1
|
Safarian A, Mirshahvalad SA, Farbod A, Nasrollahi H, Pirich C, Beheshti M. Artificial intelligence for tumor [ 18F]FDG-PET imaging: Advancement and future trends-part I. Semin Nucl Med 2025; 55:328-344. [PMID: 40158896 DOI: 10.1053/j.semnuclmed.2025.03.003] [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: 02/28/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
The advent of sophisticated image analysis techniques has facilitated the extraction of increasingly complex data, such as radiomic features, from various imaging modalities, including [18F]FDG PET/CT, a well-established cornerstone of oncological imaging. Furthermore, the use of artificial intelligence (AI) algorithms has shown considerable promise in enhancing the interpretation of these quantitative parameters. Additionally, AI-driven models enable the integration of parameters from multiple imaging modalities along with clinical data, facilitating the development of comprehensive models with significant clinical impact. However, challenges remain regarding standardization and validation of the AI-powered models, as well as their implementation in real-world clinical practice. The variability in imaging acquisition protocols, segmentation methods, and feature extraction approaches across different institutions necessitates robust harmonization efforts to ensure reproducibility and clinical utility. Moreover, the successful translation of AI models into clinical practice requires prospective validation in large cohorts, as well as seamless integration into existing workflows to assess their ability to enhance clinicians' performance. This review aims to provide an overview of the literature and highlight three key applications: diagnostic impact, prediction of treatment response, and long-term patient prognostication. In the first part, we will focus on head and neck, lung, breast, gastroesophageal, colorectal, and gynecological malignancies.
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
| | - Abolfazl Farbod
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Nasrollahi
- 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
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
2
|
Gemmell AJ, Brown CM, Ray S, Small A. Robustness of textural analysis features in quantitative 99 mTc and 177Lu SPECT-CT phantom acquisitions. EJNMMI Phys 2025; 12:40. [PMID: 40244535 PMCID: PMC12006590 DOI: 10.1186/s40658-025-00749-0] [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: 07/08/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Textural Analysis features in molecular imaging require to be robust under repeat measurement and to be independent of volume for optimum use in clinical studies. Recent EANM and SNMMI guidelines for radiomics provide advice on the potential use of phantoms to identify robust features (Hatt in EJNMMI, 2022). This study applies the suggested phantoms to use in SPECT quantification for two radionuclides, 99 mTc and 177Lu. METHODS Acquisitions were made with a uniform phantom to test volume dependency and with a customised 'Revolver' phantom, based on the PET phantom described in Hatt (EJNMMI, 2022) but with local adaptations for SPECT. Each phantom was filled separately with 99 mTc and 177Lu. Sixty-seven Textural Analysis features were extracted and tested for robustness and volume dependency. RESULTS Features showing high volume dependency or high Coefficient of Variation (indicating poor repeatability) were removed from the list of features that may be suitable for use in clinical studies. After feature reduction, there were 39 features for 99 mTc and 33 features for 177Lu remaining. CONCLUSION The use of a uniform phantom to test volume dependency and a Revolver phantom to identify repeatable Textural Analysis features is possible for quantitative SPECT using 99 mTc or 177Lu. Selection of such features is likely to be centre-dependent due to differences in camera performance as well as acquisition and reconstruction protocols.
Collapse
Affiliation(s)
- Alastair J Gemmell
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK.
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK.
| | - Colin M Brown
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, UK
| | - Alexander Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK
| |
Collapse
|
3
|
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
|
4
|
Zhang Z, Ding Y, Lin K, Ban W, Ding L, Sun Y, Fu C, Ren Y, Han C, Zhang X, Wei X, Hu S, Zhao Y, Cao L, Wang J, Nazarian S, Cao Y, Zheng L, Zhang M, Fu J, Li J, Han X, Qian D, Huang D. Development of an MRI based artificial intelligence model for the identification of underlying atrial fibrillation after ischemic stroke: a multicenter proof-of-concept analysis. EClinicalMedicine 2025; 81:103118. [PMID: 40040863 PMCID: PMC11876936 DOI: 10.1016/j.eclinm.2025.103118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Background Atrial fibrillation (AF) represents a major risk factor of ischemic stroke recurrence with serious management implications. However, it often remains undiagnosed due to lack of standard or prolonged cardiac rhythm monitoring. We aim to create a novel end-to-end artificial intelligence (AI) model that uses MRI data to rapidly identify high AF risk in patients who suffer from an acute ischemic stroke. Methods This study comprises an internal retrospective cohort and a prospective cohort from Shanghai sixth people's hospital to train and validate an MRI-based AI model. Between January 1, 2018 and December 31, 2021, 510 patients were retrospectively enrolled for algorithm development and performance was measured using fivefold cross-validation. Patients from this trial were registered with http://www.chictr.org.cn, ChiCTR2200056385. Between September 1, 2022 and July 31, 2023, 73 patients were prospectively enrolled for algorithm test. An external cohort of 175 patients from Huashan Hospital, Minhang Hospital, and Shanghai Tenth People's Hospital was also enrolled retrospectively for further model validation. A combined classifier leveraging pre-defined radiomics features and de novo features extracted by convolutional neural network (CNN) was proposed to identify underlying AF in acute ischemic stroke patients. Area under the curve (AUC), sensitivity, specificity, accuracy, positive predictive value, and negative predictive value were calculated for model evaluation. Findings The top-performing combined classifier achieved an AUC of 0.94 (95% CI, 0.90-0.98) in the internal retrospective validation group, 0.85 (95% CI, 0.79-0.91) in the external validation group, and 0.87 (95% CI, 0.90-0.98) in the prospective test group. Based on subgroup analysis, the AI model performed well in female patients, patients with NIHSS > 4 or CHA2DS2-VASc ≤ 3, with the AUC of 0.91, 0.94, and 0.90, respectively. More importantly, our proposed model identified all the AF patients that were diagnosed with Holter monitoring during index stroke admission. Interpretation Our work suggested a potential association between brain ischemic lesion pattern on MR images and underlying AF. Furthermore, with additional validation, the AI model we developed may serve as a rapid screening tool for AF in clinical practice of stroke units. Funding This work was supported by grants from the National Natural Science Foundation of China (NSFC, Grant Number: 81871102 and 82172068); Shanghai Jiao Tong University School of Medicine, Two-Hundred Talent Program as Research Doctor (Grant Number: SBR202204); Municipal Science and Technology Commission Medical Innovation Project of Shanghai, (Grant/Award Number: 20Y11910200); Research Physician Program of Shanghai Shen Kang Hospital Development Center (Grant Number: SHD2022CRD039) to Dr. Dong Huang and the SJTU Trans-med Awards Research (No. 20220101) to Dahong Qian.
Collapse
Affiliation(s)
- Zijie Zhang
- Heart Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Ding
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kaibin Lin
- Heart Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenli Ban
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Luyue Ding
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yudong Sun
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanliang Fu
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yihang Ren
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Can Han
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Zhang
- Heart Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoer Wei
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shundong Hu
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wang
- School of Computer and Computational Science, Hangzhou City University, Hangzhou, China
| | - Saman Nazarian
- Section for Cardiac Electrophysiology, Department of Medicine/Cardiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ying Cao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Lan Zheng
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Min Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianliang Fu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingbo Li
- Heart Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dahong Qian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Dong Huang
- Heart Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
6
|
Hinzpeter R, Kulanthaivelu R, Kohan A, Murad V, Mirshahvalad SA, Avery L, Ortega C, Metser U, Hope A, Yeung J, McInnis M, Veit-Haibach P. Predictive [ 18F]-FDG PET/CT-Based Radiogenomics Modelling of Driver Gene Mutations in Non-small Cell Lung Cancer. Acad Radiol 2024; 31:5314-5323. [PMID: 38997880 DOI: 10.1016/j.acra.2024.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate whether [18F]-FDG PET/CT-derived radiomics may correlate with driver gene mutations in non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS In this IRB-approved retrospective study, 203 patients with surgically treated NSCLC who underwent subsequent genomic analysis of the primary tumour at our institution between December 2004 and January 2014 were identified. Of those, 128 patients (mean age 62.4 ± 10.8 years; range: 35-84) received preoperative [18F]-FDG PET/CT as part of their initial staging and thus were included in the study. PET and CT image segmentation and feature extraction were performed semi-automatically with an open-source software platform (LIFEx, Version 6.30, lifexsoft.org). Molecular profiles using different next-generation sequencing (NGS) panels were collected from a web-based resource (cBioPortal.ca for Cancer genomics). Two statistical models were then built to evaluate the predictive ability of [18F]-FDG PET/CT-derived radiomics features for driver gene mutations in NSCLC. RESULTS More than half (68/128, 53%) of all tumour samples harboured three or more gene mutations. Overall, 55% of tumour samples demonstrated a mutation in TP53, 26% of samples had alterations in KRAS and 17% in EGFR. Extensive statistical analysis resulted in moderate to good predictive ability. The highest Youden Index for TP53 was achieved using combined PET/CT features (0.70), for KRAS using PET features only (0.57) and for EGFR using CT features only (0.60). CONCLUSION Our study demonstrated a moderate to good correlation between radiomics features and driver gene mutations in NSCLC, indicating increased predictive ability of genomic profiles using combined [18F]-FDG PET/CT-derived radiomics features.
Collapse
Affiliation(s)
- Ricarda Hinzpeter
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.).
| | - Roshini Kulanthaivelu
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andres Kohan
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Vanessa Murad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Seyed Ali Mirshahvalad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Lisa Avery
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada (L.A.); Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada (L.A.)
| | - Claudia Ortega
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Ur Metser
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andrew Hope
- Department of Radiation Oncology, University Health Network, Toronto, Canada (A.H.)
| | - Jonathan Yeung
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada (J.Y.)
| | - Micheal McInnis
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Patrick Veit-Haibach
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| |
Collapse
|
7
|
Shenouda M, Shaikh A, Deutsch I, Mitchell O, Kindler HL, Armato SG. Radiomics for differentiation of somatic BAP1 mutation on CT scans of patients with pleural mesothelioma. J Med Imaging (Bellingham) 2024; 11:064501. [PMID: 39669009 PMCID: PMC11633667 DOI: 10.1117/1.jmi.11.6.064501] [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: 07/06/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose The BRCA1-associated protein 1 (BAP1) gene is of great interest because somatic (BAP1) mutations are the most common alteration associated with pleural mesothelioma (PM). Further, germline mutation of the BAP1 gene has been linked to the development of PM. This study aimed to explore the potential of radiomics on computed tomography scans to identify somatic BAP1 gene mutations and assess the feasibility of radiomics in future research in identifying germline mutations. Approach A cohort of 149 patients with PM and known somatic BAP1 mutation status was collected, and a previously published deep learning model was used to first automatically segment the tumor, followed by radiologist modifications. Image preprocessing was performed, and texture features were extracted from the segmented tumor regions. The top features were selected and used to train 18 separate machine learning models using leave-one-out cross-validation (LOOCV). The performance of the models in distinguishing between BAP1-mutated (BAP1+) and BAP1 wild-type (BAP1-) tumors was evaluated using the receiver operating characteristic area under the curve (ROC AUC). Results A decision tree classifier achieved the highest overall AUC value of 0.69 (95% confidence interval: 0.60 and 0.77). The features selected most frequently through the LOOCV were all second-order (gray-level co-occurrence or gray-level size zone matrices) and were extracted from images with an applied transformation. Conclusions This proof-of-concept work demonstrated the potential of radiomics to differentiate among BAP1+/- in patients with PM. Future work will extend these methods to the assessment of germline BAP1 mutation status through image analysis for improved patient prognostication.
