derived from computed tomography images, and univariate and multivariate analyses by logistic regression were used to identify the significant clinical and radiological variables with PNI. A robust combined model incorporating radiomics signature and clinicoradiological features based on machine learning was developed and the SHAP was used to visualize the prediction process. A Kaplan-Meier survival analysis was performed to compare prognostic differences between PNI-positive and PNI-negative groups and was conducted to explore the prognostic information of the combined model. RESULTS Among 243 patients (mean age, 61.2 years ± 11.0 (SD); 152 men and 91 women), 108 (44.4%) were diagnosed as PNI-positive. The radiomics signature was constructed by seven radiomics features, with areas under the curves of 0.792, 0.748, and 0.729 in the training, external validation, and prospective cohorts, respectively. Three significant clinicoradiological features were selected and combined with radiomics signature to construct a combined model using machine learning. The eXtreme Gradient Boosting exhibited improved accuracy and robustness (areas under the curves of 0.884, 0.831, and 0.831, respectively). Survival analysis showed the construction combined model could be used to stratify relapse-free survival (hazard ratio, 1.933; 95% CI: 1.093-3.418; P =0.021). CONCLUSIONS We developed and validated a robust combined model incorporating radiomics signature and clinicoradiological features based on machine learning to accurately identify the PNI statuses of ICC, and visualize the prediction process through SHAP for clinical application.
Collapse
Affiliation(s)
- Ziwei Liu
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Chun Luo
- Department of Radiology, The First People’s Hospital of Foshan
| | - Xinjie Chen
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Yanqiu Feng
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
- School of Biomedical Engineering, Southern Medical University
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology
- Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, 1023 Sha-Tai South Road, Guangzhou, China
| | - Jieying Feng
- Department of Radiology, The Sixth Affiliated Hospital, South China University of Technology, Foshan
| | - Rong Zhang
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Fusheng Ouyang
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Xiaohong Li
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Zhilin Tan
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Lingda Deng
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Yifan Chen
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Zhiping Cai
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Ximing Zhang
- Department of Radiology, The First People’s Hospital of Foshan
| | - Jiehong Liu
- School of Biomedical Engineering, Southern Medical University
| | - Wei Liu
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Baoliang Guo
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| | - Qiugen Hu
- Department of Radiology,Southern Medical University (The First People’s Hospital of Shunde)
| |
Collapse
|
24
|
Lin Y, Yang Z, Chen J, Li M, Cai Z, Wang X, Zhai T, Lin Z. A contrast-enhanced CT radiomics-based model to identify candidates for deintensified chemoradiotherapy in locoregionally advanced nasopharyngeal carcinoma patients. Eur Radiol 2024; 34:1302-1313. [PMID: 37594526 DOI: 10.1007/s00330-023-09987-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVES To develop a contrast-enhanced CT (CECT) radiomics-based model to identify locoregionally advanced nasopharyngeal carcinoma (LA-NPC) patients who would benefit from deintensified chemoradiotherapy. METHODS LA-NPC patients who received low-dose concurrent cisplatin therapy (cumulative: 150 mg/m2), were randomly divided into training and validation groups. 107 radiomics features based on the primary nasopharyngeal tumor were extracted from each pre-treatment CECT scan. Through Cox regression analysis, a radiomics model and patients' corresponding radiomics scores were created with predictive independent radiomics features. T stage (T) and radiomics score (R) were compared as predictive factors. Combining the N stage (N), a clinical model (T + N), and a substitution model (R + N) were constructed. RESULTS Training and validation groups consisted of 66 and 33 patients, respectively. Three significant independent radiomics features (flatness, mean, and gray level non-uniformity in gray level dependence matrix (GLDM-GLN)) were found. The radiomics score showed better predictive ability than the T stage (concordance index (C-index): 0.67 vs. 0.61, AUC: 0.75 vs. 0.60). The R + N model had better predictive performance and more effective risk stratification than the T + N model (C-index: 0.77 vs. 0.68, AUC: 0.80 vs. 0.70). The R + N model identified a low-risk group as deintensified chemoradiotherapy candidates in which no patient developed progression within 3 years, with 5-year progression-free survival (PFS) and overall survival (OS) both 90.7% (hazard ratio (HR) = 4.132, p = 0.018). CONCLUSION Our radiomics-based model combining radiomics score and N stage can identify specific LA-NPC candidates for whom de-escalation therapy can be performed without compromising therapeutic efficacy. CLINICAL RELEVANCE STATEMENT Our study shows that the radiomics-based model (R + N) can accurately stratify patients into different risk groups, with satisfactory prognosis in the low-risk group when treated with low-dose concurrent chemotherapy, providing new options for individualized de-escalation strategies. KEY POINTS • A radiomics score, consisting of 3 predictive radiomics features (flatness, mean, and GLDM-GLN) integrated with the N stage, can identify specific LA-NPC populations for deintensified treatment. • In the selection of LA-NPC candidates for de-intensified treatment, radiomics score extracted from primary nasopharyngeal tumors based on CECT can be superior to traditional T stage classification as a predictor.
Collapse
Affiliation(s)
- Yinbing Lin
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China
- Shantou University Medical College, 22 Xinling Road, Shantou 515000, 515041, Guangdong, China
| | - Zhining Yang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China
- Nasopharyngeal Carcinoma Research Center, Shantou University Medical College, Shantou University, 7 Raoping Road, Shantou, 515000, Guangdong, China
| | - Jiechen Chen
- Shantou University Medical College, 22 Xinling Road, Shantou 515000, 515041, Guangdong, China
| | - Mei Li
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China
- Nasopharyngeal Carcinoma Research Center, Shantou University Medical College, Shantou University, 7 Raoping Road, Shantou, 515000, Guangdong, China
| | - Zeman Cai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China
- Nasopharyngeal Carcinoma Research Center, Shantou University Medical College, Shantou University, 7 Raoping Road, Shantou, 515000, Guangdong, China
| | - Xiao Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China
- Shantou University Medical College, 22 Xinling Road, Shantou 515000, 515041, Guangdong, China
| | - Tiantian Zhai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China.
- Nasopharyngeal Carcinoma Research Center, Shantou University Medical College, Shantou University, 7 Raoping Road, Shantou, 515000, Guangdong, China.
| | - Zhixiong Lin
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515000, Guangdong, China.
- Nasopharyngeal Carcinoma Research Center, Shantou University Medical College, Shantou University, 7 Raoping Road, Shantou, 515000, Guangdong, China.
| |
Collapse
|
25
|
Yu L, Zhang Z, Yi H, Wang J, Li J, Wang X, Bai H, Ge H, Zheng X, Ni J, Qi H, Guan Y, Xu W, Zhu Z, Xing L, Dekker A, Wee L, Traverso A, Ye Z, Yuan Z. A PET/CT radiomics model for predicting distant metastasis in early-stage non-small cell lung cancer patients treated with stereotactic body radiotherapy: a multicentric study. Radiat Oncol 2024; 19:10. [PMID: 38254106 PMCID: PMC10802016 DOI: 10.1186/s13014-024-02402-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVES Stereotactic body radiotherapy (SBRT) is a treatment option for patients with early-stage non-small cell lung cancer (NSCLC) who are unfit for surgery. Some patients may experience distant metastasis. This study aimed to develop and validate a radiomics model for predicting distant metastasis in patients with early-stage NSCLC treated with SBRT. METHODS Patients at five institutions were enrolled in this study. Radiomics features were extracted based on the PET/CT images. After feature selection in the training set (from Tianjin), CT-based and PET-based radiomics signatures were built. Models based on CT and PET signatures were built and validated using external datasets (from Zhejiang, Zhengzhou, Shandong, and Shanghai). An integrated model that included CT and PET radiomic signatures was developed. The performance of the proposed model was evaluated in terms of its discrimination, calibration, and clinical utility. Multivariate logistic regression was used to calculate the probability of distant metastases. The cutoff value was obtained using the receiver operator characteristic curve (ROC), and the patients were divided into high- and low-risk groups. Kaplan-Meier analysis was used to evaluate the distant metastasis-free survival (DMFS) of different risk groups. RESULTS In total, 228 patients were enrolled. The median follow-up time was 31.4 (2.0-111.4) months. The model based on CT radiomics signatures had an area under the curve (AUC) of 0.819 in the training set (n = 139) and 0.786 in the external dataset (n = 89). The PET radiomics model had an AUC of 0.763 for the training set and 0.804 for the external dataset. The model combining CT and PET radiomics had an AUC of 0.835 for the training set and 0.819 for the external dataset. The combined model showed a moderate calibration and a positive net benefit. When the probability of distant metastasis was greater than 0.19, the patient was considered to be at high risk. The DMFS of patients with high- and low-risk was significantly stratified (P < 0.001). CONCLUSIONS The proposed PET/CT radiomics model can be used to predict distant metastasis in patients with early-stage NSCLC treated with SBRT and provide a reference for clinical decision-making. In this study, the model was established by combining CT and PET radiomics signatures in a moderate-quantity training cohort of early-stage NSCLC patients treated with SBRT and was successfully validated in independent cohorts. Physicians could use this easy-to-use model to assess the risk of distant metastasis after SBRT. Identifying subgroups of patients with different risk factors for distant metastasis is useful for guiding personalized treatment approaches.
