1
|
Alizade-Harakiyan M, Khodaei A, Yousefi A, Zamani H, Mesbahi A. Decision tree-based machine learning algorithm for prediction of acute radiation esophagitis. Biochem Biophys Rep 2025; 42:101991. [PMID: 40230494 PMCID: PMC11995095 DOI: 10.1016/j.bbrep.2025.101991] [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: 01/30/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Background Radiation-induced esophagitis remains a significant challenge in thoracic and neck cancer treatment, impacting patient quality of life and potentially limiting therapeutic efficacy. This study aimed to develop and validate a decision tree-based model for predicting acute esophagitis grades in patients undergoing chemoradiotherapy. Methods Data from 100 patients receiving thoracic and neck radiotherapy were analyzed. The dataset comprised 33 features, including demographic, clinical, and dosimetric parameters. A decision tree classifier was implemented for both binary (Grade ≥2 vs. <2) and multi-class (Grades 1, 2, and 3) classification. Model performance was evaluated using standard metrics including accuracy, precision, recall, and F1-score. Results The binary classification model achieved 97 % accuracy in distinguishing acute esophagitis. The multi-class model demonstrated 98 % accuracy in predicting specific grades. Key predictive features included V40 (volume receiving 40 Gy), V60, and average esophageal dose. The model generated interpretable decision rules, with V60 ≥ 2.3 strongly indicating Grade 3 esophagitis. Conclusions The decision tree model demonstrates high accuracy in predicting radiation-induced esophagitis grades while maintaining clinical interpretability. This approach offers potential for treatment optimization and personalized risk assessment in radiotherapy planning. The model's transparency and reliability make it a promising tool for clinical decision support in radiation oncology.
Collapse
Affiliation(s)
- Mostafa Alizade-Harakiyan
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Khodaei
- Faculty of Electrical and Computer Engineering, University of Tabriz, Tabriz, Iran
| | - Ali Yousefi
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Zamani
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Mesbahi
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Park J, Hu W, Jin IH, Liu H, Zang Y. A Bayesian adaptive biomarker stratified phase II randomized clinical trial design for radiotherapies with competing risk survival outcomes. Stat Methods Med Res 2024; 33:80-95. [PMID: 38062757 PMCID: PMC11227940 DOI: 10.1177/09622802231215801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
In recent decades, many phase II clinical trials have used survival outcomes as the primary endpoints. If radiotherapy is involved, the competing risk issue often arises because the time to disease progression can be censored by the time to normal tissue complications, and vice versa. Besides, many existing research has examined that patients receiving the same radiotherapy dose may yield distinct responses due to their heterogeneous radiation susceptibility statuses. Therefore, the "one-size-fits-all" strategy often fails, and it is more relevant to evaluate the subgroup-specific treatment effect with the subgroup defined by the radiation susceptibility status. In this paper, we propose a Bayesian adaptive biomarker stratified phase II trial design evaluating the subgroup-specific treatment effects of radiotherapy. We use the cause-specific hazard approach to model the competing risk survival outcomes. We propose restricting the candidate radiation doses based on each patient's radiation susceptibility status. Only the clinically feasible personalized dose will be considered, which enhances the benefit for the patients in the trial. In addition, we propose a stratified Bayesian adaptive randomization scheme such that more patients will be randomized to the dose reporting more favorable survival outcomes. Numerical studies and an illustrative trial example have shown that the proposed design performed well and outperformed the conventional design ignoring the competing risk issue.
Collapse
Affiliation(s)
- Jina Park
- Department of Applied Statistics, Yonsei University, South Korea
- Department of Statistics and Data Science, Yonsei University, South Korea
| | | | - Ick Hoon Jin
- Department of Applied Statistics, Yonsei University, South Korea
- Department of Statistics and Data Science, Yonsei University, South Korea
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Cancer Institute of New Jersey, Rutgers University, USA
| | - Yong Zang
- Department of Biostatistics and Health Data Sciences, Center of Computational Biology and Bioinformatics, Indiana University, USA
| |
Collapse
|
3
|
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
|
4
|
Forouzannezhad P, Maes D, Hippe DS, Thammasorn P, Iranzad R, Han J, Duan C, Liu X, Wang S, Chaovalitwongse WA, Zeng J, Bowen SR. Multitask Learning Radiomics on Longitudinal Imaging to Predict Survival Outcomes following Risk-Adaptive Chemoradiation for Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:1228. [PMID: 35267535 PMCID: PMC8909466 DOI: 10.3390/cancers14051228] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
Medical imaging provides quantitative and spatial information to evaluate treatment response in the management of patients with non-small cell lung cancer (NSCLC). High throughput extraction of radiomic features on these images can potentially phenotype tumors non-invasively and support risk stratification based on survival outcome prediction. The prognostic value of radiomics from different imaging modalities and time points prior to and during chemoradiation therapy of NSCLC, relative to conventional imaging biomarker or delta radiomics models, remains uncharacterized. We investigated the utility of multitask learning of multi-time point radiomic features, as opposed to single-task learning, for improving survival outcome prediction relative to conventional clinical imaging feature model benchmarks. Survival outcomes were prospectively collected for 45 patients with unresectable NSCLC enrolled on the FLARE-RT phase II trial of risk-adaptive chemoradiation and optional consolidation PD-L1 checkpoint blockade (NCT02773238). FDG-PET, CT, and perfusion SPECT imaging pretreatment and week 3 mid-treatment was performed and 110 IBSI-compliant pyradiomics shape-/intensity-/texture-based features from the metabolic tumor volume were extracted. Outcome modeling consisted of a fused Laplacian sparse group LASSO with component-wise gradient boosting survival regression in a multitask learning framework. Testing performance under stratified 10-fold cross-validation was evaluated for multitask learning radiomics of different imaging modalities and time points. Multitask learning models were benchmarked against conventional clinical imaging and delta radiomics models and evaluated with the concordance index (c-index) and index of prediction accuracy (IPA). FDG-PET radiomics had higher prognostic value for overall survival in test folds (c-index 0.71 [0.67, 0.75]) than CT radiomics (c-index 0.64 [0.60, 0.71]) or perfusion SPECT radiomics (c-index 0.60 [0.57, 0.63]). Multitask learning of pre-/mid-treatment FDG-PET radiomics (c-index 0.71 [0.67, 0.75]) outperformed benchmark clinical imaging (c-index 0.65 [0.59, 0.71]) and FDG-PET delta radiomics (c-index 0.52 [0.48, 0.58]) models. Similarly, the IPA for multitask learning FDG-PET radiomics (30%) was higher than clinical imaging (26%) and delta radiomics (15%) models. Radiomics models performed consistently under different voxel resampling conditions. Multitask learning radiomics for outcome modeling provides a clinical decision support platform that leverages longitudinal imaging information. This framework can reveal the relative importance of different imaging modalities and time points when designing risk-adaptive cancer treatment strategies.