Collapse
Affiliation(s)
- Mena Shenouda
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | | | - Ilana Deutsch
- Northwestern University, Evanston, Illinois, United States
| | - Owen Mitchell
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Hedy L. Kindler
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Samuel G. Armato
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| |
Collapse
|
8
|
Wang Y, Yang G, Gao X, Li L, Zhu H, Yi H. Subregion-specific 18F-FDG PET-CT radiomics for the pre-treatment prediction of EGFR mutation status in solid lung adenocarcinoma. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:134-143. [PMID: 38737644 PMCID: PMC11087292 DOI: 10.62347/ddrr4923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
This study aimed to assess the efficacy of fluor-18 fluorodeoxyglucose (18F-FDG) PET/CT using sub-regional-based radiomics in predicting epidermal growth factor receptor (EGFR) mutation status in pretreatment patients with solid lung adenocarcinoma. A retrospective analysis included 269 patients (134 EGFR+ and 135 EGFR-) who underwent pretreatment 18F-FDG PET/CT scans and EGFR mutation testing. The most metabolically active intratumoral sub-region was identified, and radiomics features from whole tumors or sub-regional regions were used to build classification models. The dataset was split into a 7:3 ratio for training and independent testing. Feature subsets were determined by Pearson correlation and the Kruskal Wallis test and radiomics classifiers were built with support vector machines or logistic regressions. Evaluation metrics, including accuracy, area under the curve (AUC), sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were employed for different classifiers. Results indicated that the sub-region-based classifier outperformed the whole-tumor classifier in terms of accuracy (73.8% vs. 66.2%), AUC (0.768 vs. 0.632), specificity (65.0% vs. 50.0%), PPV (70.2% vs. 62.2%), and NPV (78.8% vs. 74.0%). The clinical classifier exhibited an accuracy of 75.0%, AUC of 0.768, sensitivity of 72.5%, specificity of 77.5%, PPV of 76.3%, and NPV of 73.8%. The combined classifier, incorporating sub-region analysis and clinical parameters, demonstrated further improvement with an accuracy of 77.5%, AUC of 0.807, sensitivity of 77.5%, specificity of 77.5%, and NPV of 77.5%. The study suggests that sub-region-based 18F-FDG PET/CT radiomics enhances EGFR mutation prediction in solid lung adenocarcinoma, providing a practical and cost-efficient alternative to invasive EGFR testing.
Collapse
Affiliation(s)
- Yun Wang
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Guang Yang
- Department of Physics, Shanghai Key Laboratory of Magnetic Resonance, East China Normal UniversityShanghai 200062, China
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Linfa Li
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Hongzhou Zhu
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| |
Collapse
|
9
|
Yuan L, An L, Zhu Y, Duan C, Kong W, Jiang P, Yu QQ. Machine Learning in Diagnosis and Prognosis of Lung Cancer by PET-CT. Cancer Manag Res 2024; 16:361-375. [PMID: 38699652 PMCID: PMC11063459 DOI: 10.2147/cmar.s451871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
As a disease with high morbidity and high mortality, lung cancer has seriously harmed people's health. Therefore, early diagnosis and treatment are more important. PET/CT is usually used to obtain the early diagnosis, staging, and curative effect evaluation of tumors, especially lung cancer, due to the heterogeneity of tumors and the differences in artificial image interpretation and other reasons, it also fails to entirely reflect the real situation of tumors. Artificial intelligence (AI) has been applied to all aspects of life. Machine learning (ML) is one of the important ways to realize AI. With the help of the ML method used by PET/CT imaging technology, there are many studies in the diagnosis and treatment of lung cancer. This article summarizes the application progress of ML based on PET/CT in lung cancer, in order to better serve the clinical. In this study, we searched PubMed using machine learning, lung cancer, and PET/CT as keywords to find relevant articles in the past 5 years or more. We found that PET/CT-based ML approaches have achieved significant results in the detection, delineation, classification of pathology, molecular subtyping, staging, and response assessment with survival and prognosis of lung cancer, which can provide clinicians a powerful tool to support and assist in critical daily clinical decisions. However, ML has some shortcomings such as slightly poor repeatability and reliability.
Collapse
Affiliation(s)
- Lili Yuan
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Lin An
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Yandong Zhu
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Chongling Duan
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Weixiang Kong
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| | - Qing-Qing Yu
- Jining NO.1 People’s Hospital, Shandong First Medical University, Jining, People’s Republic of China
| |
Collapse
|
10
|
Shao X, Ge X, Gao J, Niu R, Shi Y, Shao X, Jiang Z, Li R, Wang Y. Transfer learning-based PET/CT three-dimensional convolutional neural network fusion of image and clinical information for prediction of EGFR mutation in lung adenocarcinoma. BMC Med Imaging 2024; 24:54. [PMID: 38438844 PMCID: PMC10913633 DOI: 10.1186/s12880-024-01232-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND To introduce a three-dimensional convolutional neural network (3D CNN) leveraging transfer learning for fusing PET/CT images and clinical data to predict EGFR mutation status in lung adenocarcinoma (LADC). METHODS Retrospective data from 516 LADC patients, encompassing preoperative PET/CT images, clinical information, and EGFR mutation status, were divided into training (n = 404) and test sets (n = 112). Several deep learning models were developed utilizing transfer learning, involving CT-only and PET-only models. A dual-stream model fusing PET and CT and a three-stream transfer learning model (TS_TL) integrating clinical data were also developed. Image preprocessing includes semi-automatic segmentation, resampling, and image cropping. Considering the impact of class imbalance, the performance of the model was evaluated using ROC curves and AUC values. RESULTS TS_TL model demonstrated promising performance in predicting the EGFR mutation status, with an AUC of 0.883 (95%CI = 0.849-0.917) in the training set and 0.730 (95%CI = 0.629-0.830) in the independent test set. Particularly in advanced LADC, the model achieved an AUC of 0.871 (95%CI = 0.823-0.919) in the training set and 0.760 (95%CI = 0.638-0.881) in the test set. The model identified distinct activation areas in solid or subsolid lesions associated with wild and mutant types. Additionally, the patterns captured by the model were significantly altered by effective tyrosine kinase inhibitors treatment, leading to notable changes in predicted mutation probabilities. CONCLUSION PET/CT deep learning model can act as a tool for predicting EGFR mutation in LADC. Additionally, it offers clinicians insights for treatment decisions through evaluations both before and after treatment.
Collapse
Affiliation(s)
- Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China.
| | - Xinyu Ge
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Jianxiong Gao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Rong Niu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Yunmei Shi
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Zhenxing Jiang
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Renyuan Li
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China.
| |
Collapse
|
11
|
Zhong R, Gao T, Li J, Li Z, Tian X, Zhang C, Lin X, Wang Y, Gao L, Hu K. The global research of artificial intelligence in lung cancer: a 20-year bibliometric analysis. Front Oncol 2024; 14:1346010. [PMID: 38371616 PMCID: PMC10869611 DOI: 10.3389/fonc.2024.1346010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/18/2024] [Indexed: 02/20/2024] Open
Abstract
Background Lung cancer (LC) is the second-highest incidence and the first-highest mortality cancer worldwide. Early screening and precise treatment of LC have been the research hotspots in this field. Artificial intelligence (AI) technology has advantages in many aspects of LC and widely used such as LC early diagnosis, LC differential classification, treatment and prognosis prediction. Objective This study aims to analyze and visualize the research history, current status, current hotspots, and development trends of artificial intelligence in the field of lung cancer using bibliometric methods, and predict future research directions and cutting-edge hotspots. Results A total of 2931 articles published between 2003 and 2023 were included, contributed by 15,848 authors from 92 countries/regions. Among them, China (40%) with 1173 papers,USA (24.80%) with 727 papers and the India(10.2%) with 299 papers have made outstanding contributions in this field, accounting for 75% of the total publications. The primary research institutions were Shanghai Jiaotong University(n=66),Chinese Academy of Sciences (n=63) and Harvard Medical School (n=52).Professor Qian Wei(n=20) from Northeastern University in China were ranked first in the top 10 authors while Armato SG(n=458 citations) was the most co-cited authors. Frontiers in Oncology(121 publications; IF 2022,4.7; Q2) was the most published journal. while Radiology (3003 citations; IF 2022, 19.7; Q1) was the most co-cited journal. different countries and institutions should further strengthen cooperation between each other. The most common keywords were lung cancer, classification, cancer, machine learning and deep learning. Meanwhile, The most cited papers was Nicolas Coudray et al.2018.NAT MED(1196 Total Citations). Conclusions Research related to AI in lung cancer has significant application prospects, and the number of scholars dedicated to AI-related research on lung cancer is continually growing. It is foreseeable that non-invasive diagnosis and precise minimally invasive treatment through deep learning and machine learning will remain a central focus in the future. Simultaneously, there is a need to enhance collaboration not only among various countries and institutions but also between high-quality medical and industrial entities.
Collapse
Affiliation(s)
- Ruikang Zhong
- Beijing University of Chinese Medicine, Beijing, China
| | - Tangke Gao
- Beijing University of Chinese Medicine, Beijing, China
| | - Jinghua Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Zexing Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Xue Tian
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chi Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Ximing Lin
- Beijing University of Chinese Medicine, Beijing, China
| | - Yuehui Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Lei Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Ge X, Gao J, Niu R, Shi Y, Shao X, Wang Y, Shao X. New research progress on 18F-FDG PET/CT radiomics for EGFR mutation prediction in lung adenocarcinoma: a review. Front Oncol 2023; 13:1242392. [PMID: 38094613 PMCID: PMC10716448 DOI: 10.3389/fonc.2023.1242392] [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: 06/19/2023] [Accepted: 11/16/2023] [Indexed: 11/09/2024] Open
Abstract
Lung cancer, the most frequently diagnosed cancer worldwide, is the leading cause of cancer-associated deaths. In recent years, significant progress has been achieved in basic and clinical research concerning the epidermal growth factor receptor (EGFR), and the treatment of lung adenocarcinoma has also entered a new era of individualized, targeted therapies. However, the detection of lung adenocarcinoma is usually invasive. 18F-FDG PET/CT can be used as a noninvasive molecular imaging approach, and radiomics can acquire high-throughput data from standard images. These methods play an increasingly prominent role in diagnosing and treating cancers. Herein, we reviewed the progress in applying 18F-FDG PET/CT and radiomics in lung adenocarcinoma clinical research and how these data are analyzed via traditional statistics, machine learning, and deep learning to predict EGFR mutation status, all of which achieved satisfactory results. Traditional statistics extract features effectively, machine learning achieves higher accuracy with complex algorithms, and deep learning obtains significant results through end-to-end methods. Future research should combine these methods to achieve more accurate predictions, providing reliable evidence for the precision treatment of lung adenocarcinoma. At the same time, facing challenges such as data insufficiency and high algorithm complexity, future researchers must continuously explore and optimize to better apply to clinical practice.