Collapse
Affiliation(s)
- Lu Yu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhen Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - HeQing Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jin Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Junyi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Hui Bai
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Hong Ge
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zheng
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haoran Qi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Yong Guan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
26
|
Zhong Y, Cai C, Chen T, Gui H, Chen C, Deng J, Yang M, Yu B, Song Y, Wang T, Chen Y, Shi H, Xie D, Chen C, She Y. PET/CT-based deep learning grading signature to optimize surgical decisions for clinical stage I invasive lung adenocarcinoma and biologic basis under its prediction: a multicenter study. Eur J Nucl Med Mol Imaging 2024; 51:521-534. [PMID: 37725128 DOI: 10.1007/s00259-023-06434-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE No consensus on a grading system for invasive lung adenocarcinoma had been built over a long period of time. Until October 2020, a novel grading system was proposed to quantify the whole landscape of histologic subtypes and proportions of pulmonary adenocarcinomas. This study aims to develop a deep learning grading signature (DLGS) based on positron emission tomography/computed tomography (PET/CT) to personalize surgical treatments for clinical stage I invasive lung adenocarcinoma and explore the biologic basis under its prediction. METHODS A total of 2638 patients with clinical stage I invasive lung adenocarcinoma from 4 medical centers were retrospectively included to construct and validate the DLGS. The predictive performance of the DLGS was evaluated by the area under the receiver operating characteristic curve (AUC), its potential to optimize surgical treatments was investigated via survival analyses in risk groups defined by the DLGS, and its biological basis was explored by comparing histologic patterns, genotypic alternations, genetic pathways, and infiltration of immune cells in microenvironments between risk groups. RESULTS The DLGS to predict grade 3 achieved AUCs of 0.862, 0.844, and 0.851 in the validation set (n = 497), external cohort (n = 382), and prospective cohort (n = 600), respectively, which were significantly better than 0.814, 0.810, and 0.806 of the PET model, 0.813, 0.795, and 0.824 of the CT model, and 0.762, 0.734, and 0.751 of the clinical model. Additionally, for DLGS-defined high-risk population, lobectomy yielded an improved prognosis compared to sublobectomy p = 0.085 for overall survival [OS] and p = 0.038 for recurrence-free survival [RFS]) and systematic nodal dissection conferred a superior prognosis to limited nodal dissection (p = 0.001 for OS and p = 0.041 for RFS). CONCLUSION The DLGS harbors the potential to predict the histologic grade and personalize the surgical treatments for clinical stage I invasive lung adenocarcinoma. Its applicability to other territories should be further validated by a larger international study.
Collapse
Affiliation(s)
- Yifan Zhong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chuang Cai
- School of Computer Science and Communication Engineering , Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Gui
- Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi Medical College, Guizhou, China
| | - Jiajun Deng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo HwaMei Hospital, Chinese Academy of Sciences, Zhejiang, China
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi Medical College, Guizhou, China
| | - Tingting Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangchun Chen
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huazheng Shi
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Liu F, Xiang Z, Li Q, Fang X, Zhou J, Yang X, Lin H, Yang Q. 18F-FDG PET/CT-based radiomics model for predicting the degree of pathological differentiation in non-small cell lung cancer: a multicentre study. Clin Radiol 2024; 79:e147-e155. [PMID: 37884401 DOI: 10.1016/j.crad.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
AIM To explore the value of 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT)-based radiomics model for predicting the degree of pathological differentiation in non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS Clinical characteristics of 182 NSCLC patients from four centres were collected, and radiomics features were extracted from 18F-FDG PET/CT images. Three logistic regression prediction models were established: clinical model; radiomics model; and nomogram combining radiomics signatures and clinical features. The predictive ability of the models was assessed using receiver operating characteristics curve analysis. RESULTS Patients from centre 1 were assigned randomly to the training and internal validation cohorts (7:3 ratio); patients from centres 2-4 served as the external validation cohort. The area under the curve (AUC) values for the clinical model in the training, internal validation, and external validation cohort were 0.74 (95% confidence interval [CI] = 0.64-0.84), 0.64 (95% CI = 0.46-0.81), and 0.74 (95% CI = 0.60-0.88), respectively. In the training (AUC: 0.84 [95% CI = 0.77-0.92]), internal validation (AUC: 0.81 [95% CI = 0.67-0.95]), and external validation cohorts (AUC: 0.74 [95% CI = 0.58-0.89]), the radiomics model showed good predictive ability for differentiation. Compared to the clinical and radiomics models, the nomogram has relatively better diagnostic performance, and the AUC values for nomogram in the training, internal validation, and external validation cohort were 0.86 (95% CI = 0.78-0.93), 0.83 (95% CI = 0.70-0.96), and 0.77 (95% CI = 0.62-0.92), respectively. CONCLUSIONS The 18F-FDG PET/CT-based radiomics model showed good ability for predicting the degree of differentiation of NSCLC. The nomogram combining the radiomics signature and clinical features has relatively better diagnostic performance.
Collapse
Affiliation(s)
- F Liu
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Z Xiang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Q Li
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - X Fang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - J Zhou
- The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - X Yang
- Sichuan Science City Hospital, Mianyang, Sichuan 621000, China
| | - H Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha 410005, China
| | - Q Yang
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumour Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
28
|
Hou J, Jin H, Zhang Y, Xu Y, Cui F, Qin X, Han L, Yuan Z, Zheng G, Peng J, Shu Z, Gong X. Hybrid model of CT-fractional flow reserve, pericoronary fat attenuation index and radiomics for predicting the progression of WMH: a dual-center pilot study. Front Cardiovasc Med 2023; 10:1282768. [PMID: 38179506 PMCID: PMC10766365 DOI: 10.3389/fcvm.2023.1282768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Objective To develop and validate a hybrid model incorporating CT-fractional flow reserve (CT-FFR), pericoronary fat attenuation index (pFAI), and radiomics signatures for predicting progression of white matter hyperintensity (WMH). Methods A total of 226 patients who received coronary computer tomography angiography (CCTA) and brain magnetic resonance imaging from two hospitals were divided into a training set (n = 116), an internal validation set (n = 30), and an external validation set (n = 80). Patients who experienced progression of WMH were identified from subsequent MRI results. We calculated CT-FFR and pFAI from CCTA images using semi-automated software, and segmented the pericoronary adipose tissue (PCAT) and myocardial ROI. A total of 1,073 features were extracted from each ROI, and were then refined by Elastic Net Regression. Firstly, different machine learning algorithms (Logistic Regression [LR], Support Vector Machine [SVM], Random Forest [RF], k-nearest neighbor [KNN] and eXtreme Gradient Gradient Boosting Machine [XGBoost]) were used to evaluate the effectiveness of radiomics signatures for predicting WMH progression. Then, the optimal machine learning algorithm was used to compare the predictive performance of individual and hybrid models based on independent risk factors of WMH progression. Receiver operating characteristic (ROC) curve analysis, calibration and decision curve analysis were used to evaluate predictive performance and clinical value of the different models. Results CT-FFR, pFAI, and radiomics signatures were independent predictors of WMH progression. Based on the machine learning algorithms, the PCAT signatures led to slightly better predictions than the myocardial signatures and showed the highest AUC value in the XGBoost algorithm for predicting WMH progression (AUC: 0.731 [95% CI: 0.603-0.838] vs.0.711 [95% CI: 0.584-0.822]). In addition, pFAI provided better predictions than CT-FFR (AUC: 0.762 [95% CI: 0.651-0.863] vs. 0.682 [95% CI: 0.547-0.799]). A hybrid model that combined CT-FFR, pFAI, and two radiomics signatures provided the best predictions of WMH progression [AUC: 0.893 (95%CI: 0.815-0.956)]. Conclusion pFAI was more effective than CT-FFR, and PCAT signatures were more effective than myocardial signatures in predicting WMH progression. A hybrid model that combines pFAI, CT-FFR, and two radiomics signatures has potential use for identifying WMH progression.