Collapse
Affiliation(s)
- Parisa Forouzannezhad
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Dominic Maes
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
| | - Phawis Thammasorn
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Reza Iranzad
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jie Han
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - Chunyan Duan
- Department of Mechanical Engineering, Tongji University, Shanghai 200092, China;
| | - Xiao Liu
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Shouyi Wang
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - W. Art Chaovalitwongse
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jing Zeng
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Stephen R. Bowen
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
- Department of Radiology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Wang Y, Lin X, Sun D. A narrative review of prognosis prediction models for non-small cell lung cancer: what kind of predictors should be selected and how to improve models? ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1597. [PMID: 34790803 PMCID: PMC8576716 DOI: 10.21037/atm-21-4733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/02/2021] [Indexed: 12/18/2022]
Abstract
Objective To discover potential predictors and explore how to build better models by summarizing the existing prognostic prediction models of non-small cell lung cancer (NSCLC). Background Research on clinical prediction models of NSCLC has experienced explosive growth in recent years. As more predictors of prognosis are discovered, the choice of predictors to build models is particularly important, and in the background of more applications of next-generation sequencing technology, gene-related predictors are widely used. As it is more convenient to obtain samples and follow-up data, the prognostic model is preferred by researchers. Methods PubMed and the Cochrane Library were searched using the items “NSCLC”, “prognostic model”, “prognosis prediction”, and “survival prediction” from 1 January 1980 to 5 May 2021. Reference lists from articles were reviewed and relevant articles were identified. Conclusions The performance of gene-related models has not obviously improved. Relative to the innovation and diversity of predictors, it is more important to establish a highly stable model that is convenient for clinical application. Most of the prevalent models are highly biased and referring to PROBAST at the beginning of the study may be able to significantly control the bias. Existing models should be validated in a large external dataset to make a meaningful comparison.
Collapse
Affiliation(s)
- Yuhang Wang
- Graduate School, Tianjin Medical University, Tianjin, China
| | | | - Daqiang Sun
- Graduate School, Tianjin Medical University, Tianjin, China.,Department of Thoracic Surgery, Tianjin Chest Hospital of Nankai University, Tianjin, China
| |
Collapse
|
6
|
Oliveira C, Amstutz F, Vuong D, Bogowicz M, Hüllner M, Foerster R, Basler L, Schröder C, Eboulet EI, Pless M, Thierstein S, Peters S, Hillinger S, Tanadini-Lang S, Guckenberger M. Preselection of robust radiomic features does not improve outcome modelling in non-small cell lung cancer based on clinical routine FDG-PET imaging. EJNMMI Res 2021; 11:79. [PMID: 34417899 PMCID: PMC8380219 DOI: 10.1186/s13550-021-00809-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/08/2021] [Indexed: 12/25/2022] Open
Abstract
Background Radiomics is a promising tool for identifying imaging-based biomarkers. Radiomics-based models are often trained on single-institution datasets; however, multi-centre imaging datasets are preferred for external generalizability owing to the influence of inter-institutional scanning differences and acquisition settings. The study aim was to determine the value of preselection of robust radiomic features in routine clinical positron emission tomography (PET) images to predict clinical outcomes in locally advanced non-small cell lung cancer (NSCLC). Methods A total of 1404 primary tumour radiomic features were extracted from pre-treatment [18F]fluorodeoxyglucose (FDG)-PET scans of stage IIIA/N2 or IIIB NSCLC patients using a training cohort (n = 79; prospective Swiss multi-centre randomized phase III trial SAKK 16/00; 16 centres) and an internal validation cohort (n = 31; single centre). Robustness studies investigating delineation variation, attenuation correction and motion were performed (intraclass correlation coefficient threshold > 0.9). Two 12-/24-month event-free survival (EFS) and overall survival (OS) logistic regression models were trained using standardized imaging: (1) with robust features alone and (2) with all available features. Models were then validated using fivefold cross-validation, and validation on a separate single-centre dataset. Model performance was assessed using area under the receiver operating characteristic curve (AUC). Results Robustness studies identified 179 stable features (13%), with 25% stable features for 3D versus 4D acquisition, 31% for attenuation correction and 78% for delineation. Univariable analysis found no significant robust features predicting 12-/24-month EFS and 12-month OS (p value > 0.076). Prognostic models without robust preselection performed well for 12-month EFS in training (AUC = 0.73) and validation (AUC = 0.74). Patient stratification into two risk groups based on 12-month EFS was significant for training (p value = 0.02) and validation cohorts (p value = 0.03). Conclusions A PET-based radiomics model using a standardized, multi-centre dataset to predict EFS in locally advanced NSCLC was successfully established and validated with good performance. Prediction models with robust feature preselection were unsuccessful, indicating the need for a standardized imaging protocol. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00809-3.
Collapse
Affiliation(s)
- Carol Oliveira
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland. .,Division of Radiation Oncology, Cancer Center of Southeastern Ontario, Queen's University, Kingston, ON, Canada.
| | - Florian Amstutz
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robert Foerster
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lucas Basler
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christina Schröder
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Eric I Eboulet
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Miklos Pless
- Department of Medical Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Sandra Thierstein
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Sven Hillinger
- Department of Thoracic Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Muenzfeld H, Nowak C, Riedlberger S, Hartenstein A, Hamm B, Jahnke P, Penzkofer T. Intra-scanner repeatability of quantitative imaging features in a 3D printed semi-anthropomorphic CT phantom. Eur J Radiol 2021; 141:109818. [PMID: 34157639 DOI: 10.1016/j.ejrad.2021.109818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Radiomics has shown to provide novel diagnostic and predictive disease information based on quantitative image features in study settings. However, limited data yielded contradictory results and important questions regarding the validity of the methods remain to be answered. The purpose of this study was to evaluate how clinical imaging techniques affect the stability of radiomics features by using 3D printed anthropomorphic CT phantom to test for repeatability and reproducibility of quantitative parameters. METHODS 48 PET/CT validated lymph nodes of prostate cancer patients (24 metastatic, 24 non-metastatic) were used as a template to create a customized 3D printed anthropomorphic phantom. We subsequently scanned the phantom five times with a routine abdominal CT protocol. Images were reconstructed using iterative reconstruction and two soft tissue kernels and one bone kernel. Radiomics features were extracted and assessed for repeatability and susceptibility towards image reconstruction settings using concordance correlation coefficients. RESULTS Our analysis revealed 19 of 86 features (22 %) as highly repeatable (CCC ≥ 0.85) with low susceptibility towards image reconstruction protocols. Most features analyzed depicted critical non-repeatability with CCC's < 0.75 even under entirely consistent imaging acquisition settings. Edge enhancing kernels result in higher variances between the scans and differences in repeatability and reproducibility were detected between PSMA-positive and negative lymph nodes with overall more stable features seen in tumor positive lymph nodes. CONCLUSIONS Both, repeatability and reproducibility play a crucial role in the validation process of radiomics features in clinical routine. This phantom study shows that most radiomics features in contrast to previous studies, including phantom and clinical, do not depict sufficient intra-scanner repeatability to serve as reliable diagnostic tools.