Collapse
Affiliation(s)
- Xinyu Ge
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Jianxiong Gao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Rong Niu
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Yunmei Shi
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Yuetao Wang
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Xiaonan Shao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| |
Collapse
|
13
|
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
|
14
|
Tan JL, Xia L, Sun SG, Zeng H, Lu DY, Cheng XJ. Prediction of EGFR mutation status in lung adenocarcinoma based on 18F-FDG PET/CT radiomic features. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2023; 13:230-244. [PMID: 38023818 PMCID: PMC10656631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
The earlier identification of EGFR mutation status in lung adenocarcinoma patients is crucial for treatment decision-making. Radiomics, which involves high-throughput extraction of imaging features from medical images for quantitative analysis, can quantify tumor heterogeneity and assess tumor biology non-invasively. This field has gained attention from researchers in recent years. The aim of this study is to establish a model based on 18F-FDG PET/CT radiomic features to predict the epidermal growth factor receptor (EGFR) mutation status of lung adenocarcinoma and evaluate its performance. 155 patients with lung adenocarcinoma who underwent 18F-FDG PET/CT scans and EGFR gene detection before treatment were retrospectively analyzed. The LIFEx packages was used to perform 3D volume of interest (VOI) segmentation manually on DICOM images and extract 128 radiomic features. The Wilcoxon rank sum test and least absolute shrinkage and selection operator (LASSO) regression algorithm were applied to filter the radiomic features and establish models. The performance of the models was evaluated by the receiver operating characteristic (ROC) curve and the area under the curve (AUC). Among the models we have built, the radiomic model based on 18F-FDG PET/CT has the best prediction performance for EGFR gene mutation status, with an AUC of 0.90 (95% CI 0.84~0.96) in the training set and 0.79 (95% CI 0.64~0.94) in the test set. In conclusion, we have established a radiomics model based on 18F-FDG PET/CT, which has good predictive performance in identifying EGFR gene mutation status in lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Jian-Ling Tan
- Department of Nuclear Medicine, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan UniversityWuhan, Hubei, China
| | - Liang Xia
- Department of Nuclear Medicine, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Su-Guang Sun
- Department of Nuclear Medicine, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan UniversityWuhan, Hubei, China
| | - Hui Zeng
- Department of Nuclear Medicine, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan UniversityWuhan, Hubei, China
| | - Di-Yu Lu
- Department of Nuclear Medicine, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Xiao-Jie Cheng
- Department of Nuclear Medicine, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan UniversityWuhan, Hubei, China
| |
Collapse
|
15
|
Bevilacqua R, Barbarossa F, Fantechi L, Fornarelli D, Paci E, Bolognini S, Giammarchi C, Lattanzio F, Paciaroni L, Riccardi GR, Pelliccioni G, Biscetti L, Maranesi E. Radiomics and Artificial Intelligence for the Diagnosis and Monitoring of Alzheimer's Disease: A Systematic Review of Studies in the Field. J Clin Med 2023; 12:5432. [PMID: 37629474 PMCID: PMC10455452 DOI: 10.3390/jcm12165432] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
The use of radiomics and artificial intelligence applied for the diagnosis and monitoring of Alzheimer's disease has developed in recent years. However, this approach is not yet completely applicable in clinical practice. The aim of this paper is to provide a systematic analysis of the studies that have included the use of radiomics from different imaging techniques and artificial intelligence for the diagnosis and monitoring of Alzheimer's disease in order to improve the clinical outcomes and quality of life of older patients. A systematic review of the literature was conducted in February 2023, analyzing manuscripts and articles of the last 5 years from the PubMed, Scopus and Embase databases. All studies concerning discrimination among Alzheimer's disease, Mild Cognitive Impairment and healthy older people performing radiomics analysis through machine and deep learning were included. A total of 15 papers were included. The results showed a very good performance of this approach in the differentiating Alzheimer's disease patients-both at the dementia and pre-dementia phases of the disease-from healthy older people. In summary, radiomics and AI can be valuable tools for diagnosing and monitoring the progression of Alzheimer's disease, potentially leading to earlier and more accurate diagnosis and treatment. However, the results reported by this review should be read with great caution, keeping in mind that imaging alone is not enough to identify dementia due to Alzheimer's.
Collapse
Affiliation(s)
- Roberta Bevilacqua
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| | - Federico Barbarossa
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| | - Lorenzo Fantechi
- Unit of Nuclear Medicine, IRCCS INRCA, 60127 Ancona, Italy; (L.F.); (D.F.)
| | - Daniela Fornarelli
- Unit of Nuclear Medicine, IRCCS INRCA, 60127 Ancona, Italy; (L.F.); (D.F.)
| | - Enrico Paci
- Unit of Radiology, IRCCS INRCA, 60127 Ancona, Italy;
| | - Silvia Bolognini
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| | - Cinzia Giammarchi
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| | - Fabrizia Lattanzio
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| | - Lucia Paciaroni
- Unit of Neurology, IRCCS INRCA, 60127 Ancona, Italy; (L.P.); (G.P.)
| | | | | | | | - Elvira Maranesi
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (R.B.); (F.B.); (S.B.); (C.G.); (F.L.); (E.M.)
| |
Collapse
|
16
|
Ashrafinia S, Dalaie P, Schindler TH, Pomper MG, Rahmim A. Standardized Radiomics Analysis of Clinical Myocardial Perfusion Stress SPECT Images to Identify Coronary Artery Calcification. Cureus 2023; 15:e43343. [PMID: 37700937 PMCID: PMC10493172 DOI: 10.7759/cureus.43343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
PURPOSE Myocardial perfusion (MP) stress single-photon emission computed tomography (SPECT) is an established diagnostic test for patients suspected of coronary artery disease (CAD). Meanwhile, coronary artery calcification (CAC) scoring obtained from diagnostic CT is a highly sensitive test, offering incremental diagnostic information in identifying patients with significant CAD yet normal MP stress SPECT (MPSS) scans. However, after decades of wide utilization of MPSS, CAC is not commonly reimbursed (e.g. by the CMS), nor widely deployed in community settings. We studied the potential of complementary information deduced from the radiomics analysis of normal MPSS scans in predicting the CAC score. METHODS We collected data from 428 patients with normal (non-ischemic) MPSS (99mTc-sestamibi; consensus reading). A nuclear medicine physician verified iteratively reconstructed images (attenuation-corrected) to be free from fixed perfusion defects and artifactual attenuation. Three-dimensional images were automatically segmented into four regions of interest (ROIs), including myocardium and three vascular segments (left anterior descending [LAD]-left circumference [LCX]-right coronary artery [RCA]). We used our software package, standardized environment for radiomics analysis (SERA), to extract 487 radiomic features in compliance with the image biomarker standardization initiative (IBSI). Isotropic cubic voxels were discretized using fixed bin-number discretization (eight schemes). We first performed blind-to-outcome feature selection focusing on a priori usefulness, dynamic range, and redundancy of features. Subsequently, we performed univariate and multivariate machine learning analyses to predict CAC scores from i) selected radiomic features, ii) 10 clinical features, and iii) combined radiomics + clinical features. Univariate analysis invoked Spearman correlation with Benjamini-Hotchberg false-discovery correction. The multivariate analysis incorporated stepwise linear regression, where we randomly selected a 15% test set and divided the other 85% of data into 70% training and 30% validation sets. Training started from a constant (intercept) model, iteratively adding/removing features (stepwise regression), invoking the Akaike information criterion (AIC) to discourage overfitting. Validation was run similarly, except that the training output model was used as the initial model. We randomized training/validation sets 20 times, selecting the best model using log-likelihood for evaluation in the test set. Assessment in the test set was performed thoroughly by running the entire operation 50 times, subsequently employing Fisher's method to verify the significance of independent tests. RESULTS Unsupervised feature selection significantly reduced 8×487 features to 56. In univariate analysis, no feature survived the false-discovery rate (FDR) to directly correlate with CAC scores. Applying Fisher's method to the multivariate regression results demonstrated combining radiomics with the clinical features to enhance the significance of the prediction model across all cardiac segments. Conclusions: Our standardized and statistically robust multivariate analysis demonstrated significant prediction of the CAC score for all cardiac segments when combining MPSS radiomic features with clinical features, suggesting radiomics analysis can add diagnostic or prognostic value to standard MPSS for wide clinical usage.
Collapse
Affiliation(s)
- Saeed Ashrafinia
- Radiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Pejman Dalaie
- Radiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Martin G Pomper
- Radiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Arman Rahmim
- Physics and Astronomy, University of British Columbia, Vancouver, CAN
| |
Collapse
|
17
|
Ishimura M, Norikane T, Mitamura K, Yamamoto Y, Manabe Y, Murao M, Murota M, Kanaji N, Nishiyama Y. FDG PET texture indices as imaging biomarkers for epidermal growth factor receptor mutation status in lung adenocarcinoma. Sci Rep 2023; 13:6742. [PMID: 37185611 PMCID: PMC10130153 DOI: 10.1038/s41598-023-34061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
Identifying the epidermal growth factor receptor (EGFR) mutation status is important for the optimal treatment of patients with EGFR mutations. We investigated the relationship between 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) texture indices and EGFR mutation status in patients with newly diagnosed lung adenocarcinoma. We retrospectively analyzed data of patients with newly diagnosed lung adenocarcinoma who underwent pretreatment FDG PET/computed tomography and EGFR mutation testing between August 2014 and November 2020. Patients were divided into mutated EGFR and wild-type EGFR groups. The maximum standardized uptake value (SUVmax) and 31 texture indices for the primary tumor were calculated from PET images and compared between the two groups. Of the 66 patients included, 22 had mutated EGFR and 44 had wild-type EGFR. The SUVmax did not significantly differ between the two groups. Among the 31 evaluated texture indices, the following five showed a statistically significant difference between the groups: correlation (P = 0.003), gray-level nonuniformity for run (P = 0.042), run length nonuniformity (P = 0.02), coarseness (P = 0.006), and gray-level nonuniformity for zone (P = 0.04). Based on the preliminary results of this study in a small patient population, FDG PET texture indices may be potential imaging biomarkers for the EGFR mutation status in patients with newly diagnosed lung adenocarcinoma.
Collapse
Affiliation(s)
- Mariko Ishimura
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Takashi Norikane
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Katsuya Mitamura
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Yuka Yamamoto
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
| | - Yuri Manabe
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Mitsumasa Murao
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Makiko Murota
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Nobuhiro Kanaji
- Division of Hematology, Rheumatology, and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Yoshihiro Nishiyama
- Department of Radiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| |
Collapse
|
18
|
Du Y, Zhang S, Qiu Q, Zhang J, Fang Y, Zhao L, Wei W, Wang J, Wang J, Li X. The effect of hippocampal radiomic features and functional connectivity on the relationship between hippocampal volume and cognitive function in Alzheimer's disease. J Psychiatr Res 2023; 158:382-391. [PMID: 36646036 DOI: 10.1016/j.jpsychires.2023.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Hippocampal volume is associated with cognitive function in Alzheimer's disease (AD). Hippocampal radiomic features and resting-state functional connectivity (rs-FC) are promising biomarkers and correlate with AD pathology. However, few studies have been conducted on how hippocampal biomarkers affect the cognition-structure relationship. Therefore, we aimed to investigate the effects of hippocampal radiomic features and resting-state functional connectivity (rs-FC) on this relationship in AD. We enrolled 70 AD patients and 65 healthy controls (HCs). The FreeSurfer software was used to measure hippocampal volume. We selected hippocampal radiomic features to build a model to distinguish AD patients from HCs and used a seed-based approach to calculate the hippocampal rs-FC. Furthermore, we conducted mediation and moderation analyses to investigate the effect of hippocampal radiomic features and rs-FC on the relationship between hippocampal volume and cognition in AD. The results suggested that hippocampal radiomic features mediated the association between bilateral hippocampal volume and cognition in AD. Additionally, patients with AD showed weaker rs-FC between the bilateral hippocampus and right ventral posterior cingulate cortex and stronger rs-FC between the left hippocampus and left insula than HCs. The rs-FC between the hippocampus and insula moderated the relationship between hippocampal volume and cognition in AD, suggesting that this rs-FC could exacerbate or ameliorate the effects of hippocampal volume on cognition and may be essential in improving cognitive function in AD. Our findings may not only expand existing biological knowledge of the interrelationships among hippocampal biomarkers and cognition but also provide potential targets for treatment strategies for AD.