Collapse
Affiliation(s)
- Jie Hou
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Hui Jin
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Yongsheng Zhang
- The Hangzhou TCM Hospital (Affiliated Zhejiang Chinese Medical University), Hangzhou, Zhejiang, China
| | - Yuyun Xu
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng Cui
- The Hangzhou TCM Hospital (Affiliated Zhejiang Chinese Medical University), Hangzhou, Zhejiang, China
| | - Xue Qin
- Bengbu Medical College, Bengbu, Anhui, China
| | - Lu Han
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhongyu Yuan
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | | | - Jiaxuan Peng
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhenyu Shu
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiangyang Gong
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
Tang X, Wu F, Chen X, Ye S, Ding Z. Current status and prospect of PET-related imaging radiomics in lung cancer. Front Oncol 2023; 13:1297674. [PMID: 38164195 PMCID: PMC10757959 DOI: 10.3389/fonc.2023.1297674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Lung cancer is highly aggressive, which has a high mortality rate. Major types encompass lung adenocarcinoma, lung squamous cell carcinoma, lung adenosquamous carcinoma, small cell carcinoma, and large cell carcinoma. Lung adenocarcinoma and lung squamous cell carcinoma together account for more than 80% of cases. Diverse subtypes demand distinct treatment approaches. The application of precision medicine necessitates prompt and accurate evaluation of treatment effectiveness, contributing to the improvement of treatment strategies and outcomes. Medical imaging is crucial in the diagnosis and management of lung cancer, with techniques such as fluoroscopy, computed radiography (CR), digital radiography (DR), computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET)/CT, and PET/MRI being essential tools. The surge of radiomics in recent times offers fresh promise for cancer diagnosis and treatment. In particular, PET/CT and PET/MRI radiomics, extensively studied in lung cancer research, have made advancements in diagnosing the disease, evaluating metastasis, predicting molecular subtypes, and forecasting patient prognosis. While conventional imaging methods continue to play a primary role in diagnosis and assessment, PET/CT and PET/MRI radiomics simultaneously provide detailed morphological and functional information. This has significant clinical potential value, offering advantages for lung cancer diagnosis and treatment. Hence, this manuscript provides a review of the latest developments in PET-related radiomics for lung cancer.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, Hangzhou Wuyunshan Hospital (Hangzhou Health Promotion Research Institute), Hangzhou, China
| | - Fan Wu
- Department of Nuclear Medicine and Radiology, Shulan Hangzhou Hospital affiliated to Shulan International Medical College of Zhejiang Shuren University, Hangzhou, China
| | - Xiaofen Chen
- Department of Radiology, Hangzhou Wuyunshan Hospital (Hangzhou Health Promotion Research Institute), Hangzhou, China
| | - Shengli Ye
- Department of Nuclear Medicine and Radiology, Shulan Hangzhou Hospital affiliated to Shulan International Medical College of Zhejiang Shuren University, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Hangzhou First People’s Hospital, Hangzhou, China
| |
Collapse
|
30
|
Trivizakis E, Koutroumpa NM, Souglakos J, Karantanas A, Zervakis M, Marias K. Radiotranscriptomics of non-small cell lung carcinoma for assessing high-level clinical outcomes using a machine learning-derived multi-modal signature. Biomed Eng Online 2023; 22:125. [PMID: 38102586 PMCID: PMC10724973 DOI: 10.1186/s12938-023-01190-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Multi-omics research has the potential to holistically capture intra-tumor variability, thereby improving therapeutic decisions by incorporating the key principles of precision medicine. The purpose of this study is to identify a robust method of integrating features from different sources, such as imaging, transcriptomics, and clinical data, to predict the survival and therapy response of non-small cell lung cancer patients. METHODS 2996 radiomics, 5268 transcriptomics, and 8 clinical features were extracted from the NSCLC Radiogenomics dataset. Radiomics and deep features were calculated based on the volume of interest in pre-treatment, routine CT examinations, and then combined with RNA-seq and clinical data. Several machine learning classifiers were used to perform survival analysis and assess the patient's response to adjuvant chemotherapy. The proposed analysis was evaluated on an unseen testing set in a k-fold cross-validation scheme. Score- and concatenation-based multi-omics were used as feature integration techniques. RESULTS Six radiomics (elongation, cluster shade, entropy, variance, gray-level non-uniformity, and maximal correlation coefficient), six deep features (NasNet-based activations), and three transcriptomics (OTUD3, SUCGL2, and RQCD1) were found to be significant for therapy response. The examined score-based multi-omic improved the AUC up to 0.10 on the unseen testing set (0.74 ± 0.06) and the balance between sensitivity and specificity for predicting therapy response for 106 patients, resulting in less biased models and improving upon the either highly sensitive or highly specific single-source models. Six radiomics (kurtosis, GLRLM- and GLSZM-based non-uniformity from images with no filtering, biorthogonal, and daubechies wavelets), seven deep features (ResNet-based activations), and seven transcriptomics (ELP3, ZZZ3, PGRMC2, TRAK1, ATIC, USP7, and PNPLA2) were found to be significant for the survival analysis. Accordingly, the survival analysis for 115 patients was also enhanced up to 0.20 by the proposed score-based multi-omics in terms of the C-index (0.79 ± 0.03). CONCLUSIONS Compared to single-source models, multi-omics integration has the potential to improve prediction performance, increase model stability, and reduce bias for both treatment response and survival analysis.
Collapse
Affiliation(s)
- Eleftherios Trivizakis
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013, Heraklion, Greece.
- Medical School, University of Crete, 71003, Heraklion, Greece.
| | - Nikoletta-Maria Koutroumpa
- Medical School, University of Crete, 71003, Heraklion, Greece
- School of Electrical and Computer Engineering, Technical University of Crete, 73100, Chania, Greece
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 71003, Heraklion, Greece
- Department of Medical Oncology, University Hospital of Heraklion, 71500, Heraklion, Greece
| | - Apostolos Karantanas
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013, Heraklion, Greece
- Department of Radiology, Medical School, University of Crete, 71003, Heraklion, Greece
| | - Michalis Zervakis
- School of Electrical and Computer Engineering, Technical University of Crete, 73100, Chania, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, 71410, Heraklion, Greece
| |
Collapse
|
31
|
Luining WI, Oprea-Lager DE, Vis AN, van Moorselaar RJA, Knol RJJ, Wondergem M, Boellaard R, Cysouw MCF. Optimization and validation of 18F-DCFPyL PET radiomics-based machine learning models in intermediate- to high-risk primary prostate cancer. PLoS One 2023; 18:e0293672. [PMID: 37943772 PMCID: PMC10635444 DOI: 10.1371/journal.pone.0293672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Radiomics extracted from prostate-specific membrane antigen (PSMA)-PET modeled with machine learning (ML) may be used for prediction of disease risk. However, validation of previously proposed approaches is lacking. We aimed to optimize and validate ML models based on 18F-DCFPyL-PET radiomics for the prediction of lymph-node involvement (LNI), extracapsular extension (ECE), and postoperative Gleason score (GS) in primary prostate cancer (PCa) patients. METHODS Patients with intermediate- to high-risk PCa who underwent 18F-DCFPyL-PET/CT before radical prostatectomy with pelvic lymph-node dissection were evaluated. The training dataset included 72 patients, the internal validation dataset 24 patients, and the external validation dataset 27 patients. PSMA-avid intra-prostatic lesions were delineated semi-automatically on PET and 480 radiomics features were extracted. Conventional PET-metrics were derived for comparative analysis. Segmentation, preprocessing, and ML methods were optimized in repeated 5-fold cross-validation (CV) on the training dataset. The trained models were tested on the combined validation dataset. Combat harmonization was applied to external radiomics data. Model performance was assessed using the receiver-operating-characteristics curve (AUC). RESULTS The CV-AUCs in the training dataset were 0.88, 0.79 and 0.84 for LNI, ECE, and GS, respectively. In the combined validation dataset, the ML models could significantly predict GS with an AUC of 0.78 (p<0.05). However, validation AUCs for LNI and ECE prediction were not significant (0.57 and 0.63, respectively). Conventional PET metrics-based models had comparable AUCs for LNI (0.59, p>0.05) and ECE (0.66, p>0.05), but a lower AUC for GS (0.73, p<0.05). In general, Combat harmonization improved external validation AUCs (-0.03 to +0.18). CONCLUSION In internal and external validation, 18F-DCFPyL-PET radiomics-based ML models predicted high postoperative GS but not LNI or ECE in intermediate- to high-risk PCa. Therefore, the clinical benefit seems to be limited. These results underline the need for external and/or multicenter validation of PET radiomics-based ML model analyses to assess their generalizability.