Collapse
Affiliation(s)
- Hanna Muenzfeld
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany.
| | - Claus Nowak
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Germany; Berlin Institute of Health, Berlin, Germany
| | | | | | - Bernd Hamm
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany
| | - Paul Jahnke
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany
| | - Tobias Penzkofer
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany; Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
8
|
Multiple Testing, Cut-Point Optimization, and Signs of Publication Bias in Prognostic FDG-PET Imaging Studies of Head and Neck and Lung Cancer: A Review and Meta-Analysis. Diagnostics (Basel) 2020; 10:diagnostics10121030. [PMID: 33271785 PMCID: PMC7761090 DOI: 10.3390/diagnostics10121030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/23/2022] Open
Abstract
Positron emission tomography (PET) imaging with 2-deoxy-2-[18F]-fluorodeoxyglucose (FDG) was proposed as prognostic marker in radiotherapy. Various uptake metrics and cut points were used, potentially leading to inflated effect estimates. Here, we performed a meta-analysis and systematic review of the prognostic value of pretreatment FDG–PET in head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC), with tests for publication bias. Hazard ratio (HR) for overall survival (OS), disease free survival (DFS), and local control was extracted or derived from the 57 studies included. Test for publication bias was performed, and the number of statistical tests and cut-point optimizations were registered. Eggers regression related to correlation of SUVmax with OS/DFS yielded p = 0.08/p = 0.02 for HNSCC and p < 0.001/p = 0.014 for NSCLC. No outcomes showed significant correlation with SUVmax, when adjusting for publication bias effect, whereas all four showed a correlation in the conventional meta-analysis. The number of statistical tests and cut points were high with no indication of improvement over time. Our analysis showed significant evidence of publication bias leading to inflated estimates of the prognostic value of SUVmax. We suggest that improved management of these complexities, including predefined statistical analysis plans, are critical for a reliable assessment of FDG–PET.
Collapse
|
9
|
de la Pinta C, Barrios-Campo N, Sevillano D. Radiomics in lung cancer for oncologists. J Clin Transl Res 2020; 6:127-134. [PMID: 33521373 PMCID: PMC7837741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/12/2020] [Accepted: 06/08/2020] [Indexed: 11/26/2022] Open
Abstract
UNLABELLED Radiomics has revolutionized the world of medical imaging. The aim of this review is to guide oncologists in radiomics and its applications in diagnosis, prediction of response and damage, prediction of survival, and prognosis in lung cancer. In this review, we analyzed published literature on PubMed and MEDLINE with papers published in the last 10 years. We included papers in English language with information about radiomics features and diagnostic, predictive, and prognosis of radiomics in lung cancer. All citations were evaluated for relevant content and validation. RELEVANCE FOR PATIENTS The evolution of technology allows the development of computer algorithms that facilitate the diagnosis and evaluation of response after different oncological treatments and their non-invasive follow-up.
Collapse
Affiliation(s)
| | - Nuria Barrios-Campo
- Department of Biomedical Engineering, Madrid Polytechnic University, Madrid, Spain
| | - David Sevillano
- Department of Medical Physics, Ramón y Cajal Hospital, Madrid, Spain
| |
Collapse
|
10
|
2-[ 18F]FDG PET/CT radiomics in lung cancer: An overview of the technical aspect and its emerging role in management of the disease. Methods 2020; 188:84-97. [PMID: 32497604 DOI: 10.1016/j.ymeth.2020.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the most common cancer, worldwide, and a major health issue with a remarkable mortality rate. 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (2-[18F]FDG PET/CT) plays an indispensable role in the management of lung cancer patients. Long-established quantitative parameters such as size, density, and metabolic activity have been and are being employed in the current practice to enhance interpretation and improve diagnostic and prognostic value. The introduction of radiomics analysis revolutionized the quantitative evaluation of medical imaging, revealing data within images beyond visual interpretation. The "big data" are extracted from high-quality images and are converted into information that correlates to relevant genetic, pathologic, clinical, or prognostic features. Technically advanced, diverse methods have been implemented in different studies. The standardization of image acquisition, segmentation and features analysis is still a debated issue. Importantly, a body of features has been extracted and employed for diagnosis, staging, risk stratification, prognostication, and therapeutic response. 2-[18F]FDG PET/CT-derived features show promising value in non-invasively diagnosing the malignant nature of pulmonary nodules, differentiating lung cancer subtypes, and predicting response to different therapies as well as survival. In this review article, we aimed to provide an overview of the technical aspects used in radiomics analysis in non-small cell lung cancer (NSCLC) and elucidate the role of 2-[18F]FDG PET/CT-derived radiomics in the diagnosis, prognostication, and therapeutic response.
Collapse
|
11
|
Qin Q, Shi A, Zhang R, Wen Q, Niu T, Chen J, Qiu Q, Wan Y, Sun X, Xing L. Cone-beam CT radiomics features might improve the prediction of lung toxicity after SBRT in stage I NSCLC patients. Thorac Cancer 2020; 11:964-972. [PMID: 32061061 PMCID: PMC7113065 DOI: 10.1111/1759-7714.13349] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background Stereotactic body radiotherapy (SBRT) is the standard care for inoperable early stage non‐small cell lung cancer (NSCLC). The purpose of our study was to investigate whether a prediction model based on cone‐beam CT (CBCT) plus pretreatment CT radiomics features could improve the prediction of tumor control and lung toxicity after SBRT in comparison to a model based on pretreatment CT radiomics features alone. Methods A total of 34 cases of stage I NSCLC patients who received SBRT were included in the study. The pretreatment planning CT and serial CBCT radiomics features were analyzed using the imaging biomarker explorer (IBEX) software platform. Multivariate logistic regression was conducted for the association between progression‐free survival (PFS), lung toxicity and features. The predictive capabilities of the models based on CBCT and CT features were compared using receiver operating characteristic (ROC) curves. Results Five CBCT features and two planning CT features were correlated with disease progression. Six CBCT features and two planning CT features were related to lung injury. The ROC curves indicated that the model based on the CBCT plus planning CT features might be better than the model based on the planning CT features in predicting lung injury. The other ROC curves indicated that the model based on the planning CT features was similar to the model based on the CBCT plus planning CT features in predicting disease progression. Conclusions Both pretreatment CT and CBCT radiomics features could predict disease progression and lung injury. A model with CBCT plus pretreatment CT radiomics features might improve the prediction of lung toxicity in comparison with a model with pretreatment CT features alone. Key points Significant findings of the study: A model with cone‐beam CT radiomics features plus pre‐treatment CT radiomics features might improve the prediction of lung toxicity after SBRT in stage I NSCLC patients. What this study adds: In the prediction of PFS and lung toxicity in early‐stage NSCLC patients treated with SBRT, CBCT radiomics could be another effective method.