Collapse
Affiliation(s)
- Yang Du
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shaowei Zhang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qi Qiu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jianye Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yuan Fang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Lu Zhao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Wenjing Wei
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jinghua Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jinhong Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Xia Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
19
|
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: 2] [Impact Index Per Article: 1.0] [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
|
20
|
Agüloğlu N, Aksu A, Unat DS, Akyol M. The prognostic relationship of 18F-FDG PET/CT metabolic and volumetric parameters in metastatic ALK + NSCLC. Nucl Med Commun 2022; 43:1217-1224. [PMID: 36345766 DOI: 10.1097/mnm.0000000000001625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The aim of this study is to determine the role of metabolic and volumetric parameters obtained from 18Fluorine-Fluorodeoxyglucose PET/computed tomography (18F-FDG PET/CT) imaging on progression-free survival (PFS) and overall survival (OS) in patients with advanced nonsquamous cell lung carcinoma (NSCLC) with anaplastic lymphoma kinase (ALK) rearrangement. METHODS Pre and post-treatment PET/CT images of the ALK + NSCLC patients between January 2015 and July 2020 were evaluated. The highest standardized uptake value (SUVmax), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) values were obtained from pre-tyrosine kinase inhibitor (TKI) basal PET/CT (PETpre) and post-TKI PET/CT (PETpost) images. Total MTV (tMTV) and total TLG (tTLG) values were calculated by summing MTV and TLG values in all tumor foci. The change (Δ) in pSUVmax, pMTV, pTLG, tMTV and tTLG before and after treatment was calculated.The relationship of these parameters with OS and PFS was analyzed. RESULTS tTLGpre, tMTVpre, pTLGpre, pMTVpre, ∆SUVmax, ∆tMTV and ∆tTLG values were found to be associated with OS; ∆tMTV, ∆tTLG, tTLGpre, tMTVpre, pTLGpre and pMTVpre were associated with PFS. The cutoff values in both predicting OS and PFS were calculated as -31.6 and 391.1 for ∆tMTV and tTLGpre, respectively. In Cox regression analysis, ∆tMTV and stage for OS and ∆tMTV and tTLGpre for PFS were obtained as prognostic factors. CONCLUSIONS Metabolic and volumetric parameters, especially TLG values in the whole body before treatment and change in whole body MTV value, obtained from PET/CT may be useful in predicting prognosis and determining treatment strategies for patients with advanced ALK + NSCLC.
Collapse
Affiliation(s)
- Nurşin Agüloğlu
- Department of Nuclear Medicine, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, İzmir
| | - Ayşegül Aksu
- Department of Nuclear Medicine, Başakşehir Çam and Sakura City Hospital, İstanbul
| | - Damla S Unat
- Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital İzmir, Turkey
| | - Murat Akyol
- Department of Medical Oncology, Bakirçay University Medical School İzmir, Turkey
| |
Collapse
|
21
|
Hu N, Yan G, Wu Y, Wang L, Wang Y, Xiang Y, Lei P, Luo P. Recent and current advances in PET/CT imaging in the field of predicting epidermal growth factor receptor mutations in non-small cell lung cancer. Front Oncol 2022; 12:879341. [PMID: 36276079 PMCID: PMC9582655 DOI: 10.3389/fonc.2022.879341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are a significant treatment strategy for the management of non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutation status. Currently, EGFR mutation status is established based on tumor tissue acquired by biopsy or resection, so there is a compelling need to develop non-invasive, rapid, and accurate gene mutation detection methods. Non-invasive molecular imaging, such as positron emission tomography/computed tomography (PET/CT), has been widely applied to obtain the tumor molecular and genomic features for NSCLC treatment. Recent studies have shown that PET/CT can precisely quantify EGFR mutation status in NSCLC patients for precision therapy. This review article discusses PET/CT advances in predicting EGFR mutation status in NSCLC and their clinical usefulness.
Collapse
Affiliation(s)
- Na Hu
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Gang Yan
- Department of Nuclear Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yuhui Wu
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Li Wang
- School of Nursing, Guizhou Medical University, Guiyang, China
| | - Yang Wang
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yining Xiang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Pinggui Lei
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China,School of Public Health, Guizhou Medical University, Guiyang, China,*Correspondence: Pinggui Lei, ; Peng Luo,
| | - Peng Luo
- School of Public Health, Guizhou Medical University, Guiyang, China,*Correspondence: Pinggui Lei, ; Peng Luo,
| |
Collapse
|
22
|
Hannequin P, Decroisette C, Kermanach P, Berardi G, Bourbonne V. FDG PET and CT radiomics in diagnosis and prognosis of non-small-cell lung cancer. Transl Lung Cancer Res 2022; 11:2051-2063. [PMID: 36386457 PMCID: PMC9641045 DOI: 10.21037/tlcr-22-158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/22/2022] [Indexed: 09/13/2023]
Abstract
BACKGROUND 18F-FDG PET and CT radiomics has been the object of a wide research for over 20 years but its contribution to clinical practice remains not yet well established. We have investigated its impact versus that of only histo-clinical data, for the routine management of non-small-cell lung cancer (NSCLC). METHODS Our patients were retrospectively considered. They all had a FDG PET-CT and immuno-histo-chemistry (IHC) to assess PD-L1 expression at the beginning of the disease. A prognosis univariate and multivariate Cox survival analyses was performed for overall survival (OS) and progression free survival (PFS) prediction, including a training/testing procedure. Two sets of 47 PET and 47 CT radiomics features (RFs) were extracted. Difference between RFs according to PD-L1 expression, the histology status and the stage level were tested using suited non parametric statistical tests and the receiver operating characteristics (ROC) curve and the area under curve (AUC). RESULTS From 2017 to 2019, 212 NSCLC patients treated in our institution were included. The main conventional prognostic variables were stage and gender with a low added prognostic value in the models including PET and CT RFs. Neither PET nor CT RFs were significant to separate the different levels of PD-L1 expression. Several RFs differ between adenocarcinoma (ADC) and squamous cell carcinoma (SCC) tumours and a large number of PET and CT RFs are significantly linked to patient stage. CONCLUSIONS In our population, PET and CT RFs show their intrinsic power to predict survival but do not significantly improve OS and PFS prediction in the different multivariate models, in comparison to conventional data. It would seem necessary to carry out one's own survival analysis before determining a radiomics signature.
Collapse
Affiliation(s)
- Pascal Hannequin
- Annecy Nuclear Medicine Center, Le Pericles, B Allée de la Mandallaz, Metz-Tessy, France
| | - Chantal Decroisette
- Pneumology Department, CHANGE Annecy, 1 Avenue de l’hôpital, Metz-Tessy, France
| | - Pascale Kermanach
- Mont Blanc Histo-Pathology Laboratory, 40 Route de l’Aiglière, Argonay, France
| | - Giulia Berardi
- Pneumology Department, University Hospital la Tronche, Boulevard de la Chantourne, La Tronche, France
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, 2 Avenue Foch, Brest, France
| |
Collapse
|
23
|
Li S, Li Y, Zhao M, Wang P, Xin J. Combination of 18F-Fluorodeoxyglucose PET/CT Radiomics and Clinical Features for Predicting Epidermal Growth Factor Receptor Mutations in Lung Adenocarcinoma. Korean J Radiol 2022; 23:921-930. [PMID: 36047542 PMCID: PMC9434738 DOI: 10.3348/kjr.2022.0295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To identify epidermal growth factor receptor (EGFR) mutations in lung adenocarcinoma based on 18F-fluorodeoxyglucose (FDG) PET/CT radiomics and clinical features and to distinguish EGFR exon 19 deletion (19 del) and exon 21 L858R missense (21 L858R) mutations using FDG PET/CT radiomics. MATERIALS AND METHODS We retrospectively analyzed 179 patients with lung adenocarcinoma. They were randomly assigned to training (n = 125) and testing (n = 54) cohorts in a 7:3 ratio. A total of 2632 radiomics features were extracted from the tumor region of interest from the PET (1316) and CT (1316) images. Six PET/CT radiomics features that remained after the feature selection step were used to calculate the radiomics model score (rad-score). Subsequently, a combined clinical and radiomics model was constructed based on sex, smoking history, tumor diameter, and rad-score. The performance of the combined model in identifying EGFR mutations was assessed using a receiver operating characteristic (ROC) curve. Furthermore, in a subsample of 99 patients, a PET/CT radiomics model for distinguishing 19 del and 21 L858R EGFR mutational subtypes was established, and its performance was evaluated. RESULTS The area under the ROC curve (AUROC) and accuracy of the combined clinical and PET/CT radiomics models were 0.882 and 81.6%, respectively, in the training cohort and 0.837 and 74.1%, respectively, in the testing cohort. The AUROC and accuracy of the radiomics model for distinguishing between 19 del and 21 L858R EGFR mutational subtypes were 0.708 and 66.7%, respectively, in the training cohort and 0.652 and 56.7%, respectively, in the testing cohort. CONCLUSION The combined clinical and PET/CT radiomics model could identify the EGFR mutational status in lung adenocarcinoma with moderate accuracy. However, distinguishing between EGFR 19 del and 21 L858R mutational subtypes was more challenging using PET/CT radiomics.
Collapse
Affiliation(s)
- Shen Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yadi Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Zhao
- Pharmaceutical Diagnostics, GE Healthcare, Beijing, China
| | - Pengyuan Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
24
|
Agüloğlu N, Aksu A, Akyol M, Katgı N, Doksöz TÇ. IMPORTANCE of PRETREATMENT 18F-FDG PET/CT TEXTURE ANALYSIS in PREDICTING EGFR and ALK MUTATION in PATIENTS with NON-SMALL CELL LUNG CANCER. Nuklearmedizin 2022; 61:433-439. [PMID: 35977671 DOI: 10.1055/a-1868-4918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
OBJECTIVE Identification of anaplastic lymphoma kinase (ALK) and epidermal growth factor receptor (EGFR) mutation types is of great importance before treatment with tyrosine kinase inhibitors (TKIs). Radiomics is a new strategy for noninvasively predicting the genetic status of cancer. We aimed to evaluate the predictive power of 18F-FDG PET/CT-based radiomic features for mutational status before treatment in non-small cell lung cancer (NSCLC) and to develop a predictive model based on radiomic features. METHODS Images of patients who underwent 18F-FDG PET/CT for initial staging with the diagnosis of NSCLC between January 2015 and July 2020 were evaluated using LIFEx software. The region of interest (ROI) of the primary tumor was established and volumetric and textural features were obtained. Clinical data and radiomic data were evaluated with machine learning (ML) algorithms to create a model. RESULTS For EGFR mutation prediction, the most successful machine learning algorithm obtained with GLZLM_GLNU and clinical data was Naive Bayes (AUC: 0.751, MCC: 0.347, acc: 71.4%). For ALK rearrangement prediction, the most successful machine learning algorithm obtained with GLCM_correlation, GLZLM_LZHGE and clinical data was evaluated as Naive Bayes (AUC: 0.682, MCC: 0.221, acc: 77.4%). CONCLUSIONS In our study, we created prediction models based on radiomic analysis of 18F-FDG PET/CT images. Tissue analysis with ML algorithms are non-invasive methods for predicting ALK rearrangement and EGFR mutation status in NSCLC, which may be useful for targeted therapy selection in a clinical setting.
Collapse
Affiliation(s)
| | - Ayşegül Aksu
- Bahçeşehir Çam ve Sakura Hastanesi, İstanbul, Turkey
| | | | | | | |
Collapse
|
25
|
Yin X, Liao H, Yun H, Lin N, Li S, Xiang Y, Ma X. Artificial intelligence-based prediction of clinical outcome in immunotherapy and targeted therapy of lung cancer. Semin Cancer Biol 2022; 86:146-159. [PMID: 35963564 DOI: 10.1016/j.semcancer.2022.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
Lung cancer accounts for the main proportion of malignancy-related deaths and most patients are diagnosed at an advanced stage. Immunotherapy and targeted therapy have great advances in application in clinics to treat lung cancer patients, yet the efficacy is unstable. The response rate of these therapies varies among patients. Some biomarkers have been proposed to predict the outcomes of immunotherapy and targeted therapy, including programmed cell death-ligand 1 (PD-L1) expression and oncogene mutations. Nevertheless, the detection tests are invasive, time-consuming, and have high demands on tumor tissue. The predictive performance of conventional biomarkers is also unsatisfactory. Therefore, novel biomarkers are needed to effectively predict the outcomes of immunotherapy and targeted therapy. The application of artificial intelligence (AI) can be a possible solution, as it has several advantages. AI can help identify features that are unable to be used by humans and perform repetitive tasks. By combining AI methods with radiomics, pathology, genomics, transcriptomics, proteomics, and clinical data, the integrated model has shown predictive value in immunotherapy and targeted therapy, which significantly improves the precision treatment of lung cancer patients. Herein, we reviewed the application of AI in predicting the outcomes of immunotherapy and targeted therapy in lung cancer patients, and discussed the challenges and future directions in this field.