Collapse
Affiliation(s)
- Wietske I. Luining
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniela E. Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
| | - Reindert J. A. van Moorselaar
- Department of Urology, Amsterdam University Medical Centers, Prostate Cancer Network Netherlands, Amsterdam, The Netherlands
| | - Remco J. J. Knol
- Department of Nuclear Medicine, Northwest Clinics, Alkmaar, The Netherlands
| | - Maurits Wondergem
- Department of Nuclear Medicine, Northwest Clinics, Alkmaar, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Matthijs C. F. Cysouw
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Erasmus LT, Strange TA, Agrawal R, Strange CD, Ahuja J, Shroff GS, Truong MT. Lung Cancer Staging: Imaging and Potential Pitfalls. Diagnostics (Basel) 2023; 13:3359. [PMID: 37958255 PMCID: PMC10649001 DOI: 10.3390/diagnostics13213359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths in men and women in the United States. Accurate staging is needed to determine prognosis and devise effective treatment plans. The International Association for the Study of Lung Cancer (IASLC) has made multiple revisions to the tumor, node, metastasis (TNM) staging system used by the Union for International Cancer Control and the American Joint Committee on Cancer to stage lung cancer. The eighth edition of this staging system includes modifications to the T classification with cut points of 1 cm increments in tumor size, grouping of lung cancers associated with partial or complete lung atelectasis or pneumonitis, grouping of tumors with involvement of a main bronchus regardless of distance from the carina, and upstaging of diaphragmatic invasion to T4. The N classification describes the spread to regional lymph nodes and no changes were proposed for TNM-8. In the M classification, metastatic disease is divided into intra- versus extrathoracic metastasis, and single versus multiple metastases. In order to optimize patient outcomes, it is important to understand the nuances of the TNM staging system, the strengths and weaknesses of various imaging modalities used in lung cancer staging, and potential pitfalls in image interpretation.
Collapse
Affiliation(s)
- Lauren T. Erasmus
- Department of Anatomy and Cell Biology, Faculty of Sciences, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Taylor A. Strange
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Rishi Agrawal
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Chad D. Strange
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Jitesh Ahuja
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Girish S. Shroff
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| | - Mylene T. Truong
- Department of Thoracic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.A.); (C.D.S.); (J.A.)
| |
Collapse
|
33
|
Tang X, Li Y, Shen LT, Yan WF, Qian WL, Yang ZG. CT Radiomics Predict EGFR-T790M Resistance Mutation in Advanced Non-Small Cell Lung Cancer Patients After Progression on First-line EGFR-TKI. Acad Radiol 2023; 30:2574-2587. [PMID: 36941156 DOI: 10.1016/j.acra.2023.01.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 03/23/2023]
Abstract
RATIONALE AND OBJECTIVES We aim to explore the value of chest CT radiomics in predicting the epidermal growth factor receptor (EGFR)-T790M resistance mutation of advanced non-small cell lung cancer (NSCLC) patients after the failure of first-line EGFR-tyrosine kinase inhibitor (EGFR-TKI). MATERIALS AND METHODS A total of 211 and 135 advanced NSCLC patients with tumor tissue-based (Cohort-1) or circulating tumor DNA (ctDNA)-based (Cohort-2) EGFR-T790M testing were included, respectively. Cohort-1 was used for modeling and Cohort-2 was for models' validation. Radiomic features were extracted from tumor lesions on chest nonenhanced CT (NECT) and/or contrast-enhanced CT (CECT). We used eight feature selectors and eight classifier algorithms to establish radiomic models. Models were evaluated by area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS CT morphological manifestations of peripheral location and pleural indentation sign were associated with EGFR-T790M. For NECT, CECT, and NECT+CECT radiomic features, the feature selector and classifier algorithms of LASSO and Stepwise logistic regression, Boruta and SVM, and LASSO and SVM were chosen to develop the optimal model, respectively (AUC: 0.844, 0.811, and 0.897). All models performed well in calibration curves and DCA. Independent validation of models in Cohort-2 revealed that both NECT and CECT models individually had limited power for predicting EGFR-T790M mutation detected by ctDNA (AUC: 0.649, 0.675), while the NECT+CECT radiomic model had a satisfactory AUC (0.760). CONCLUSION This study proved the feasibility of using CT radiomic features to predict the EGFR-T790M resistance mutation, which could be helpful in guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li-Ting Shen
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Feng Yan
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Lei Qian
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
34
|
Pan F, Feng L, Liu B, Hu Y, Wang Q. Application of radiomics in diagnosis and treatment of lung cancer. Front Pharmacol 2023; 14:1295511. [PMID: 38027000 PMCID: PMC10646419 DOI: 10.3389/fphar.2023.1295511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Radiomics has become a research field that involves the process of converting standard nursing images into quantitative image data, which can be combined with other data sources and subsequently analyzed using traditional biostatistics or artificial intelligence (Al) methods. Due to the capture of biological and pathophysiological information by radiomics features, these quantitative radiomics features have been proven to provide fast and accurate non-invasive biomarkers for lung cancer risk prediction, diagnosis, prognosis, treatment response monitoring, and tumor biology. In this review, radiomics has been emphasized and discussed in lung cancer research, including advantages, challenges, and drawbacks.
Collapse
Affiliation(s)
- Feng Pan
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of CT, Jilin Province FAW General Hospital, Changchun, China
| | - Li Feng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baocai Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qian Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
35
|
Khodabakhshi Z, Amini M, Hajianfar G, Oveisi M, Shiri I, Zaidi H. Dual-Centre Harmonised Multimodal Positron Emission Tomography/Computed Tomography Image Radiomic Features and Machine Learning Algorithms for Non-small Cell Lung Cancer Histopathological Subtype Phenotype Decoding. Clin Oncol (R Coll Radiol) 2023; 35:713-725. [PMID: 37599160 DOI: 10.1016/j.clon.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/10/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
AIMS We aimed to build radiomic models for classifying non-small cell lung cancer (NSCLC) histopathological subtypes through a dual-centre dataset and comprehensively evaluate the effect of ComBat harmonisation on the performance of single- and multimodality radiomic models. MATERIALS AND METHODS A public dataset of NSCLC patients from two independent centres was used. Two image fusion methods, namely guided filtering-based fusion and image fusion based on visual saliency map and weighted least square optimisation, were used. Radiomic features were extracted from each scan, including first-order, texture and moment-invariant features. Subsequently, ComBat harmonisation was applied to the extracted features from computed tomography (CT), positron emission tomography (PET) and fused images to correct the centre effect. For feature selection, least absolute shrinkage and selection operator (Lasso) and recursive feature elimination (RFE) were investigated. For machine learning, logistic regression (LR), support vector machine (SVM) and AdaBoost were evaluated for classifying NSCLC subtypes. Training and evaluation of the models were carried out in a robust framework to offset plausible errors and performance was reported using area under the curve, balanced accuracy, sensitivity and specificity before and after harmonisation. N-way ANOVA was used to assess the effect of different factors on the performance of the models. RESULTS Support vector machine fed with selected features by recursive feature elimination from a harmonised PET feature set achieved the highest performance (area under the curve = 0.82) in classifying NSCLC histopathological subtypes. Although the performance of the models did not significantly improve for CT images after harmonisation, the performance of PET and guided filtering-based fusion feature signatures significantly improved for almost all models. Although the selection of the image modality and feature selection methods was effective on the performance of the model (ANOVA P-values <0.001), machine learning and harmonisation did not change the performance significantly (ANOVA P-values = 0.839 and 0.292, respectively). CONCLUSION This study confirmed the potential of radiomic analysis on PET, CT and hybrid images for histopathological classification of NSCLC subtypes.