Collapse
Affiliation(s)
- Qingjin Qin
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Anhui Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China.,Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ran Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China.,Shandong University Cheeloo College of Medicine, Jinan, China
| | - Qiang Wen
- Department of Oncology, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tianye Niu
- Nuclear & Radiological Engineering and Medical Physics Programs Woodruff School of Mechanical Engineering Georgia Institute of Technology, Atlanta, Georgia
| | - Jinhu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Qingtao Qiu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Yidong Wan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang
| | - Xiaorong Sun
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China.,Shandong University Cheeloo College of Medicine, Jinan, China.,Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Ligang Xing
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| |
Collapse
|
12
|
Refaee T, Wu G, Ibrahim A, Halilaj I, Leijenaar RTH, Rogers W, Gietema HA, Hendriks LEL, Lambin P, Woodruff HC. The Emerging Role of Radiomics in COPD and Lung Cancer. Respiration 2020; 99:99-107. [PMID: 31991420 DOI: 10.1159/000505429] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/12/2019] [Indexed: 12/24/2022] Open
Abstract
Medical imaging plays a key role in evaluating and monitoring lung diseases such as chronic obstructive pulmonary disease (COPD) and lung cancer. The application of artificial intelligence in medical imaging has transformed medical images into mineable data, by extracting and correlating quantitative imaging features with patients' outcomes and tumor phenotype - a process termed radiomics. While this process has already been widely researched in lung oncology, the evaluation of COPD in this fashion remains in its infancy. Here we outline the main applications of radiomics in lung cancer and briefly review the workflow from image acquisition to the evaluation of model performance. Finally, we discuss the current assessments of COPD and the potential application of radiomics in COPD.
Collapse
Affiliation(s)
- Turkey Refaee
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands, .,Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia,
| | - Guangyao Wu
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Abdallah Ibrahim
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, Centre Hospitalier Universitaire de Liège, Liège, Belgium.,Department of Nuclear Medicine and Comprehensive Diagnostic Center Aachen (CDCA), University Hospital RWTH Aachen University, Aachen, Germany
| | - Iva Halilaj
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - William Rogers
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Hester A Gietema
- Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
13
|
Cheze Le Rest C, Hustinx R. Are radiomics ready for clinical prime-time in PET/CT imaging? THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:347-354. [DOI: 10.23736/s1824-4785.19.03210-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
Radiomics and Texture Analysis in Laryngeal Cancer. Looking for New Frontiers in Precision Medicine through Imaging Analysis. Cancers (Basel) 2019; 11:cancers11101409. [PMID: 31547210 PMCID: PMC6826870 DOI: 10.3390/cancers11101409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/24/2022] Open
Abstract
Radiomics and texture analysis represent a new option in our biomarkers arsenal. These techniques extract a large number of quantitative features, analyzing their properties to incorporate them in clinical decision-making. Laryngeal cancer represents one of the most frequent cancers in the head and neck area. We hypothesized that radiomics features can be included as a laryngeal cancer precision medicine tool, as it is able to non-invasively characterize the overall tumor accounting for heterogeneity, being a prognostic and/or predictive biomarker derived from routine, standard of care, imaging data, and providing support during the follow up of the patient, in some cases avoiding the need for biopsies. The larynx represents a unique diagnostic and therapeutic challenge for clinicians due to its complex tridimensional anatomical structure. Its complex regional and functional anatomy makes it necessary to enhance our diagnostic tools in order to improve decision-making protocols, aimed at better survival and functional results. For this reason, this technique can be an option for monitoring the evolution of the disease, especially in surgical and non-surgical organ preservation treatments. This concise review article will explain basic concepts about radiomics and discuss recent progress and results related to laryngeal cancer.
Collapse
|
15
|
Schwier M, van Griethuysen J, Vangel MG, Pieper S, Peled S, Tempany C, Aerts HJWL, Kikinis R, Fennessy FM, Fedorov A. Repeatability of Multiparametric Prostate MRI Radiomics Features. Sci Rep 2019; 9:9441. [PMID: 31263116 PMCID: PMC6602944 DOI: 10.1038/s41598-019-45766-z] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 06/12/2019] [Indexed: 12/17/2022] Open
Abstract
In this study we assessed the repeatability of radiomics features on small prostate tumors using test-retest Multiparametric Magnetic Resonance Imaging (mpMRI). The premise of radiomics is that quantitative image-based features can serve as biomarkers for detecting and characterizing disease. For such biomarkers to be useful, repeatability is a basic requirement, meaning its value must remain stable between two scans, if the conditions remain stable. We investigated repeatability of radiomics features under various preprocessing and extraction configurations including various image normalization schemes, different image pre-filtering, and different bin widths for image discretization. Although we found many radiomics features and preprocessing combinations with high repeatability (Intraclass Correlation Coefficient > 0.85), our results indicate that overall the repeatability is highly sensitive to the processing parameters. Neither image normalization, using a variety of approaches, nor the use of pre-filtering options resulted in consistent improvements in repeatability. We urge caution when interpreting radiomics features and advise paying close attention to the processing configuration details of reported results. Furthermore, we advocate reporting all processing details in radiomics studies and strongly recommend the use of open source implementations.
Collapse
Affiliation(s)
- Michael Schwier
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Mark G Vangel
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Sharon Peled
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Clare Tempany
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hugo J W L Aerts
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ron Kikinis
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Fraunhofer MEVIS, Bremen, Germany
- Mathematics/Computer Science Faculty, University of Bremen, Bremen, Germany
| | - Fiona M Fennessy
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andriy Fedorov
- Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
The role of functional imaging in lung cancer. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
17
|
Heterogeneity analysis of 18F-FDG PET imaging in oncology: clinical indications and perspectives. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0299-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Sosa Iglesias V, Giuranno L, Dubois LJ, Theys J, Vooijs M. Drug Resistance in Non-Small Cell Lung Cancer: A Potential for NOTCH Targeting? Front Oncol 2018; 8:267. [PMID: 30087852 PMCID: PMC6066509 DOI: 10.3389/fonc.2018.00267] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is a major cause for therapeutic failure in non-small cell lung cancer (NSCLC) leading to tumor recurrence and disease progression. Cell intrinsic mechanisms of resistance include changes in the expression of drug transporters, activation of pro-survival, and anti-apoptotic pathways, as well as non-intrinsic influences of the tumor microenvironment. It has become evident that tumors are composed of a heterogeneous population of cells with different genetic, epigenetic, and phenotypic characteristics that result in diverse responses to therapy, and underlies the emergence of resistant clones. This tumor heterogeneity is driven by subpopulations of tumor cells termed cancer stem cells (CSCs) that have tumor-initiating capabilities, are highly self-renewing, and retain the ability for multi-lineage differentiation. CSCs have been identified in NSCLC and have been associated with chemo- and radiotherapy resistance. Stem cell pathways are frequently deregulated in cancer and are implicated in recurrence after treatment. Here, we focus on the NOTCH signaling pathway, which has a role in stem cell maintenance in non-squamous non-small lung cancer, and we critically assess the potential for targeting the NOTCH pathway to overcome resistance to chemotherapeutic and targeted agents using both preclinical and clinical evidence.