Collapse
Affiliation(s)
- Xiaomeng Yin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hu Liao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hong Yun
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Nan Lin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Shen Li
- West China School of Medicine, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Yu Xiang
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
26
|
Ghosh D, Mastej E, Jain R, Choi YS. Causal Inference in Radiomics: Framework, Mechanisms, and Algorithms. Front Neurosci 2022; 16:884708. [PMID: 35812228 PMCID: PMC9261933 DOI: 10.3389/fnins.2022.884708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/20/2022] [Indexed: 12/30/2022] Open
Abstract
The widespread use of machine learning algorithms in radiomics has led to a proliferation of flexible prognostic models for clinical outcomes. However, a limitation of these techniques is their black-box nature, which prevents the ability for increased mechanistic phenomenological understanding. In this article, we develop an inferential framework for estimating causal effects with radiomics data. A new challenge is that the exposure of interest is latent so that new estimation procedures are needed. We leverage a multivariate version of partial least squares for causal effect estimation. The methodology is illustrated with applications to two radiomics datasets, one in osteosarcoma and one in glioblastoma.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
- *Correspondence: Debashis Ghosh
| | - Emily Mastej
- Computational Biosciences Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rajan Jain
- Department of Radiology and Neurosurgery, New York University Langone Medical Center, New York, NY, United States
| | - Yoon Seong Choi
- Department of Radiology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
27
|
Manafi-Farid R, Askari E, Shiri I, Pirich C, Asadi M, Khateri M, Zaidi H, Beheshti M. [ 18F]FDG-PET/CT radiomics and artificial intelligence in lung cancer: Technical aspects and potential clinical applications. Semin Nucl Med 2022; 52:759-780. [PMID: 35717201 DOI: 10.1053/j.semnuclmed.2022.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Lung cancer is the second most common cancer and the leading cause of cancer-related death worldwide. Molecular imaging using [18F]fluorodeoxyglucose Positron Emission Tomography and/or Computed Tomography ([18F]FDG-PET/CT) plays an essential role in the diagnosis, evaluation of response to treatment, and prediction of outcomes. The images are evaluated using qualitative and conventional quantitative indices. However, there is far more information embedded in the images, which can be extracted by sophisticated algorithms. Recently, the concept of uncovering and analyzing the invisible data extracted from medical images, called radiomics, is gaining more attention. Currently, [18F]FDG-PET/CT radiomics is growingly evaluated in lung cancer to discover if it enhances the diagnostic performance or implication of [18F]FDG-PET/CT in the management of lung cancer. In this review, we provide a short overview of the technical aspects, as they are discussed in different articles of this special issue. We mainly focus on the diagnostic performance of the [18F]FDG-PET/CT-based radiomics and the role of artificial intelligence in non-small cell lung cancer, impacting the early detection, staging, prediction of tumor subtypes, biomarkers, and patient's outcomes.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Emran Askari
- Department of Nuclear Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mahboobeh Asadi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maziar Khateri
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib 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
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
28
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 1, Supradiaphragmatic Cancers. Diagnostics (Basel) 2022; 12:1329. [PMID: 35741138 PMCID: PMC9221970 DOI: 10.3390/diagnostics12061329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Radiomics is an upcoming field in nuclear oncology, both promising and technically challenging. To summarize the already undertaken work on supradiaphragmatic neoplasia and assess its quality, we performed a literature search in the PubMed database up to 18 February 2022. Inclusion criteria were: studies based on human data; at least one specified tumor type; supradiaphragmatic malignancy; performing radiomics on PET imaging. Exclusion criteria were: studies only based on phantom or animal data; technical articles without a clinically oriented question; fewer than 30 patients in the training cohort. A review database containing PMID, year of publication, cancer type, and quality criteria (number of patients, retrospective or prospective nature, independent validation cohort) was constructed. A total of 220 studies met the inclusion criteria. Among them, 119 (54.1%) studies included more than 100 patients, 21 studies (9.5%) were based on prospectively acquired data, and 91 (41.4%) used an independent validation set. Most studies focused on prognostic and treatment response objectives. Because the textural parameters and methods employed are very different from one article to another, it is complicated to aggregate and compare articles. New contributions and radiomics guidelines tend to help improving quality of the reported studies over the years.
Collapse
Affiliation(s)
- David Morland
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Elizabeth Katherine Anna Triumbari
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
29
|
Chen Q, Li Y, Cheng Q, Van Valkenburgh J, Sun X, Zheng C, Zhang R, Yuan R. EGFR Mutation Status and Subtypes Predicted by CT-Based 3D Radiomic Features in Lung Adenocarcinoma. Onco Targets Ther 2022; 15:597-608. [PMID: 35669165 PMCID: PMC9165655 DOI: 10.2147/ott.s352619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
Objective In this study, we aim to establish a non-invasive tool to predict epidermal growth factor receptor (EGFR) mutation status and subtypes based on radiomic features of computed tomography (CT). Methods A total of 233 lung adenocarcinoma patients were investigated and randomly divided into the training and test cohorts. In this study, 2300 radiomic features were extracted from original and filtered (Exponential, Laplacian of Gaussian, Logarithm, Gabor, Wavelet) CT images. The radiomic features were divided into four categories, including histogram, volumetric, morphologic, and texture features. An RF-BFE algorithm was developed to select the features for building the prediction models. Clinicopathological features (including age, gender, smoking status, TNM staging, maximum diameter, location, and growth pattern) were combined to establish an integrated model with radiomic features. ROC curve and AUC quantified the effectiveness of the predictor of EGFR mutation status and subtypes. Results A set of 10 features were selected to predict EGFR mutation status between EGFR mutant and wild type, while 9 selected features were used to predict mutation subtypes between exon 19 deletion and exon 21 L858R mutation. To predict the EGFR mutation status, the AUC of the training cohort was 0.778 and the AUC of the test cohort was 0.765. To predict the EGFR mutation subtypes, the AUC of training cohort was 0.725 and the AUC of test cohort was 0.657. The integrated model showed the most optimal predictive performance with EGFR mutation status (AUC = 0.870 and 0.759) and subtypes (AUC = 0.797 and 0.554) in the training and test cohorts. Conclusion CT-based radiomic features can extract information on tumor heterogeneity in lung adenocarcinoma. In addition, we have established a radiomic model and an integrated model to non-invasively predict the EGFR mutation status and subtypes of lung adenocarcinoma, which is conducive to saving clinical costs and guiding targeted therapy.
Collapse
Affiliation(s)
- Quan Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Yan Li
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qiguang Cheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Juno Van Valkenburgh
- Department of Radiology, Molecular Imaging Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiaotian Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Ruiguang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Correspondence: Ruiguang Zhang, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China, Email
| | - Rong Yuan
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
- Rong Yuan, Department of Radiology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China, Email
| |
Collapse
|
30
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
31
|
Katal S, Eibschutz LS, Saboury B, Gholamrezanezhad A, Alavi A. Advantages and Applications of Total-Body PET Scanning. Diagnostics (Basel) 2022; 12:diagnostics12020426. [PMID: 35204517 PMCID: PMC8871405 DOI: 10.3390/diagnostics12020426] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Recent studies have focused on the development of total-body PET scanning in a variety of fields such as clinical oncology, cardiology, personalized medicine, drug development and toxicology, and inflammatory/infectious disease. Given its ultrahigh detection sensitivity, enhanced temporal resolution, and long scan range (1940 mm), total-body PET scanning can not only image faster than traditional techniques with less administered radioactivity but also perform total-body dynamic acquisition at a longer delayed time point. These unique characteristics create several opportunities to improve image quality and can provide a deeper understanding regarding disease detection, diagnosis, staging/restaging, response to treatment, and prognostication. By reviewing the advantages of total-body PET scanning and discussing the potential clinical applications for this innovative technology, we can address specific issues encountered in routine clinical practice and ultimately improve patient care.
Collapse
Affiliation(s)
- Sanaz Katal
- Independent Researcher, Melbourne 3000, Australia;
| | - Liesl S. Eibschutz
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90007, USA; (L.S.E.); (A.G.)
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD 20892, USA;
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90007, USA; (L.S.E.); (A.G.)
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
32
|
Bu L, Tu N, Wang K, Zhou Y, Xie X, Han X, Lin H, Feng H. Relationship between 18F-FDG PET/CT Semi-Quantitative Parameters and International Association for the Study of Lung Cancer, American Thoracic Society/European Respiratory Society Classification in Lung Adenocarcinomas. Korean J Radiol 2022; 23:112-123. [PMID: 34983098 PMCID: PMC8743143 DOI: 10.3348/kjr.2021.0455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/30/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022] Open
Abstract
Objective To investigate the relationship between 18F-FDG PET/CT semi-quantitative parameters and the International Association for the Study of Lung Cancer, American Thoracic Society/European Respiratory Society (IASLC/ATS/ERS) histopathologic classification, including histological subtypes, proliferation activity, and somatic mutations. Materials and Methods This retrospective study included 419 patients (150 males, 269 females; median age, 59.0 years; age range, 23.0–84.0 years) who had undergone surgical removal of stage IA–IIIA lung adenocarcinoma and had preoperative PET/CT data of lung tumors. The maximum standardized uptake values (SUVmax), background-subtracted volume (BSV), and background-subtracted lesion activity (BSL) derived from PET/CT were measured. The IASLC/ATS/ERS subtypes, Ki67 score, and epidermal growth factor/anaplastic lymphoma kinase (EGFR/ALK) mutation status were evaluated. The PET/CT semi-quantitative parameters were compared between the tumor subtypes using the Mann–Whitney U test or the Kruskal–Wallis test. The optimum cutoff values of the PET/CT semi-quantitative parameters for distinguishing the IASLC/ATS/ERS subtypes were calculated using receiver operating characteristic curve analysis. The correlation between the PET/CT semi-quantitative parameters and pathological parameters was analyzed using Spearman’s correlation. Statistical significance was set at p < 0.05. Results SUVmax, BSV, and BSL values were significantly higher in invasive adenocarcinoma (IA) than in minimally IA (MIA), and the values were higher in MIA than in adenocarcinoma in situ (AIS) (all p < 0.05). Remarkably, an SUVmax of 0.90 and a BSL of 3.62 were shown to be the optimal cutoff values for differentiating MIA from AIS, manifesting as pure ground-glass nodules with 100% sensitivity and specificity. Metabolic-volumetric parameters (BSV and BSL) were better potential independent factors than metabolic parameters (SUVmax) in differentiating growth patterns. SUVmax and BSL, rather than BSV, were strongly or moderately correlated with Ki67 in most subtypes, except for the micropapillary and solid predominant groups. PET/CT parameters were not correlated with EGFR/ALK mutation status. Conclusion As noninvasive surrogates, preoperative PET/CT semi-quantitative parameters could imply IASLC/ATS/ERS subtypes and Ki67 index and thus may contribute to improved management of precise surgery and postoperative adjuvant therapy.