Collapse
Affiliation(s)
- Z Khodabakhshi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - M Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - G Hajianfar
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - M Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran; Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK; Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - I Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - H Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
36
|
Zhou Y, Zhang B, Han J, Dai N, Jia T, Huang H, Deng S, Sang S. Development of a radiomic-clinical nomogram for prediction of survival in patients with diffuse large B-cell lymphoma treated with chimeric antigen receptor T cells. J Cancer Res Clin Oncol 2023; 149:11549-11560. [PMID: 37395846 DOI: 10.1007/s00432-023-05038-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND In our current work, an 18F-FDG PET/CT radiomics-based model was developed to assess the progression-free survival (PFS) and overall survival (OS) of patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) who received chimeric antigen receptor (CAR)-T cell therapy. METHODS A total of 61 DLBCL cases receiving 18F-FDG PET/CT before CAR-T cell infusion were included in the current analysis, and these patients were randomly assigned to a training cohort (n = 42) and a validation cohort (n = 19). Radiomic features from PET and CT images were obtained using LIFEx software, and radiomics signatures (R-signatures) were then constructed by choosing the optimal parameters according to their PFS and OS. Subsequently, the radiomics model and clinical model were constructed and validated. RESULTS The radiomics model that integrated R-signatures and clinical risk factors showed superior prognostic performance compared with the clinical models in terms of both PFS (C-index: 0.710 vs. 0.716; AUC: 0.776 vs. 0.712) and OS (C-index: 0.780 vs. 0.762; AUC: 0.828 vs. 0.728). For validation, the C-index of the two approaches was 0.640 vs. 0.619 and 0.676 vs. 0.699 for predicting PFS and OS, respectively. Moreover, the AUC was 0.886 vs. 0.635 and 0.778 vs. 0.705, respectively. The calibration curves indicated good agreement, and the decision curve analysis suggested that the net benefit of radiomics models was higher than that of clinical models. CONCLUSIONS PET/CT-derived R-signature could be a potential prognostic biomarker for R/R DLBCL patients undergoing CAR-T cell therapy. Moreover, the risk stratification could be further enhanced when the PET/CT-derived R-signature was combined with clinical factors.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jiangqin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Na Dai
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Tongtong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haiwen Huang
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
37
|
Klement RJ, Sweeney RA. Metabolic factors associated with the prognosis of oligometastatic patients treated with stereotactic body radiotherapy. Cancer Metastasis Rev 2023; 42:927-940. [PMID: 37261610 DOI: 10.1007/s10555-023-10110-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
Over the past two decades, it has been established that cancer patients with oligometastases, i.e., only a few detectable metastases confined to one or a few organs, may benefit from an aggressive local treatment approach such as the application of high-precision stereotactic body radiotherapy (SBRT). Specifically, some studies have indicated that achieving long-term local tumor control of oligometastases is associated with prolonged overall survival. This motivates investigations into which factors may modify the dose-response relationship of SBRT by making metastases more or less radioresistant. One such factor relates to the uptake of the positron emission tomography tracer 2-deoxy-2-[18F]fluoro-D-glucose (FDG) which reflects the extent of tumor cell glycolysis or the Warburg effect, respectively. Here we review the biological mechanisms how the Warburg effect drives tumor cell radioresistance and metastasis and draw connections to clinical studies reporting associations between high FDG uptake and worse clinical outcomes after SBRT for oligometastases. We further review the evidence for distinct metabolic phenotypes of metastases preferentially seeding to specific organs and their possible translation into distinct radioresistance. Finally, evidence that obesity and hyperglycemia also affect outcomes after SBRT will be presented. While delivered dose is the main determinant of a high local tumor control probability, there might be clinical scenarios when metabolic targeting could make the difference between achieving local control or not, for example when doses have to be compromised in order to spare neighboring high-risk organs, or when tumors are expected to be highly therapy-resistant due to heavy pretreatment such as chemotherapy and/or radiotherapy.
Collapse
Affiliation(s)
- Rainer J Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany.
| | - Reinhart A Sweeney
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Robert-Koch-Straße 10, 97422, Schweinfurt, Germany
| |
Collapse
|
38
|
Shao J, Feng J, Li J, Liang S, Li W, Wang C. Novel tools for early diagnosis and precision treatment based on artificial intelligence. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:148-160. [PMID: 39171128 PMCID: PMC11332840 DOI: 10.1016/j.pccm.2023.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer has the highest mortality rate among all cancers in the world. Hence, early diagnosis and personalized treatment plans are crucial to improving its 5-year survival rate. Chest computed tomography (CT) serves as an essential tool for lung cancer screening, and pathology images are the gold standard for lung cancer diagnosis. However, medical image evaluation relies on manual labor and suffers from missed diagnosis or misdiagnosis, and physician heterogeneity. The rapid development of artificial intelligence (AI) has brought a whole novel opportunity for medical task processing, demonstrating the potential for clinical application in lung cancer diagnosis and treatment. AI technologies, including machine learning and deep learning, have been deployed extensively for lung nodule detection, benign and malignant classification, and subtype identification based on CT images. Furthermore, AI plays a role in the non-invasive prediction of genetic mutations and molecular status to provide the optimal treatment regimen, and applies to the assessment of therapeutic efficacy and prognosis of lung cancer patients, enabling precision medicine to become a reality. Meanwhile, histology-based AI models assist pathologists in typing, molecular characterization, and prognosis prediction to enhance the efficiency of diagnosis and treatment. However, the leap to extensive clinical application still faces various challenges, such as data sharing, standardized label acquisition, clinical application regulation, and multimodal integration. Nevertheless, AI holds promising potential in the field of lung cancer to improve cancer care.
Collapse
Affiliation(s)
- Jun Shao
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaming Feng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shufan Liang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
39
|
Chen M, Copley SJ, Viola P, Lu H, Aboagye EO. Radiomics and artificial intelligence for precision medicine in lung cancer treatment. Semin Cancer Biol 2023; 93:97-113. [PMID: 37211292 DOI: 10.1016/j.semcancer.2023.05.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/14/2023] [Accepted: 05/17/2023] [Indexed: 05/23/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. It exhibits, at the mesoscopic scale, phenotypic characteristics that are generally indiscernible to the human eye but can be captured non-invasively on medical imaging as radiomic features, which can form a high dimensional data space amenable to machine learning. Radiomic features can be harnessed and used in an artificial intelligence paradigm to risk stratify patients, and predict for histological and molecular findings, and clinical outcome measures, thereby facilitating precision medicine for improving patient care. Compared to tissue sampling-driven approaches, radiomics-based methods are superior for being non-invasive, reproducible, cheaper, and less susceptible to intra-tumoral heterogeneity. This review focuses on the application of radiomics, combined with artificial intelligence, for delivering precision medicine in lung cancer treatment, with discussion centered on pioneering and groundbreaking works, and future research directions in the area.