Collapse
Affiliation(s)
- Venus Sosa Iglesias
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Lorena Giuranno
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Ludwig J Dubois
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Jan Theys
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Marc Vooijs
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| |
Collapse
|
19
|
Pu Y, Zhang JX, Liu H, Appelbaum D, Meng J, Penney BC. Developing and validating a novel metabolic tumor volume risk stratification system for supplementing non-small cell lung cancer staging. Eur J Nucl Med Mol Imaging 2018; 45:2079-2092. [PMID: 29882161 DOI: 10.1007/s00259-018-4059-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/23/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE We hypothesized that whole-body metabolic tumor volume (MTVwb) could be used to supplement non-small cell lung cancer (NSCLC) staging due to its independent prognostic value. The goal of this study was to develop and validate a novel MTVwb risk stratification system to supplement NSCLC staging. METHODS We performed an IRB-approved retrospective review of 935 patients with NSCLC and FDG-avid tumor divided into modeling and validation cohorts based on the type of PET/CT scanner used for imaging. In addition, sensitivity analysis was conducted by dividing the patient population into two randomized cohorts. Cox regression and Kaplan-Meier survival analyses were performed to determine the prognostic value of the MTVwb risk stratification system. RESULTS The cut-off values (10.0, 53.4 and 155.0 mL) between the MTVwb quartiles of the modeling cohort were applied to both the modeling and validation cohorts to determine each patient's MTVwb risk stratum. The survival analyses showed that a lower MTVwb risk stratum was associated with better overall survival (all p < 0.01), independent of TNM stage together with other clinical prognostic factors, and the discriminatory power of the MTVwb risk stratification system, as measured by Gönen and Heller's concordance index, was not significantly different from that of TNM stage in both cohorts. Also, the prognostic value of the MTVwb risk stratum was robust in the two randomized cohorts. The discordance rate between the MTVwb risk stratum and TNM stage or substage was 45.1% in the modeling cohort and 50.3% in the validation cohort. CONCLUSION This study developed and validated a novel MTVwb risk stratification system, which has prognostic value independent of the TNM stage and other clinical prognostic factors in NSCLC, suggesting that it could be used for further NSCLC pretreatment assessment and for refining treatment decisions in individual patients.
Collapse
Affiliation(s)
- Yonglin Pu
- Department of Radiology, The University of Chicago, 5841 S. Maryland Ave., MC 2026, Chicago, IL, 60637, USA.
| | - James X Zhang
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital and Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Daniel Appelbaum
- Department of Radiology, The University of Chicago, 5841 S. Maryland Ave., MC 2026, Chicago, IL, 60637, USA
| | - Jianfeng Meng
- Department of Respiratory Medicine, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541002, Guangxi Zhuang Autonomous Region, China
| | - Bill C Penney
- Department of Radiology, The University of Chicago, 5841 S. Maryland Ave., MC 2026, Chicago, IL, 60637, USA
| |
Collapse
|
20
|
Carvalho S, Leijenaar RTH, Troost EGC, van Timmeren JE, Oberije C, van Elmpt W, de Geus-Oei LF, Bussink J, Lambin P. 18F-fluorodeoxyglucose positron-emission tomography (FDG-PET)-Radiomics of metastatic lymph nodes and primary tumor in non-small cell lung cancer (NSCLC) - A prospective externally validated study. PLoS One 2018; 13:e0192859. [PMID: 29494598 PMCID: PMC5832210 DOI: 10.1371/journal.pone.0192859] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/31/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Lymph node stage prior to treatment is strongly related to disease progression and poor prognosis in non-small cell lung cancer (NSCLC). However, few studies have investigated metabolic imaging features derived from pre-radiotherapy 18F-fluorodeoxyglucose (FDG) positron-emission tomography (PET) of metastatic hilar/mediastinal lymph nodes (LNs). We hypothesized that these would provide complementary prognostic information to FDG-PET descriptors to only the primary tumor (tumor). METHODS Two independent cohorts of 262 and 50 node-positive NSCLC patients were used for model development and validation. Image features (i.e. Radiomics) including shape and size, first order statistics, texture, and intensity-volume histograms (IVH) (http://www.radiomics.io/) were evaluated by univariable Cox regression on the development cohort. Prognostic modeling was conducted with a 10-fold cross-validated least absolute shrinkage and selection operator (LASSO), automatically selecting amongst FDG-PET-Radiomics descriptors from (1) tumor, (2) LNs or (3) both structures. Performance was assessed with the concordance-index. Development data are publicly available at www.cancerdata.org and Dryad (doi:10.5061/dryad.752153b). RESULTS Common SUV descriptors (maximum, peak, and mean) were significantly related to overall survival when extracted from LNs, as were LN volume and tumor load (summed tumor and LNs' volumes), though this was not true for either SUV metrics or tumor's volume. Feature selection exclusively from imaging information based on FDG-PET-Radiomics, exhibited performances of (1) 0.53 -external 0.54, when derived from the tumor, (2) 0.62 -external 0.56 from LNs, and (3) 0.62 -external 0.59 from both structures, including at least one feature from each sub-category, except IVH. CONCLUSION Combining imaging information based on FDG-PET-Radiomics features from tumors and LNs is desirable to achieve a higher prognostic discriminative power for NSCLC.
Collapse
Affiliation(s)
- Sara Carvalho
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
| | - Ralph T. H. Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
| | - Esther G. C. Troost
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Medical Faculty and University Hospital Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany
- OncoRay, National Centre for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus of Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Janna E. van Timmeren
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
| | - Cary Oberije
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology and Nuclear Medicine, Radboud UMC, Nijmegen, the Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW–School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC +), Maastricht, the Netherlands
- * E-mail:
| |
Collapse
|
21
|
Lazzeroni M, Uhrdin J, Carvalho S, van Elmpt W, Lambin P, Dasu A, Wersäll P, Toma-Dasu I. Evaluation of third treatment week as temporal window for assessing responsiveness on repeated FDG-PET-CT scans in Non-Small Cell Lung Cancer patients. Phys Med 2018. [DOI: 10.1016/j.ejmp.2018.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Shi L, He Y, Yuan Z, Benedict S, Valicenti R, Qiu J, Rong Y. Radiomics for Response and Outcome Assessment for Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2018; 17:1533033818782788. [PMID: 29940810 PMCID: PMC6048673 DOI: 10.1177/1533033818782788] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/09/2018] [Accepted: 05/16/2018] [Indexed: 12/24/2022] Open
Abstract
Routine follow-up visits and radiographic imaging are required for outcome evaluation and tumor recurrence monitoring. Yet more personalized surveillance is required in order to sufficiently address the nature of heterogeneity in nonsmall cell lung cancer and possible recurrences upon completion of treatment. Radiomics, an emerging noninvasive technology using medical imaging analysis and data mining methodology, has been adopted to the area of cancer diagnostics in recent years. Its potential application in response assessment for cancer treatment has also drawn considerable attention. Radiomics seeks to extract a large amount of valuable information from patients' medical images (both pretreatment and follow-up images) and quantitatively correlate image features with diagnostic and therapeutic outcomes. Radiomics relies on computers to identify and analyze vast amounts of quantitative image features that were previously overlooked, unmanageable, or failed to be identified (and recorded) by human eyes. The research area has been focusing on the predictive accuracy of pretreatment features for outcome and response and the early discovery of signs of tumor response, recurrence, distant metastasis, radiation-induced lung injury, death, and other outcomes, respectively. This review summarized the application of radiomics in response assessments in radiotherapy and chemotherapy for non-small cell lung cancer, including image acquisition/reconstruction, region of interest definition/segmentation, feature extraction, and feature selection and classification. The literature search for references of this article includes PubMed peer-reviewed publications over the last 10 years on the topics of radiomics, textural features, radiotherapy, chemotherapy, lung cancer, and response assessment. Summary tables of radiomics in response assessment and treatment outcome prediction in radiation oncology have been developed based on the comprehensive review of the literature.