Collapse
Affiliation(s)
- Lihong Bu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Tu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Wang
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Zhou
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinli Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiqin Lin
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyan Feng
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
33
|
Caruso D, Polici M, Lauri C, Laghi A. Radiomics and artificial intelligence. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
34
|
Guo H, Xu K, Duan G, Wen L, He Y. Progress and future prospective of FDG-PET/CT imaging combined with optimized procedures in lung cancer: toward precision medicine. Ann Nucl Med 2022; 36:1-14. [PMID: 34727331 DOI: 10.1007/s12149-021-01683-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022]
Abstract
With a 5-year overall survival of approximately 20%, lung cancer has always been the number one cancer-specific killer all over the world. As a fusion of positron emission computed tomography (PET) and computed tomography (CT), PET/CT has revolutionized cancer imaging over the past 20 years. In this review, we focused on the optimization of the function of 18F-flurodeoxyglucose (FDG)-PET/CT in diagnosis, prognostic prediction and therapy management of lung cancers by computer programs. FDG-PET/CT has demonstrated a surprising role in development of therapeutic biomarkers, prediction of therapeutic responses and long-term survival, which could be conducive to solving existing dilemmas. Meanwhile, novel tracers and optimized procedures are also developed to control the quality and improve the effect of PET/CT. With the continuous development of some new imaging agents and their clinical applications, application value of PET/CT has broad prospects in this area.
Collapse
Affiliation(s)
- Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
- School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai, 200092, China
| | - Kandi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
- School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai, 200092, China
| | - Guangxin Duan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Ling Wen
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
- School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
35
|
Chen W, Hua Y, Mao D, Wu H, Tan M, Ma W, Huang X, Lu J, Li C, Li M. A Computed Tomography-Derived Radiomics Approach for Predicting Uncommon EGFR Mutation in Patients With NSCLC. Front Oncol 2021; 11:722106. [PMID: 34976788 PMCID: PMC8716946 DOI: 10.3389/fonc.2021.722106] [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: 06/08/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
PURPOSE This study aims to develop a CT-based radiomics approach for identifying the uncommon epidermal growth factor receptor (EGFR) mutation in patients with non-small cell lung cancer (NSCLC). METHODS This study involved 223 NSCLC patients (107 with uncommon EGFR mutation-positive and 116 with uncommon EGFR mutation-negative). A total of 1,269 radiomics features were extracted from the non-contrast-enhanced CT images after image segmentation and preprocessing. Support vector machine algorithm was used for feature selection and model construction. Receiver operating characteristic curve analysis was applied to evaluate the performance of the radiomics signature, the clinicopathological model, and the integrated model. A nomogram was developed and evaluated by using the calibration curve and decision curve analysis. RESULTS The radiomics signature demonstrated a good performance for predicting the uncommon EGFR mutation in the training cohort (area under the curve, AUC = 0.802; 95% confidence interval, CI: 0.736-0.858) and was verified in the validation cohort (AUC = 0.791, 95% CI: 0.642-0.899). The integrated model combined radiomics signature with clinicopathological independent predictors exhibited an incremental performance compared with the radiomics signature or the clinicopathological model. A nomogram based on the integrated model was developed and showed good calibration (Hosmer-Lemeshow test, P = 0.92 in the training cohort and 0.608 in the validation cohort) and discrimination capacity (AUC of 0.816 in the training cohort and 0.795 in the validation cohort). CONCLUSION Radiomics signature combined with the clinicopathological features can predict uncommon EGFR mutation in NSCLC patients.
Collapse
Affiliation(s)
| | - Yanqing Hua
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Sheng S, Guo B, Wang Z, Zhang Z, Zhou J, Huo Z. Aberrant Methylation and Immune Microenvironment Are Associated With Overexpressed Fibronectin 1: A Diagnostic and Prognostic Target in Head and Neck Squamous Cell Carcinoma. Front Mol Biosci 2021; 8:753563. [PMID: 34746236 PMCID: PMC8563786 DOI: 10.3389/fmolb.2021.753563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Fibronectin 1 (FN1) is involved in cell adhesion and migration processes such as metastasis, wound healing, embryogenesis, blood coagulation, and host defense. However, the role of FN1 in the diagnosis and prognosis of head and neck squamous cell carcinoma (HNSCC) is far from understood. Methods: FN1 expression profiles and clinical parameters from multiple HNSCC datasets were applied to evaluate the association between FN1 expression and HNSCC survival. We also identified FN1 expression in the mRNA and protein levels in 20 pairs of clinical samples by quantitative polymerase chain reaction (qPCR) and immunohistochemistry. Receiver operator characteristic (ROC) analysis was used to demonstrate the potential diagnostic value of FN1 in HNSCC. Aberrant methylation PPI networks were established using multiple bioinformatic tools based on TCGA database. The immune microenvironment and levels of immune checkpoints were investigated between groups with high and low FN1 expression. Results: FN1 was significantly upregulated in HNSCC compared with para-carcinoma tissues on the basis of TCGA database and our clinical samples. Univariate and multivariate Cox regression analysis revealed that FN1 could be an independent indicator for prognosis of HNSCC. GO enrichment and KEGG pathway analysis demonstrated that cell adhesion, focal adhesion, and the PI3K-Akt signaling pathway might be involved in the potential mechanisms of FN1's prognostic performance in HNSCC. Methylation of FN1 was also higher and closely associated with poorer survival in HNSCC. In addition, FN1 expression was positively correlated with three DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B). Furthermore, FN1 was positively associated with CD4+ T cells, endothelial cells, macrophages, and NK cells and negatively correlated with CD8+ T cells Conclusion: FN1 might be an independent prognostic biomarker for HNSCC patients. Hypermethylation, the aberrant proportions of immune cells, and the PI3K/Akt signaling pathway might be involved in the mechanism of FN1's oncogene role in HNSCC.
Collapse
Affiliation(s)
- Surui Sheng
- Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Guo
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhentao Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihua Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jieyu Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zirong Huo
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Liberini V, Mariniello A, Righi L, Capozza M, Delcuratolo MD, Terreno E, Farsad M, Volante M, Novello S, Deandreis D. NSCLC Biomarkers to Predict Response to Immunotherapy with Checkpoint Inhibitors (ICI): From the Cells to In Vivo Images. Cancers (Basel) 2021; 13:4543. [PMID: 34572771 PMCID: PMC8464855 DOI: 10.3390/cancers13184543] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death, and it is usually diagnosed in advanced stages (stage III or IV). Recently, the availability of targeted strategies and of immunotherapy with checkpoint inhibitors (ICI) has favorably changed patient prognosis. Treatment outcome is closely related to tumor biology and interaction with the tumor immune microenvironment (TME). While the response in molecular targeted therapies relies on the presence of specific genetic alterations in tumor cells, accurate ICI biomarkers of response are lacking, and clinical outcome likely depends on multiple factors that are both host and tumor-related. This paper is an overview of the ongoing research on predictive factors both from in vitro/ex vivo analysis (ranging from conventional pathology to molecular biology) and in vivo analysis, where molecular imaging is showing an exponential growth and use due to technological advancements and to the new bioinformatics approaches applied to image analyses that allow the recovery of specific features in specific tumor subclones.
Collapse
Affiliation(s)
- Virginia Liberini
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Turin, Italy;
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy
| | - Annapaola Mariniello
- Thoracic Oncology Unit, Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy; (A.M.); (M.D.D.); (S.N.)
| | - Luisella Righi
- Pathology Unit, Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy; (L.R.); (M.V.)
| | - Martina Capozza
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Marco Donatello Delcuratolo
- Thoracic Oncology Unit, Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy; (A.M.); (M.D.D.); (S.N.)
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Mohsen Farsad
- Nuclear Medicine, Central Hospital Bolzano, 39100 Bolzano, Italy;
| | - Marco Volante
- Pathology Unit, Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy; (L.R.); (M.V.)
| | - Silvia Novello
- Thoracic Oncology Unit, Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy; (A.M.); (M.D.D.); (S.N.)
| | - Désirée Deandreis
- Department of Medical Science, Division of Nuclear Medicine, University of Turin, 10126 Turin, Italy;
| |
Collapse
|
38
|
Yin G, Wang Z, Song Y, Li X, Chen Y, Zhu L, Su Q, Dai D, Xu W. Prediction of EGFR Mutation Status Based on 18F-FDG PET/CT Imaging Using Deep Learning-Based Model in Lung Adenocarcinoma. Front Oncol 2021; 11:709137. [PMID: 34367993 PMCID: PMC8340023 DOI: 10.3389/fonc.2021.709137] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Objective The purpose of this study was to develop a deep learning-based system to automatically predict epidermal growth factor receptor (EGFR) mutant lung adenocarcinoma in 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT). Methods Three hundred and one lung adenocarcinoma patients with EGFR mutation status were enrolled in this study. Two deep learning models (SECT and SEPET) were developed with Squeeze-and-Excitation Residual Network (SE-ResNet) module for the prediction of EGFR mutation with CT and PET images, respectively. The deep learning models were trained with a training data set of 198 patients and tested with a testing data set of 103 patients. Stacked generalization was used to integrate the results of SECT and SEPET. Results The AUCs of the SECT and SEPET were 0.72 (95% CI, 0.62–0.80) and 0.74 (95% CI, 0.65–0.82) in the testing data set, respectively. After integrating SECT and SEPET with stacked generalization, the AUC was further improved to 0.84 (95% CI, 0.75–0.90), significantly higher than SECT (p<0.05). Conclusion The stacking model based on 18F-FDG PET/CT images is capable to predict EGFR mutation status of patients with lung adenocarcinoma automatically and non-invasively. The proposed model in this study showed the potential to help clinicians identify suitable advanced patients with lung adenocarcinoma for EGFR‐targeted therapy.
Collapse
Affiliation(s)
- Guotao Yin
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Yingchao Song
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Yiwen Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Lei Zhu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Qian Su
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| |
Collapse
|
39
|
Primary metabolic tumor volume from 18F-FDG PET/CT associated with epidermal growth factor receptor mutation in lung adenocarcinoma patients. Nucl Med Commun 2021; 41:1210-1217. [PMID: 32815896 DOI: 10.1097/mnm.0000000000001274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE To explore the potential parameters from F-FDG PET/CT that might be associated with the epidermal growth factor receptor (EGFR) gene mutation status in lung adenocarcinoma (ADC) patients. METHODS Data of the test cohort of 191 patients and the validation cohort of 55 patients with newly diagnosed ADC were retrospectively reviewed. All patients underwent F-FDG PET/CT scans and EGFR mutation tests prior to treatment. The metabolic parameters obtained from F-FDG PET/CT combining with clinical characteristics were analyzed by using univariate and multivariate logistic regression analyses. Then two cohorts were enrolled to validate the predictive model by area under the receiver-operating characteristic curve (AUC), respectively. RESULTS EGFR mutation-positive was seen of 33.0% (63/191) and 32.7% (18/55) in two cohorts, respectively. In univariate analysis, female, nonsmokers, metabolic parameters of primary tumor [mean standardized uptake value, metabolic tumor volume (pMTV), and total lesion glycolysis], non-necrosis of primary tumor, and serum tumor markers [carbohydrate antigen 19-9, squamous cell carcinoma antigen, and precursor of gastrin releasing peptide (proGRP)] were significantly relevant with EGFR mutation. In multivariate analysis with adjustment of age and TNM stage, pMTV (<8.13 cm), proGRP (≥38.44 pg/ml) and women were independent significant predictors for EGFR mutation. The AUC for the predictive value of these factors was 0.739 [95% confidence interval (CI) 0.665-0.813] in the cohort of 191 patients and 0.716 (95% CI 0.567-0.865) in the cohort of 55 patients, respectively. CONCLUSION Low pMTV (<8.13 cm) was an independent predictor and could be integrated with women and high proGRP (≥38.44 pg/ml) to enhance the discriminability on the EGFR mutation status in ADC patients.