Collapse
Affiliation(s)
- Mitchell Chen
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK; Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| | - Susan J Copley
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK; Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| | - Patrizia Viola
- North West London Pathology, Charing Cross Hospital, Fulham Palace Rd, London W6 8RF, UK
| | - Haonan Lu
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK.
| |
Collapse
|
40
|
Xing W, Gao W, Lv X, Zhao Z, Xu X, Wu Z, Mao G, Chen J. Artificial intelligence predicts lung cancer radiotherapy response: A meta-analysis. Artif Intell Med 2023; 142:102585. [PMID: 37316099 DOI: 10.1016/j.artmed.2023.102585] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/06/2023] [Accepted: 05/16/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Artificial intelligence (AI) technology has clustered patients based on clinical features into sub-clusters to stratify high-risk and low-risk groups to predict outcomes in lung cancer after radiotherapy and has gained much more attention in recent years. Given that the conclusions vary considerably, this meta-analysis was conducted to investigate the combined predictive effect of AI models on lung cancer. METHODS This study was performed according to PRISMA guidelines. PubMed, ISI Web of Science, and Embase databases were searched for relevant literature. Outcomes, including overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), and local control (LC), were predicted using AI models in patients with lung cancer after radiotherapy, and were used to calculate the pooled effect. Quality, heterogeneity, and publication bias of the included studies were also evaluated. RESULTS Eighteen articles with 4719 patients were eligible for this meta-analysis. The combined hazard ratios (HRs) of the included studies for OS, LC, PFS, and DFS of lung cancer patients were 2.55 (95 % confidence interval (CI) = 1.73-3.76), 2.45 (95 % CI = 0.78-7.64), 3.84 (95 % CI = 2.20-6.68), and 2.66 (95 % CI = 0.96-7.34), respectively. The combined area under the receiver operating characteristics curve (AUC) of the included articles on OS and LC in patients with lung cancer was 0.75 (95 % CI = 0.67-0.84), and 0.80 (95%CI = 0.0.68-0.95), respectively. CONCLUSION The clinical feasibility of predicting outcomes using AI models after radiotherapy in patients with lung cancer was demonstrated. Large-scale, prospective, multicenter studies should be conducted to more accurately predict the outcomes in patients with lung cancer.
Collapse
Affiliation(s)
- Wenmin Xing
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Wenyan Gao
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences&Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Xiaoling Lv
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Zhenlei Zhao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Xiaogang Xu
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Zhibing Wu
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China.
| | - Jun Chen
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, China.
| |
Collapse
|
41
|
Long ZC, Ding XC, Zhang XB, Sun PP, Hao FR, Li ZR, Hu M. The Efficacy of Pretreatment 18F-FDG PET-CT-Based Deep Learning Network Structure to Predict Survival in Nasopharyngeal Carcinoma. Clin Med Insights Oncol 2023; 17:11795549231171793. [PMID: 37251551 PMCID: PMC10214083 DOI: 10.1177/11795549231171793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
Background Previous studies have shown that the 5-year survival rates of patients with nasopharyngeal carcinoma (NPC) were still not ideal despite great improvement in NPC treatments. To achieve individualized treatment of NPC, we have been looking for novel models to predict the prognosis of patients with NPC. The objective of this study was to use a novel deep learning network structural model to predict the prognosis of patients with NPC and to compare it with the traditional PET-CT model combining metabolic parameters and clinical factors. Methods A total of 173 patients were admitted to 2 institutions between July 2014 and April 2020 for the retrospective study; each received a PET-CT scan before treatment. The least absolute shrinkage and selection operator (LASSO) was employed to select some features, including SUVpeak-P, T3, age, stage II, MTV-P, N1, stage III and pathological type, which were associated with overall survival (OS) of patients. We constructed 2 survival prediction models: an improved optimized adaptive multimodal task (a 3D Coordinate Attention Convolutional Autoencoder and an uncertainty-based jointly Optimizing Cox Model, CACA-UOCM for short) and a clinical model. The predictive power of these models was assessed using the Harrell Consistency Index (C index). Overall survival of patients with NPC was compared by Kaplan-Meier and Log-rank tests. Results The results showed that CACA-UOCM model could estimate OS (C index, 0.779 for training, 0.774 for validation, and 0.819 for testing) and divide patients into low and high mortality risk groups, which were significantly associated with OS (P < .001). However, the C-index of the model based only on clinical variables was only 0.42. Conclusions The deep learning network model based on 18F-FDG PET/CT can serve as a reliable and powerful predictive tool for NPC and provide therapeutic strategies for individual treatment.
Collapse
Affiliation(s)
- Zi-Chan Long
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xing-Chen Ding
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xian-Bin Zhang
- Department of General Surgery and Integrated Chinese and Western Medicine, Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Peng-Peng Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fu-Rong Hao
- Department of Radiation Oncology, Weifang People's Hospital, Weifang, China
| | | | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
42
|
Chen K, Hou L, Chen M, Li S, Shi Y, Raynor WY, Yang H. Predicting the Efficacy of SBRT for Lung Cancer with 18F-FDG PET/CT Radiogenomics. Life (Basel) 2023; 13:life13040884. [PMID: 37109413 PMCID: PMC10142286 DOI: 10.3390/life13040884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Purpose: to develop a radiogenomic model on the basis of 18F-FDG PET/CT radiomics and clinical-parameter EGFR for predicting PFS stratification in lung-cancer patients after SBRT treatment. Methods: A total of 123 patients with lung cancer who had undergone 18F-FDG PET/CT examination before SBRT from September 2014 to December 2021 were retrospectively analyzed. All patients’ PET/CT images were manually segmented, and the radiomic features were extracted. LASSO regression was used to select radiomic features. Logistic regression analysis was used to screen clinical features to establish the clinical EGFR model, and a radiogenomic model was constructed by combining radiomics and clinical EGFR. We used the receiver operating characteristic curve and calibration curve to assess the efficacy of the models. The decision curve and influence curve analysis were used to evaluate the clinical value of the models. The bootstrap method was used to validate the radiogenomic model, and the mean AUC was calculated to assess the model. Results: A total of 2042 radiomics features were extracted. Five radiomic features were related to the PFS stratification of lung-cancer patients with SBRT. T-stage and overall stages (TNM) were independent factors for predicting PFS stratification. AUCs under the ROC curve of the radiomics, clinical EGFR, and radiogenomic models were 0.84, 0.67, and 0.86, respectively. The calibration curve shows that the predicted value of the radiogenomic model was in good agreement with the actual value. The decision and influence curve showed that the model had high clinical application values. After Bootstrap validation, the mean AUC of the radiogenomic model was 0.850(95%CI 0.849–0.851). Conclusions: The radiogenomic model based on 18F-FDG PET/CT radiomics and clinical EGFR has good application value in predicting the PFS stratification of lung-cancer patients after SBRT treatment.
Collapse
Affiliation(s)
- Kuifei Chen
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Liqiao Hou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Meng Chen
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Shuling Li
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Yangyang Shi
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85724, USA
| | - William Y. Raynor
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Haihua Yang
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
- Correspondence: or
| |
Collapse
|
43
|
Zhang J, Zhu H, Wang J, Chen Y, Li Y, Chen X, Chen M, Cai Z, Liu W. Machine learning in non-small cell lung cancer radiotherapy: A bibliometric analysis. Front Oncol 2023; 13:1082423. [PMID: 37025583 PMCID: PMC10072228 DOI: 10.3389/fonc.2023.1082423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundMachine learning is now well-developed in non-small cell lung cancer (NSCLC) radiotherapy. But the research trend and hotspots are still unclear. To investigate the progress in machine learning in radiotherapy NSCLC, we performed a bibliometric analysis of associated research and discuss the current research hotspots and potential hot areas in the future.MethodsThe involved researches were obtained from the Web of Science Core Collection database (WoSCC). We used R-studio software, the Bibliometrix package and VOSviewer (Version 1.6.18) software to perform bibliometric analysis.ResultsWe found 197 publications about machine learning in radiotherapy for NSCLC in the WoSCC, and the journal Medical Physics contributed the most articles. The University of Texas MD Anderson Cancer Center was the most frequent publishing institution, and the United States contributed most of the publications. In our bibliometric analysis, “radiomics” was the most frequent keyword, and we found that machine learning is mainly applied to analyze medical images in the radiotherapy of NSCLC.ResultsThe research we identified about machine learning in NSCLC radiotherapy was mainly related to the radiotherapy planning of NSCLC and the prediction of treatment effects and adverse events in NSCLC patients who were under radiotherapy. Our research has added new insights into machine learning in NSCLC radiotherapy and could help researchers better identify hot research areas in the future.