Collapse
Affiliation(s)
- Liting Shi
- Department of Radiology, Taishan Medical University, Tai’an, China
| | - Yaoyao He
- Department of Radiology, Taishan Medical University, Tai’an, China
| | - Zilong Yuan
- Department of Radiology, Hubei Cancer Hospital, Wuhan, China
| | - Stanley Benedict
- Department of Radiation Oncology, University of California Davis
Comprehensive Cancer Center, Sacramento, CA, USA
| | - Richard Valicenti
- Department of Radiation Oncology, University of California Davis
Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jianfeng Qiu
- Department of Radiology, Taishan Medical University, Tai’an, China
| | - Yi Rong
- Department of Radiation Oncology, University of California Davis
Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
23
|
Whole body metabolic tumor volume is a prognostic marker in patients with newly diagnosed stage 3B non-small cell lung cancer, confirmed with external validation. Eur J Hybrid Imaging 2017; 1:8. [PMID: 29782599 PMCID: PMC5954780 DOI: 10.1186/s41824-017-0013-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose TNM Stage 3B encompasses a wide range of primary tumor and nodal metastatic tumor burden. This study aimed to evaluate the prognostic value of quantitative FDG PET/CT parameters in patients with newly diagnosed Stage 3B Non-Small Cell Lung Cancer (NSCLC). Materials and Methods Institutional review board approved retrospective study identified patients diagnosed with Stage 3B NSCLC (8th edition TNM classification) on baseline FDG PET/CT at two medical centers (Medical centers A and B), between Feb 2004 and Dec 2014. Patients were excluded if they had prior NSCLC treatment or recent diagnosis of a second primary cancer. Quantitative FDG PET/CT parameters including whole body metabolic tumor volume (MTVwb), total lesion glycolysis (TLGwb), and maximum standardized uptake value (SUVmaxwb) were measured from baseline PET/CT using Edge method with Mimvista software. The primary endpoint was overall survival (OS). Cox proportional hazard regression and Kaplan-Meier overall survival analyses were used to test for an association between OS and quantitative FDG PET/CT parameters. The distributions of MTVwb, TLGwb, SUVmaxwb were skewed, so a natural logarithm transformation was applied and the transformed variables [(ln(MTVwb), ln(TLGwb), and ln(SUVmaxwb)] were used in the analysis. Results The training set included 110 patients from center A with Stage 3B NSCLC. 78.2% of patients expired during follow-up. Median OS was 14 months. 1-year, 2-year, and 5-year OS was 56.5%, 34.6% and 13.9%, respectively. Univariate Cox regression analysis showed no significant difference in OS on the basis of age, gender, histology, ln(TLGwb), or ln(SUVmaxwb). ln(MTVwb) was positively associated with OS [hazard ratio (HR) of 1.23, p = 0.037]. This association persisted on multivariate Cox regression analysis (HR 1.28, p = 0.043), with adjustments for age, gender, treatment and tumor histology. External validation with 44 patients from center B confirmed increasing MTVwb was associated significantly worse OS. An MTVwb cut-off point of 85.6 mL significantly stratified Stage 3B NSCLC patient prognosis. Conclusion MTVwb is a prognostic marker for OS in patients with Stage 3B NSCLC, independent of age, gender, treatment, and tumor histology.
Collapse
|
24
|
de Jong EEC, van Elmpt W, Hoekstra OS, Groen HJM, Smit EF, Boellaard R, Lambin P, Dingemans AMC. Quality assessment of positron emission tomography scans: recommendations for future multicentre trials. Acta Oncol 2017; 56:1459-1464. [PMID: 28830270 DOI: 10.1080/0284186x.2017.1346824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Standardization protocols and guidelines for positron emission tomography (PET) in multicenter trials are available, despite a large variability in image acquisition and reconstruction parameters exist. In this study, we investigated the compliance of PET scans to the guidelines of the European Association of Nuclear Medicine (EANM). From these results, we provide recommendations for future multicenter studies using PET. MATERIAL AND METHODS Patients included in a multicenter randomized phase II study had repeated PET scans for early response assessment. Relevant acquisition and reconstruction parameters were extracted from the digital imaging and communications in medicine (DICOM) header of the images. The PET image parameters were compared to the guidelines of the EANM for tumor imaging version 1.0 recommended parameters. RESULTS From the 223 included patients, 167 baseline scans and 118 response scans were available from 15 hospitals. Scans of 19% of the patients had an uptake time that fulfilled the Uniform Protocols for Imaging in Clinical Trials response assessment criteria. The average quality score over all hospitals was 69%. Scans with a non-compliant uptake time had a larger standard deviation of the mean standardized uptake value (SUVmean) of the liver than scans with compliant uptake times. CONCLUSIONS Although a standardization protocol was agreed on, there was a large variability in imaging parameters. For future, multicenter studies including PET imaging a prospective central quality review during patient inclusion is needed to improve compliance with image standardization protocols as defined by EANM.