Collapse
|
40
|
Smith BJ, Buatti JM, Bauer C, Ulrich EJ, Ahmadvand P, Budzevich MM, Gillies RJ, Goldgof D, Grkovski M, Hamarneh G, Kinahan PE, Muzi JP, Muzi M, Laymon CM, Mountz JM, Nehmeh S, Oborski MJ, Zhao B, Sunderland JJ, Beichel RR. Multisite Technical and Clinical Performance Evaluation of Quantitative Imaging Biomarkers from 3D FDG PET Segmentations of Head and Neck Cancer Images. ACTA ACUST UNITED AC 2021; 6:65-76. [PMID: 32548282 PMCID: PMC7289247 DOI: 10.18383/j.tom.2020.00004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Quantitative imaging biomarkers (QIBs) provide medical image-derived intensity, texture, shape, and size features that may help characterize cancerous tumors and predict clinical outcomes. Successful clinical translation of QIBs depends on the robustness of their measurements. Biomarkers derived from positron emission tomography images are prone to measurement errors owing to differences in image processing factors such as the tumor segmentation method used to define volumes of interest over which to calculate QIBs. We illustrate a new Bayesian statistical approach to characterize the robustness of QIBs to different processing factors. Study data consist of 22 QIBs measured on 47 head and neck tumors in 10 positron emission tomography/computed tomography scans segmented manually and with semiautomated methods used by 7 institutional members of the NCI Quantitative Imaging Network. QIB performance is estimated and compared across institutions with respect to measurement errors and power to recover statistical associations with clinical outcomes. Analysis findings summarize the performance impact of different segmentation methods used by Quantitative Imaging Network members. Robustness of some advanced biomarkers was found to be similar to conventional markers, such as maximum standardized uptake value. Such similarities support current pursuits to better characterize disease and predict outcomes by developing QIBs that use more imaging information and are robust to different processing factors. Nevertheless, to ensure reproducibility of QIB measurements and measures of association with clinical outcomes, errors owing to segmentation methods need to be reduced.
Collapse
Affiliation(s)
| | | | | | - Ethan J Ulrich
- Electrical and Computer Engineering.,Biomedical Engineering, The University of Iowa, Iowa City, IA
| | - Payam Ahmadvand
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Mikalai M Budzevich
- H. Lee Moffitt Cancer Center & Research Institute, Department of Cancer Physiology, FL
| | - Robert J Gillies
- H. Lee Moffitt Cancer Center & Research Institute, Department of Cancer Physiology, FL
| | - Dmitry Goldgof
- Department of Computer Science and Engineering, University of South Florida, FL
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Paul E Kinahan
- Department of Radiology, The University of Washington Medical Center, Seattle, WA
| | - John P Muzi
- Department of Radiology, The University of Washington Medical Center, Seattle, WA
| | - Mark Muzi
- Department of Radiology, The University of Washington Medical Center, Seattle, WA
| | - Charles M Laymon
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA.,Department of Radiology, University of Pittsburgh, Pittsburgh, PA
| | - James M Mountz
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA
| | - Sadek Nehmeh
- Department of Radiology, Weill Cornell Medical College, NY; and
| | - Matthew J Oborski
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center, New York, NY
| | | | | |
Collapse
|
41
|
La Greca Saint-Esteven A, Vuong D, Tschanz F, van Timmeren JE, Dal Bello R, Waller V, Pruschy M, Guckenberger M, Tanadini-Lang S. Systematic Review on the Association of Radiomics with Tumor Biological Endpoints. Cancers (Basel) 2021; 13:cancers13123015. [PMID: 34208595 PMCID: PMC8234501 DOI: 10.3390/cancers13123015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Radiomics supposes an alternative non-invasive tumor characterization tool, which has experienced increased interest with the advent of more powerful computers and more sophisticated machine learning algorithms. Nonetheless, the incorporation of radiomics in cancer clinical-decision support systems still necessitates a thorough analysis of its relationship with tumor biology. Herein, we present a systematic review focusing on the clinical evidence of radiomics as a surrogate method for tumor molecular profile characterization. An extensive literature review was conducted in PubMed, including papers on radiomics and a selected set of clinically relevant and commonly used tumor molecular markers. We summarized our findings based on different cancer entities, additionally evaluating the effect of different modalities for the prediction of biomarkers at each tumor site. Results suggest the existence of an association between the studied biomarkers and radiomics from different modalities and different tumor sites, even though a larger number of multi-center studies are required to further validate the reported outcomes.
Collapse
Affiliation(s)
- Agustina La Greca Saint-Esteven
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
- Correspondence:
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Fabienne Tschanz
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Janita E. van Timmeren
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Riccardo Dal Bello
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Verena Waller
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Martin Pruschy
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| |
Collapse
|
42
|
Dong Y, Hou L, Yang W, Han J, Wang J, Qiang Y, Zhao J, Hou J, Song K, Ma Y, Kazihise NGF, Cui Y, Yang X. Multi-channel multi-task deep learning for predicting EGFR and KRAS mutations of non-small cell lung cancer on CT images. Quant Imaging Med Surg 2021; 11:2354-2375. [PMID: 34079707 PMCID: PMC8107307 DOI: 10.21037/qims-20-600] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Predicting the mutation statuses of 2 essential pathogenic genes [epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma (KRAS)] in non-small cell lung cancer (NSCLC) based on CT is valuable for targeted therapy because it is a non-invasive and less costly method. Although deep learning technology has realized substantial computer vision achievements, CT imaging being used to predict gene mutations remains challenging due to small dataset limitations. METHODS We propose a multi-channel and multi-task deep learning (MMDL) model for the simultaneous prediction of EGFR and KRAS mutation statuses based on CT images. First, we decomposed each 3D lung nodule into 9 views. Then, we used the pre-trained inception-attention-resnet model for each view to learn the features of the nodules. By combining 9 inception-attention-resnet models to predict the types of gene mutations in lung nodules, the models were adaptively weighted, and the proposed MMDL model could be trained end-to-end. The MMDL model utilized multiple channels to characterize the nodule more comprehensively and integrate patient personal information into our learning process. RESULTS We trained the proposed MMDL model using a dataset of 363 patients collected by our partner hospital and conducted a multi-center validation on 162 patients in The Cancer Imaging Archive (TCIA) public dataset. The accuracies for the prediction of EGFR and KRAS mutations were, respectively, 79.43% and 72.25% in the training dataset and 75.06% and 69.64% in the validation dataset. CONCLUSIONS The experimental results demonstrated that the proposed MMDL model outperformed the latest methods in predicting EGFR and KRAS mutations in NSCLC.
Collapse
Affiliation(s)
- Yunyun Dong
- School of Software, Taiyuan University of Technology, Taiyuan, China
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Lina Hou
- Department of Radiology, Shanxi Province Cancer Hospital, Taiyuan, China
| | - Wenkai Yang
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Jiahao Han
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Jiawen Wang
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Yan Qiang
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Juanjuan Zhao
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Jiaxin Hou
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Kai Song
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Yulan Ma
- School of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | | | - Yanfen Cui
- Department of Radiology, Shanxi Province Cancer Hospital, Taiyuan, China
| | - Xiaotang Yang
- Department of Radiology, Shanxi Province Cancer Hospital, Taiyuan, China
| |
Collapse
|
43
|
Guo Y, Zhu H, Yao Z, Liu F, Yang D. The diagnostic and predictive efficacy of 18F-FDG PET/CT metabolic parameters for EGFR mutation status in non-small-cell lung cancer: A meta-analysis. Eur J Radiol 2021; 141:109792. [PMID: 34062472 DOI: 10.1016/j.ejrad.2021.109792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the predictive performance of the maximum standardized uptake value (SUVmax) and mean standardized uptake value (SUVmean) of primary lesions based on 18 F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) for EGFR mutation status in patients with non-small cell lung cancer (NSCLC). METHODS The PubMed/Medline, Embase, Cochrane Library and Web of Science databases were searched as of January 1, 2021. Studies whose reported data could be used to construct contingency tables were included. Study characteristics were extracted, and methodological quality assessment was conducted by two separate reviewers using the Quality Assessment of Diagnostic Accuracy Studies. The pooled sensitivity, specificity and area under the summary receiver operating characteristic curve (AUROC) were calculated. The possible causes of heterogeneity were analysed by meta-regression. RESULTS The 18 included studies had a total of 4024 patients. The majority of the studies showed a low to unclear risk of bias and concerns of applicability. For differentiating EGFR-mutant NSCLC from wild-type NSCLC, the pooled sensitivity and specificity were 71 % and 60 % for SUVmax and 64 % and 63 % for SUVmean, respectively. The summary AUROCs of SUVmax and SUVmean were 0.69 (95 % CI, 0.65-0.73) and 0.68 (95 % CI, 0.64-0.72), respectively. The meta-regression analysis indicated that blindness to EGFR mutation test results, the number of readers and the number of PET/CT scanners were possible causes of heterogeneity. CONCLUSIONS Our meta-analysis implied that SUVmax and SUVmean of primary lesions from 18F-FDG PET/CT harboured moderate predictive efficacy for the EGFR mutation status of NSCLC.
Collapse
Affiliation(s)
- Yue Guo
- Department of Nuclear Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, PR China.
| | - Hui Zhu
- Department of Nuclear Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, PR China.
| | - Zhiming Yao
- Department of Nuclear Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, PR China.
| | - Fugeng Liu
- Department of Nuclear Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, PR China.
| | - Dawei Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No.95 Yongan Road, West District, Beijing 100050, PR China.
| |
Collapse
|
44
|
Bevilacqua A, Calabrò D, Malavasi S, Ricci C, Casadei R, Campana D, Baiocco S, Fanti S, Ambrosini V. A [68Ga]Ga-DOTANOC PET/CT Radiomic Model for Non-Invasive Prediction of Tumour Grade in Pancreatic Neuroendocrine Tumours. Diagnostics (Basel) 2021; 11:diagnostics11050870. [PMID: 34065981 PMCID: PMC8150289 DOI: 10.3390/diagnostics11050870] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
Predicting grade 1 (G1) and 2 (G2) primary pancreatic neuroendocrine tumour (panNET) is crucial to foresee panNET clinical behaviour. Fifty-one patients with G1-G2 primary panNET demonstrated by pre-surgical [68Ga]Ga-DOTANOC PET/CT and diagnostic conventional imaging were grouped according to the tumour grade assessment method: histology on the whole excised primary lesion (HS) or biopsy (BS). First-order and second-order radiomic features (RFs) were computed from SUV maps for the whole tumour volume on HS. The RFs showing the lowest p-values and the highest area under the curve (AUC) were selected. Three radiomic models were assessed: A (trained on HS, validated on BS), B (trained on BS, validated on HS), and C (using the cross-validation on the whole dataset). The second-order normalized homogeneity and entropy was the most effective RFs couple predicting G2 and G1. The best performance was achieved by model A (test AUC = 0.90, sensitivity = 0.88, specificity = 0.89), followed by model C (median test AUC = 0.87, sensitivity = 0.83, specificity = 0.82). Model B performed worse. Using HS to train a radiomic model leads to the best prediction, although a “hybrid” (HS+BS) population performs better than biopsy-only. The non-invasive prediction of panNET grading may be especially useful in lesions not amenable to biopsy while [68Ga]Ga-DOTANOC heterogeneity might recommend FDG PET/CT.
Collapse
Affiliation(s)
- Alessandro Bevilacqua
- Advanced Research Center for Electronic Systems (ARCES), University of Bologna, I-40125 Bologna, Italy; (S.M.); (S.B.)
- Department of Computer Science and Engineering (DISI), University of Bologna, I-40136 Bologna, Italy
- Correspondence: ; Tel.: +39-051-209-5409
| | - Diletta Calabrò
- Department of Nuclear Medicine, DIMES, Alma Mater Studiorum, University of Bologna, I-40126 Bologna, Italy; (D.C.); (S.F.)
| | - Silvia Malavasi
- Advanced Research Center for Electronic Systems (ARCES), University of Bologna, I-40125 Bologna, Italy; (S.M.); (S.B.)
- Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, I-40126 Bologna, Italy
| | - Claudio Ricci
- IRCCS Azienda Ospedaliero Universitaria di Bologna, I-40138 Bologna, Italy; (C.R.); (R.C.); (D.C.)
- Department of Surgery, DIMEC Alma Mater Studiorum, University of Bologna, S.Orsola-Malpighi Hospital, I-40138 Bologna, Italy
- NET Team Bologna, ENETS Center of Excellence, I-40138 Bologna, Italy
| | - Riccardo Casadei
- IRCCS Azienda Ospedaliero Universitaria di Bologna, I-40138 Bologna, Italy; (C.R.); (R.C.); (D.C.)
- Department of Surgery, DIMEC Alma Mater Studiorum, University of Bologna, S.Orsola-Malpighi Hospital, I-40138 Bologna, Italy
- NET Team Bologna, ENETS Center of Excellence, I-40138 Bologna, Italy
| | - Davide Campana
- IRCCS Azienda Ospedaliero Universitaria di Bologna, I-40138 Bologna, Italy; (C.R.); (R.C.); (D.C.)
- NET Team Bologna, ENETS Center of Excellence, I-40138 Bologna, Italy
- Department of Oncology, DIMES Alma Mater Studiorum, University of Bologna, S.Orsola-Malpighi Hospital, I-40126 Bologna, Italy
| | - Serena Baiocco
- Advanced Research Center for Electronic Systems (ARCES), University of Bologna, I-40125 Bologna, Italy; (S.M.); (S.B.)
| | - Stefano Fanti
- Department of Nuclear Medicine, DIMES, Alma Mater Studiorum, University of Bologna, I-40126 Bologna, Italy; (D.C.); (S.F.)
- IRCCS Azienda Ospedaliero Universitaria di Bologna, I-40138 Bologna, Italy; (C.R.); (R.C.); (D.C.)
- NET Team Bologna, ENETS Center of Excellence, I-40138 Bologna, Italy
| | - Valentina Ambrosini
- Department of Nuclear Medicine, DIMES, Alma Mater Studiorum, University of Bologna, I-40126 Bologna, Italy; (D.C.); (S.F.)
- IRCCS Azienda Ospedaliero Universitaria di Bologna, I-40138 Bologna, Italy; (C.R.); (R.C.); (D.C.)
- NET Team Bologna, ENETS Center of Excellence, I-40138 Bologna, Italy
| |
Collapse
|
45
|
He J, Wang Q, Zhang Y, Wu H, Zhou Y, Zhao S. Preoperative prediction of regional lymph node metastasis of colorectal cancer based on 18F-FDG PET/CT and machine learning. Ann Nucl Med 2021; 35:617-627. [PMID: 33738763 DOI: 10.1007/s12149-021-01605-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/10/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE To establish and validate a regional lymph node (LN) metastasis prediction model of colorectal cancer (CRC) based on 18F-FDG PET/CT and radiomic features using machine-learning methods. METHODS A total of 199 colorectal cancer patients underwent pre-therapy diagnostic 18F-FDG PET/CT scans and CRC radical surgery. The Chang-Gung Image Texture Analysis toolbox (CGITA) was used to extract 70 PET radiomic features reflecting 18F-FDG uptake heterogeneity of tumors. The least absolute shrinkage and selection operator (LASSO) algorithm was used to select radiomic features and develop a radiomic signature score (Rad-score). The training set was used to establish five machine-learning prediction models and the test set was used to test the efficacy of the models. The effectiveness of the models was compared by ROC analysis. RESULTS The CRC patients were divided into a training set (n = 144) and a test set (n = 55). Two radiomic features were selected to build the Rad-score. Five machine-learning algorithms including logistic regression, support vector machine (SVM), random forest, neural network and eXtreme gradient boosting (XGBoost) were used to established models. Among the five machine-learning models, logistic regression (AUC 0.866, 95% CI 0.808-0.925) and XGBoost (AUC 0.903, 95% CI 0.855-0.951) models performed the best. In the training set, the AUC of these two models were significantly higher than that of the LN metastasis status reported by 18F-FDG PET/CT for differentiating positive and negative regional LN metastases in CRC (all p < 0.05). Good efficacy of the above two models was also achieved in the test set. We created a nomogram based on the logistic regression model that visualized the results and provided an easy-to-use method for predicting regional LN metastasis in patients with CRC. CONCLUSION In this study, five machine-learning models for preoperative prediction of regional LN metastasis of CRC based on 18F-FDG PET/CT and PET-based radiomic features were successfully developed and validated. Among them, the logistic regression and XGBoost models performed the best, with higher efficacy than 18F-FDG PET/CT in both the training and test sets.
Collapse
Affiliation(s)
- Jiahong He
- Department of Radiology, The Second Affiliated Hospital of Shenzhen University, The People's Hospital of Baoan Shenzhen, Shenzhen, 518100, Guangdong, China.
| | - Quanshi Wang
- PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yin Zhang
- PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hubing Wu
- PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yongsheng Zhou
- Department of Radiology, The Second Affiliated Hospital of Shenzhen University, The People's Hospital of Baoan Shenzhen, Shenzhen, 518100, Guangdong, China
| | - Shuangquan Zhao
- Department of Radiology, The Second Affiliated Hospital of Shenzhen University, The People's Hospital of Baoan Shenzhen, Shenzhen, 518100, Guangdong, China
| |
Collapse
|
46
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
47
|
Abstract
Positron emission tomography (PET)/computed tomography (CT) are nuclear diagnostic imaging modalities that are routinely deployed for cancer staging and monitoring. They hold the advantage of detecting disease related biochemical and physiologic abnormalities in advance of anatomical changes, thus widely used for staging of disease progression, identification of the treatment gross tumor volume, monitoring of disease, as well as prediction of outcomes and personalization of treatment regimens. Among the arsenal of different functional imaging modalities, nuclear imaging has benefited from early adoption of quantitative image analysis starting from simple standard uptake value normalization to more advanced extraction of complex imaging uptake patterns; thanks to application of sophisticated image processing and machine learning algorithms. In this review, we discuss the application of image processing and machine/deep learning techniques to PET/CT imaging with special focus on the oncological radiotherapy domain as a case study and draw examples from our work and others to highlight current status and future potentials.
Collapse
Affiliation(s)
- Lise Wei
- Department of Radiation Oncology, Physics Division, University of Michigan, Ann Arbor, MI
| | - Issam El Naqa
- Department of Radiation Oncology, Physics Division, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
48
|
Tomaszewski MR, Gillies RJ. The Biological Meaning of Radiomic Features. Radiology 2021; 298:505-516. [PMID: 33399513 PMCID: PMC7924519 DOI: 10.1148/radiol.2021202553] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
An earlier incorrect version appeared online. This article was corrected on February 10, 2021.
Collapse
Affiliation(s)
- Michal R. Tomaszewski
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| | - Robert J. Gillies
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| |
Collapse
|
49
|
A Systematic Review of PET Textural Analysis and Radiomics in Cancer. Diagnostics (Basel) 2021; 11:diagnostics11020380. [PMID: 33672285 PMCID: PMC7926413 DOI: 10.3390/diagnostics11020380] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although many works have supported the utility of PET radiomics, several authors have raised concerns over the robustness and replicability of the results. This study aimed to perform a systematic review on the topic of PET radiomics and the used methodologies. Methods: PubMed was searched up to 15 October 2020. Original research articles based on human data specifying at least one tumor type and PET image were included, excluding those that apply only first-order statistics and those including fewer than 20 patients. Each publication, cancer type, objective and several methodological parameters (number of patients and features, validation approach, among other things) were extracted. Results: A total of 290 studies were included. Lung (28%) and head and neck (24%) were the most studied cancers. The most common objective was prognosis/treatment response (46%), followed by diagnosis/staging (21%), tumor characterization (18%) and technical evaluations (15%). The average number of patients included was 114 (median = 71; range 20–1419), and the average number of high-order features calculated per study was 31 (median = 26, range 1–286). Conclusions: PET radiomics is a promising field, but the number of patients in most publications is insufficient, and very few papers perform in-depth validations. The role of standardization initiatives will be crucial in the upcoming years.
Collapse
|
50
|
Yang Y, Yang J, Shen L, Chen J, Xia L, Ni B, Ge L, Wang Y, Lu S. A multi-omics-based serial deep learning approach to predict clinical outcomes of single-agent anti-PD-1/PD-L1 immunotherapy in advanced stage non-small-cell lung cancer. Am J Transl Res 2021; 13:743-756. [PMID: 33594323 PMCID: PMC7868825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023]
Abstract
Only 20% NSCLC patients benefit from immunotherapy with a durable response. Current biomarkers are limited by the availability of samples and do not accurately predict who will benefit from immunotherapy. To develop a unified deep learning model to integrate multimodal serial information from CT with laboratory and baseline clinical information. We retrospectively analyzed 1633 CT scans and 3414 blood samples from 200 advanced stage NSCLC patients who received single anti-PD-1/PD-L1 agent between April 2016 and December 2019. Multidimensional information, including serial radiomics, laboratory data and baseline clinical data, was used to develop and validate deep learning models to identify immunotherapy responders and nonresponders. A Simple Temporal Attention (SimTA) module was developed to process asynchronous time-series imaging and laboratory data. Using cross-validation, the 90-day deep learning-based predicting model showed a good performance in distinguishing responders from nonresponders, with an area under the curve (AUC) of 0.80 (95% CI: 0.74-0.86). Before immunotherapy, we stratified the patients into high- and low-risk nonresponders using the model. The low-risk group had significantly longer progression-free survival (PFS) (8.4 months, 95% CI: 5.49-11.31 vs. 1.5 months, 95% CI: 1.29-1.71; HR 3.14, 95% CI: 2.27-4.33; log-rank test, P<0.01) and overall survival (OS) (26.7 months, 95% CI: 18.76-34.64 vs. 8.6 months, 95% CI: 4.55-12.65; HR 2.46, 95% CI: 1.73-3.51; log-rank test, P<0.01) than the high-risk group. An exploratory analysis of 93 patients with stable disease (SD) [after first efficacy assessment according to the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1] also showed that the 90-day model had a good prediction of survival and low-risk patients had significantly longer PFS (11.1 months, 95% CI: 10.24-11.96 vs. 3.3 months, 95% CI: 0.34-6.26; HR 2.93, 95% CI: 1.69-5.10; log-rank test, P<0.01) and OS (31.7 months, 95% CI: 23.64-39.76 vs. 17.2 months, 95% CI: 7.22-27.18; HR 2.22, 95% CI: 1.17-4.20; log-rank test, P=0.01) than high-risk patients. In conclusion, the SimTA-based multi-omics serial deep learning provides a promising methodology for predicting response of advanced NSCLC patients to anti-PD-1/PD-L1 monotherapy. Moreover, our model could better differentiate survival benefit among SD patients than the traditional RECIST evaluation method.
Collapse
Affiliation(s)
- Yi Yang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Jiancheng Yang
- Department of Electronic Engineering, Shanghai Jiao Tong UniversityShanghai, China
- MoE Key Lab of Artificial Intelligence, Artificial Intelligence Institute, Shanghai Jiao Tong UniversityShanghai, China
- Dianei TechnologyShanghai, China
| | - Lan Shen
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | | | - Liliang Xia
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Bingbing Ni
- Department of Electronic Engineering, Shanghai Jiao Tong UniversityShanghai, China
- MoE Key Lab of Artificial Intelligence, Artificial Intelligence Institute, Shanghai Jiao Tong UniversityShanghai, China
| | - Liang Ge
- Dianei TechnologyShanghai, China
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|