Collapse
Affiliation(s)
- Jiaming Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huijun Zhu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jue Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yulu Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yihe Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinyu Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Menghua Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhengwen Cai
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Zhengwen Cai, ; Wenqi Liu,
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Zhengwen Cai, ; Wenqi Liu,
| |
Collapse
|
44
|
Liu J, Xia X, Zou Q, Xie X, Lei Y, Wan Q, Li X. Diagnostic performance of diffusion-weighted imaging versus 18F-FDG PET/CT in differentiating pulmonary lesions: an updated meta-analysis of comparative studies. BMC Med Imaging 2023; 23:37. [PMID: 36899303 PMCID: PMC10007793 DOI: 10.1186/s12880-023-00990-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE To compare the diagnostic accuracy of diffusion-weighted imaging (DWI) and 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) for differentiating pulmonary nodules and masses. METHODS We systematically searched six databases, including PubMed, EMBASE, the Cochrane Library, and three Chinese databases, to identify studies that used both DWI and PET/CT to differentiate pulmonary nodules. The diagnostic performance of DWI and PET/CT was compared and pooled sensitivity and specificity were calculated along with 95% confidence intervals (CIs). The Quality Assessment of Diagnostic Accuracy Studies 2 was used to assess the quality of the included studies, and STATA 16.0 software was utilized to perform statistical analysis. RESULTS Overall, 10 studies that enrolled a total of 871 patients with 948 pulmonary nodules were included in this meta-analysis. DWI had greater pooled sensitivity (0.85 [95% CI 0.77-0.90]) and specificity (0.91 [95% CI 0.82-0.96]) than PET/CT (sensitivity, 0.82 [95% CI 0.70-0.90]); specificity, (0.81, [95% CI 0.72-0.87]). The area under the curve of DWI and PET/CT were 0.94 (95% CI 0.91-0.96) and 0.87 (95% CI 0.84-0.90) (Z = 1.58, P > 0.05), respectively. The diagnostic odds ratio of DWI (54.46, [95% CI 17.98-164.99]) was superior to that of PET/CT (15.77, [95% CI 8.19-30.37]). The Deeks' funnel plot asymmetry test showed no publication bias. The Spearman correlation coefficient test revealed no significant threshold effect. Lesion diameter and reference standard could be potential causes for the heterogeneity of both DWI and PET/CT studies, and quantitative or semi-quantitative parameters used would be a potential source of bias for PET/CT studies. CONCLUSION As a radiation-free technique, DWI may have similar performance compare with PET/CT in differentiating malignant pulmonary nodules or masses from benign ones.
Collapse
Affiliation(s)
- Jieqiong Liu
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China
| | - Xiaoying Xia
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China
| | - Qiao Zou
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China
| | - Xiaobin Xie
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China
| | - Yongxia Lei
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China
| | - Qi Wan
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China.
| | - Xinchun Li
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Yanjiangxilu No 151, Guangzhou, 510120, China.
| |
Collapse
|
45
|
Prognostic analysis of curatively resected pancreatic cancer using harmonized positron emission tomography radiomic features. Eur J Hybrid Imaging 2023; 7:5. [PMID: 36872413 PMCID: PMC9986192 DOI: 10.1186/s41824-023-00163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Texture features reflecting tumour heterogeneity enable us to investigate prognostic factors. The R package ComBat can harmonize the quantitative texture features among several positron emission tomography (PET) scanners. We aimed to identify prognostic factors among harmonized PET radiomic features and clinical information from pancreatic cancer patients who underwent curative surgery. METHODS Fifty-eight patients underwent preoperative enhanced dynamic computed tomography (CT) scanning and fluorodeoxyglucose PET/CT using four PET scanners. Using LIFEx software, we measured PET radiomic parameters including texture features with higher order and harmonized these PET parameters. For progression-free survival (PFS) and overall survival (OS), we evaluated clinical information, including age, TNM stage, and neural invasion, and the harmonized PET radiomic features based on univariate Cox proportional hazard regression. Next, we analysed the prognostic indices by multivariate Cox proportional hazard regression (1) by using either significant (p < 0.05) or borderline significant (p = 0.05-0.10) indices in the univariate analysis (first multivariate analysis) or (2) by using the selected features with random forest algorithms (second multivariate analysis). Finally, we checked these multivariate results by log-rank test. RESULTS Regarding the first multivariate analysis for PFS after univariate analysis, age was the significant prognostic factor (p = 0.020), and MTV and GLCM contrast were borderline significant (p = 0.051 and 0.075, respectively). Regarding the first multivariate analysis of OS, neural invasion, Shape sphericity and GLZLM LZLGE were significant (p = 0.019, 0.042 and 0.0076). In the second multivariate analysis, only MTV was significant (p = 0.046) for PFS, whereas GLZLM LZLGE was significant (p = 0.047), and Shape sphericity was borderline significant (p = 0.088) for OS. In the log-rank test, age, MTV and GLCM contrast were borderline significant for PFS (p = 0.08, 0.06 and 0.07, respectively), whereas neural invasion and Shape sphericity were significant (p = 0.03 and 0.04, respectively), and GLZLM LZLGE was borderline significant for OS (p = 0.08). CONCLUSIONS Other than the clinical factors, MTV and GLCM contrast for PFS and Shape sphericity and GLZLM LZLGE for OS may be prognostic PET parameters. A prospective multicentre study with a larger sample size may be warranted.
Collapse
|
46
|
Zhang N, Zhang X, Li J, Ren J, Li L, Dong W, Liu Y. CT-derived radiomic analysis for predicting the survival rate of patients with non-small cell lung cancer receiving radiotherapy. Phys Med 2023; 107:102546. [PMID: 36796178 DOI: 10.1016/j.ejmp.2023.102546] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 12/09/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Radiomics provides an opportunity to minimize adverse effects and optimize the efficacy of treatments noninvasively. This study aims to develop a computed tomography (CT) derived radiomic signature to predict radiological response for the patients with non-small cell lung cancer (NSCLC) receiving radiotherapy. METHODS Total 815 NSCLC patients receiving radiotherapy were sourced from public datasets. Using CT images of 281 NSCLC patients, we adopted genetic algorithm to establish a predictive radiomic signature for radiotherapy that had optimal C-index value by Cox model. Survival analysis and receiver operating characteristic curve were performed to estimate the predictive performance of the radiomic signature. Furthermore, radiogenomics analysis was performed in a dataset with matched images and transcriptome data. RESULTS Radiomic signature consisting of three features was established and then validated in the validation dataset (log-rank P = 0.0047) including 140 patient, and showed a significant predictive power in two independent datasets totaling 395 NSCLC patients with binary 2-year survival endpoint. Furthermore, the novel proposed radiomic nomogram significantly improved the prognostic performance (concordance index) of clinicopathological factors. Radiogenomics analysis linked our signature with important tumor biological processes (e.g. Mismatch repair, Cell adhesion molecules and DNA replication) associated with clinical outcomes. CONCLUSIONS The radiomic signature, reflecting tumor biological processes, could noninvasively predict therapeutic efficacy of NSCLC patients receiving radiotherapy and demonstrate unique advantage for clinical application.