Collapse
Affiliation(s)
- Evelyn E. C. de Jong
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Otto S. Hoekstra
- Department of Nuclear Medicine & PET Research, VU University Medical Center, Amsterdam, Netherlands
| | - Harry J. M. Groen
- Department of Pulmonary Diseases, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Egbert F. Smit
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, Netherlands
- Department of Thoracic Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Anne-Marie C. Dingemans
- Department of Pulmonology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
25
|
van Timmeren JE, Leijenaar RTH, van Elmpt W, Reymen B, Lambin P. Feature selection methodology for longitudinal cone-beam CT radiomics. Acta Oncol 2017; 56:1537-1543. [PMID: 28826307 DOI: 10.1080/0284186x.2017.1350285] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cone-beam CT (CBCT) scans are typically acquired daily for positioning verification of non-small cell lung cancer (NSCLC) patients. Quantitative information, derived using radiomics, can potentially contribute to (early) treatment adaptation. The aims of this study were to (1) describe and investigate a methodology for feature selection of a longitudinal radiomics approach (2) investigate which time-point during treatment is potentially useful for early treatment response assessment. MATERIAL AND METHODS For 90 NSCLC patients CBCT scans of the first two fractions of treatment (considered as 'test-retest' scans) were analyzed, as well as weekly CBCT images. One hundred and sixteen radiomic features were extracted from the GTV of all scans and subsequently absolute and relative differences were calculated between weekly CBCT images and the CBCT of the first fraction. Test-retest scans were used to determine the smallest detectable change (C = 1.96 * SD) allowing for feature selection by choosing a minimum number of patients for which a feature should change more than 'C' to be considered as relevant. Analysis of which features change at which moment during treatment was used to investigate which time-point is potentially relevant to extract longitudinal radiomics information for early treatment response assessment. RESULTS A total of six absolute delta features changed for at least ten patients at week 2 of treatment and increased to 61 at week 3, 79 at week 4 and 85 at week 5. There was 93% overlap between features selected at week 3 and the other weeks. CONCLUSIONS This study describes a feature selection methodology for longitudinal radiomics that is able to select reproducible delta radiomics features that are informative due to their change during treatment, which can potentially be used for treatment decisions concerning adaptive radiotherapy. Nonetheless, the prognostic value of the selected delta radiomic features should be investigated in future studies.
Collapse
Affiliation(s)
- Janna E. van Timmeren
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, The Netherlands
| | - Ralph T. H. Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, The Netherlands
| | - Bart Reymen
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, The Netherlands
| |
Collapse
|
26
|
Im HJ, Bradshaw T, Solaiyappan M, Cho SY. Current Methods to Define Metabolic Tumor Volume in Positron Emission Tomography: Which One is Better? Nucl Med Mol Imaging 2017; 52:5-15. [PMID: 29391907 DOI: 10.1007/s13139-017-0493-6] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/17/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Numerous methods to segment tumors using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) have been introduced. Metabolic tumor volume (MTV) refers to the metabolically active volume of the tumor segmented using FDG PET, and has been shown to be useful in predicting patient outcome and in assessing treatment response. Also, tumor segmentation using FDG PET has useful applications in radiotherapy treatment planning. Despite extensive research on MTV showing promising results, MTV is not used in standard clinical practice yet, mainly because there is no consensus on the optimal method to segment tumors in FDG PET images. In this review, we discuss currently available methods to measure MTV using FDG PET, and assess the advantages and disadvantages of the methods.
Collapse
Affiliation(s)
- Hyung-Jun Im
- 1Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI USA.,2Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Tyler Bradshaw
- 1Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Meiyappan Solaiyappan
- 3Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Steve Y Cho
- 1Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI USA.,3Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA.,4University of Wisconsin Carbone Cancer Center, Madison, WI USA
| |
Collapse
|
27
|
Antunovic L, Gallivanone F, Sollini M, Sagona A, Invento A, Manfrinato G, Kirienko M, Tinterri C, Chiti A, Castiglioni I. [ 18F]FDG PET/CT features for the molecular characterization of primary breast tumors. Eur J Nucl Med Mol Imaging 2017; 44:1945-1954. [PMID: 28711994 DOI: 10.1007/s00259-017-3770-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE The aim of this study was to evaluate the role of imaging features derived from [18F]FDG-PET/CT to provide in vivo characterization of breast cancer (BC). METHODS Images from 43 patients with a first diagnosis of BC were reviewed. Images were acquired before any treatment. Histological data were derived from pretreatment biopsy or surgical histological specimen; these included tumor type, grade, ER and PgR receptor status, lymphovascular invasion, Ki67 index, HER2 status, and molecular subtype. Standard parameters (SUVmean, TLG, MTV) and advanced imaging features (histogram-based and shape and size features) were evaluated. Univariate analysis, hierarchical clustering analysis, and exact Fisher's test were used for statistical analysis of data. Imaging-derived metrics were reduced evaluating the mutual correlation within group of features as well as the mutual correlation between groups of features to form a signature. RESULTS A significant correlation was found between some advanced imaging features and the histological type. Different molecular subtypes were characterized by different values of two histogram-based features (median and energy). A significant association was observed between the imaging signature and luminal A and luminal B HER2 negative molecular subtype and also when considering luminal A, luminal B HER2-negative and HER2-positive groups. Similar results were found between the signature and all five molecular subtypes and also when considering the histological types of BC. CONCLUSIONS Our results suggest a complementary role of standard PET imaging parameters and advanced imaging features for the in vivo biological characterization of BC lesions.
Collapse
Affiliation(s)
- Lidija Antunovic
- Nuclear Medicine Department, Humanitas Research Hospital, Via A. Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Francesca Gallivanone
- Laboratory of Innovation and Integration in Molecular Medicine, Institute of Molecular Bioimaging and Physiology, National Research Council, Via F. Cervi 93, 20090, Segrate, Milan, Italy
| | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Via A. Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Andrea Sagona
- Breast Unit, Humanitas Research Hospital, Via A. Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Alessandra Invento
- Breast Unit, Integrated University Hospital, Piazzale A. Stefani 1, Borgo Trento, 37126, Verona, Italy
| | - Giulia Manfrinato
- Residency Program in Nuclear Medicine, University of Milan, Via A. di Rudini 8, 20100, Milan, Italy
| | - Margarita Kirienko
- Department of Biomedical Sciences, Humanitas University, Via A. Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Corrado Tinterri
- Breast Unit, Humanitas Research Hospital, Via A. Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Arturo Chiti
- Nuclear Medicine Department, Humanitas Research Hospital, Via A. Manzoni 56, 20089, Rozzano, Milan, Italy. .,Department of Biomedical Sciences, Humanitas University, Via A. Manzoni 113, 20089, Rozzano, Milan, Italy.
| | - Isabella Castiglioni
- Laboratory of Innovation and Integration in Molecular Medicine, Institute of Molecular Bioimaging and Physiology, National Research Council, Via F. Cervi 93, 20090, Segrate, Milan, Italy
| |
Collapse
|
28
|
van Timmeren JE, Leijenaar RT, van Elmpt W, Reymen B, Oberije C, Monshouwer R, Bussink J, Brink C, Hansen O, Lambin P. Survival prediction of non-small cell lung cancer patients using radiomics analyses of cone-beam CT images. Radiother Oncol 2017; 123:363-369. [DOI: 10.1016/j.radonc.2017.04.016] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/20/2017] [Accepted: 04/17/2017] [Indexed: 01/20/2023]
|
29
|
Sollini M, Cozzi L, Antunovic L, Chiti A, Kirienko M. PET Radiomics in NSCLC: state of the art and a proposal for harmonization of methodology. Sci Rep 2017; 7:358. [PMID: 28336974 PMCID: PMC5428425 DOI: 10.1038/s41598-017-00426-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/23/2017] [Indexed: 12/21/2022] Open
Abstract
Imaging with positron emission tomography (PET)/computed tomography (CT) is crucial in the management of cancer because of its value in tumor staging, response assessment, restaging, prognosis and treatment responsiveness prediction. In the last years, interest has grown in texture analysis which provides an "in-vivo" lesion characterization, and predictive information in several malignances including NSCLC; however several drawbacks and limitations affect these studies, especially because of lack of standardization in features calculation, definitions and methodology reporting. The present paper provides a comprehensive review of literature describing the state-of-the-art of FDG-PET/CT texture analysis in NSCLC, suggesting a proposal for harmonization of methodology.