Collapse
Affiliation(s)
- Nannan Zhang
- Modern Educational Technology and Experiment Center, Harbin Normal University, Harbin, China
| | - Xinxin Zhang
- College of Life Science and Technology, Harbin Normal University, Harbin, China
| | - Junheng Li
- Basic Medicine College, Harbin Medical University, Harbin, China
| | - Jie Ren
- Basic Medicine College, Harbin Medical University, Harbin, China
| | - Luyang Li
- Basic Medicine College, Harbin Medical University, Harbin, China
| | - Wenlei Dong
- Department of Radiotherapy Technology Center, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Yixin Liu
- Basic Medicine College, Harbin Medical University, Harbin, China.
| |
Collapse
|
47
|
Chen X, He L, Li Q, Liu L, Li S, Zhang Y, Liu Z, Huang Y, Mao Y, Chen X. Non-invasive prediction of microsatellite instability in colorectal cancer by a genetic algorithm-enhanced artificial neural network-based CT radiomics signature. Eur Radiol 2023; 33:11-22. [PMID: 35771245 DOI: 10.1007/s00330-022-08954-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/08/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The stratification of microsatellite instability (MSI) status assists clinicians in making treatment decisions for colorectal cancer (CRC) patients. This study aimed to establish a CT-based radiomics signature to predict MSI status in patients with CRC. METHODS A total of 837 CRC patients who underwent preoperative enhanced CT and had available MSI status data were recruited from two hospitals. Radiomics features were extracted from segmented tumours, and a series of data balancing and feature selection strategies were used to select MSI-related features. Finally, an MSI-related radiomics signature was constructed using a genetic algorithm-enhanced artificial neural network model. Combined and clinical models were constructed using multivariate logistic regression analyses by integrating the clinical factors with or without the signature. A Kaplan-Meier survival analysis was conducted to explore the prognostic information of the signature in patients with CRC. RESULTS Ten features were selected to construct a signature which showed robust performance in both the internal and external validation cohorts, with areas under the curves (AUC) of 0.788 and 0.775, respectively. The performance of the signature was comparable to that of the combined model (AUCs of 0.777 and 0.767, respectively) and it outperformed the clinical model constituting age and tumour location (AUCs of 0.768 and 0.623, respectively). Survival analysis demonstrated that the signature could stratify patients with stage II CRC according to prognosis (HR: 0.402, p = 0.029). CONCLUSIONS This study built a robust radiomics signature for identifying the MSI status of CRC patients, which may assist individualised treatment decisions. KEY POINTS • Our well-designed modelling strategies helped overcome the problem of data imbalance caused by the low incidence of MSI. • Genetic algorithm-enhanced artificial neural network-based CT radiomics signature can effectively distinguish the MSI status of CRC patients. • Kaplan-Meier survival analysis demonstrated that our signature could significantly stratify stage II CRC patients into high- and low-risk groups.
Collapse
Affiliation(s)
- Xiaobo Chen
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lan He
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China
| | - Qingshu Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Liu Liu
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Suyun Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yuan Zhang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, 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, 510080, China.
| | - Yun Mao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, 510180, China.
| |
Collapse
|
48
|
Huang Y, Jiang X, Xu H, Zhang D, Liu LN, Xia YX, Xu DK, Wu HJ, Cheng G, Shi YH. Preoperative prediction of mediastinal lymph node metastasis in non-small cell lung cancer based on 18F-FDG PET/CT radiomics. Clin Radiol 2023; 78:8-17. [PMID: 36192203 DOI: 10.1016/j.crad.2022.08.140] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/14/2022] [Accepted: 08/26/2022] [Indexed: 01/07/2023]
Abstract
AIM To establish and verify a 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT)-based radiomics nomogram to predict mediastinal lymph node metastasis (LNM) in non-small cell lung cancer (NSCLC) patients preoperatively. MATERIALS AND METHODS This retrospective study enrolled 155 NSCLC patients (primary cohort, n=93; validation cohort, n=62). For each patient, 2,704 radiomic features were extracted from the primary lung cancer regions. Four procedures including the Mann-Whitney U-test, Spearman's correlation analysis, minimum redundancy-maximum relevance (mRMR), and least absolute shrinkage and selection operator (LASSO) binary logistic regression were utilised for determining essential features and establishing a radiomics signature. After that, a nomogram was established. The nomogram's potential was assessed based on its discrimination, calibration, and clinical usefulness. The radiomics signature and nomogram predictive performances were evaluated with respect to the area under the receiver operating characteristic curve (AUC), specificity, accuracy, and sensitivity. RESULTS The radiomics signature composed of eight selected features had good discriminatory performance of LNM versus non-LNM groups an AUC of 0.851 and 0.826 in primary and validation cohorts, respectively. The nomogram also indicated good discrimination with an AUC of 0.869 and 0.847 in the primary and validation cohorts, respectively. Furthermore, good calibration was demonstrated utilising the nomogram. CONCLUSIONS An 18F-FDG PET/CT-based radiomics nomogram that integrates the radiomics signature and age was promoted to predict mediastinal LNM within NSCLC patients, which could potentially facilitate individualised therapy for mediastinal LNM before treatment. The nomogram was beneficial in clinical practice, as illustrated by decision curve analysis.
Collapse
Affiliation(s)
- Y Huang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - X Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - H Xu
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - D Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - L-N Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Y-X Xia
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - D-K Xu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - H-J Wu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - G Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Y-H Shi
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
49
|
Akamatsu G, Tsutsui Y, Daisaki H, Mitsumoto K, Baba S, Sasaki M. A review of harmonization strategies for quantitative PET. Ann Nucl Med 2023; 37:71-88. [PMID: 36607466 PMCID: PMC9902332 DOI: 10.1007/s12149-022-01820-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
PET can reveal in vivo biological processes at the molecular level. PET-derived quantitative values have been used as a surrogate marker for clinical decision-making in numerous clinical studies and trials. However, quantitative values in PET are variable depending on technical, biological, and physical factors. The variability may have a significant impact on a study outcome. Appropriate scanner calibration and quality control, standardization of imaging protocols, and any necessary harmonization strategies are essential to make use of PET as a biomarker with low bias and variability. This review summarizes benefits, limitations, and remaining challenges for harmonization of quantitative PET, including whole-body PET in oncology, brain PET in neurology, PET/MR, and non-18F PET imaging. This review is expected to facilitate harmonization of quantitative PET and to promote the contribution of PET-derived biomarkers to research and development in medicine.
Collapse
Affiliation(s)
- Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan. .,Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Yuji Tsutsui
- Department of Radiological Science, Faculty of Health Science, Junshin Gakuen University, 1-1-1 Chikushigaoka, Minami-ku, Fukuoka, 815-8510 Japan
| | - Hiromitsu Daisaki
- Department of Radiological Technology, Gunma Prefectural College of Health Sciences, 323-1 Kamioki-machi, Maebashi, Gunma 371-0052 Japan
| | - Katsuhiko Mitsumoto
- Department of Clinical Radiology Service, Kyoto University Hospital, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Shingo Baba
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Masayuki Sasaki
- Department of Medical Quantum Science, Faculty of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| |
Collapse
|
50
|
Hu Q, Li K, Yang C, Wang Y, Huang R, Gu M, Xiao Y, Huang Y, Chen L. The role of artificial intelligence based on PET/CT radiomics in NSCLC: Disease management, opportunities, and challenges. Front Oncol 2023; 13:1133164. [PMID: 36959810 PMCID: PMC10028142 DOI: 10.3389/fonc.2023.1133164] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Objectives Lung cancer has been widely characterized through radiomics and artificial intelligence (AI). This review aims to summarize the published studies of AI based on positron emission tomography/computed tomography (PET/CT) radiomics in non-small-cell lung cancer (NSCLC). Materials and methods A comprehensive search of literature published between 2012 and 2022 was conducted on the PubMed database. There were no language or publication status restrictions on the search. About 127 articles in the search results were screened and gradually excluded according to the exclusion criteria. Finally, this review included 39 articles for analysis. Results Classification is conducted according to purposes and several studies were identified at each stage of disease:1) Cancer detection (n=8), 2) histology and stage of cancer (n=11), 3) metastases (n=6), 4) genotype (n=6), 5) treatment outcome and survival (n=8). There is a wide range of heterogeneity among studies due to differences in patient sources, evaluation criteria and workflow of radiomics. On the whole, most models show diagnostic performance comparable to or even better than experts, and the common problems are repeatability and clinical transformability. Conclusion AI-based PET/CT Radiomics play potential roles in NSCLC clinical management. However, there is still a long way to go before being translated into clinical application. Large-scale, multi-center, prospective research is the direction of future efforts, while we need to face the risk of repeatability of radiomics features and the limitation of access to large databases.
Collapse
Affiliation(s)
- Qiuyuan Hu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yue Wang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Rong Huang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Mingqiu Gu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yuqiang Xiao
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| | - Long Chen
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| |
Collapse
|