Collapse
Affiliation(s)
- M Sollini
- Department of Biomedical Sciences, Humanitas University, via Manzoni, 113-20089, Rozzano, (Milan), Italy.
| | - L Cozzi
- Department of Biomedical Sciences, Humanitas University, via Manzoni, 113-20089, Rozzano, (Milan), Italy
- Radiotherapy and Radiosurgery Unit, Humanitas Clinical and Research Center, via Manzoni, 56-20089, Rozzano, (Milan), Italy
| | - L Antunovic
- Nuclear Medicine Unit, Humanitas Clinical and Research Center, via Manzoni, 56-20089, Rozzano, (Milan), Italy
| | - A Chiti
- Department of Biomedical Sciences, Humanitas University, via Manzoni, 113-20089, Rozzano, (Milan), Italy
- Nuclear Medicine Unit, Humanitas Clinical and Research Center, via Manzoni, 56-20089, Rozzano, (Milan), Italy
| | - M Kirienko
- Department of Biomedical Sciences, Humanitas University, via Manzoni, 113-20089, Rozzano, (Milan), Italy
| |
Collapse
|
30
|
Lambin P, Zindler J, Vanneste BGL, De Voorde LV, Eekers D, Compter I, Panth KM, Peerlings J, Larue RTHM, Deist TM, Jochems A, Lustberg T, van Soest J, de Jong EEC, Even AJG, Reymen B, Rekers N, van Gisbergen M, Roelofs E, Carvalho S, Leijenaar RTH, Zegers CML, Jacobs M, van Timmeren J, Brouwers P, Lal JA, Dubois L, Yaromina A, Van Limbergen EJ, Berbee M, van Elmpt W, Oberije C, Ramaekers B, Dekker A, Boersma LJ, Hoebers F, Smits KM, Berlanga AJ, Walsh S. Decision support systems for personalized and participative radiation oncology. Adv Drug Deliv Rev 2017; 109:131-153. [PMID: 26774327 DOI: 10.1016/j.addr.2016.01.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/08/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
A paradigm shift from current population based medicine to personalized and participative medicine is underway. This transition is being supported by the development of clinical decision support systems based on prediction models of treatment outcome. In radiation oncology, these models 'learn' using advanced and innovative information technologies (ideally in a distributed fashion - please watch the animation: http://youtu.be/ZDJFOxpwqEA) from all available/appropriate medical data (clinical, treatment, imaging, biological/genetic, etc.) to achieve the highest possible accuracy with respect to prediction of tumor response and normal tissue toxicity. In this position paper, we deliver an overview of the factors that are associated with outcome in radiation oncology and discuss the methodology behind the development of accurate prediction models, which is a multi-faceted process. Subsequent to initial development/validation and clinical introduction, decision support systems should be constantly re-evaluated (through quality assurance procedures) in different patient datasets in order to refine and re-optimize the models, ensuring the continuous utility of the models. In the reasonably near future, decision support systems will be fully integrated within the clinic, with data and knowledge being shared in a standardized, dynamic, and potentially global manner enabling truly personalized and participative medicine.
Collapse
Affiliation(s)
- Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Jaap Zindler
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ben G L Vanneste
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lien Van De Voorde
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Daniëlle Eekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Inge Compter
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kranthi Marella Panth
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jurgen Peerlings
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ruben T H M Larue
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Timo M Deist
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arthur Jochems
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tim Lustberg
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Johan van Soest
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evelyn E C de Jong
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Aniek J G Even
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bart Reymen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Nicolle Rekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marike van Gisbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Erik Roelofs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sara Carvalho
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Catharina M L Zegers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maria Jacobs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janita van Timmeren
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patricia Brouwers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jonathan A Lal
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ludwig Dubois
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ala Yaromina
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evert Jan Van Limbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maaike Berbee
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Cary Oberije
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bram Ramaekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Liesbeth J Boersma
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kim M Smits
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Adriana J Berlanga
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sean Walsh
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
31
|
Ma X, Siegelman J, Paik DS, Mulshine JL, St Pierre S, Buckler AJ. Volumes Learned: It Takes More Than Size to "Size Up" Pulmonary Lesions. Acad Radiol 2016; 23:1190-8. [PMID: 27287713 DOI: 10.1016/j.acra.2016.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 04/08/2016] [Accepted: 04/10/2016] [Indexed: 12/17/2022]
Abstract
RATIONALE AND OBJECTIVES This study aimed to review the current understanding and capabilities regarding use of imaging for noninvasive lesion characterization and its relationship to lung cancer screening and treatment. MATERIALS AND METHODS Our review of the state of the art was broken down into questions about the different lung cancer image phenotypes being characterized, the role of imaging and requirements for increasing its value with respect to increasing diagnostic confidence and quantitative assessment, and a review of the current capabilities with respect to those needs. RESULTS The preponderance of the literature has so far been focused on the measurement of lesion size, with increasing contributions being made to determine the formal performance of scanners, measurement tools, and human operators in terms of bias and variability. Concurrently, an increasing number of investigators are reporting utility and predictive value of measures other than size, and sensitivity and specificity is being reported. Relatively little has been documented on quantitative measurement of non-size features with corresponding estimation of measurement performance and reproducibility. CONCLUSIONS The weight of the evidence suggests characterization of pulmonary lesions built on quantitative measures adds value to the screening for, and treatment of, lung cancer. Advanced image analysis techniques may identify patterns or biomarkers not readily assessed by eye and may also facilitate management of multidimensional imaging data in such a way as to efficiently integrate it into the clinical workflow.
Collapse
Affiliation(s)
- Xiaonan Ma
- Elucid Bioimaging Inc., 225 Main Street, Wenham, MA 01984.
| | - Jenifer Siegelman
- Department of Radiology, Brigham and Women's Hospital, Boston Massachusetts; Department of Radiology (hospital-based), Harvard Medical School, Boston, Massachusetts
| | - David S Paik
- Elucid Bioimaging Inc., 225 Main Street, Wenham, MA 01984
| | - James L Mulshine
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | | |
Collapse
|
32
|
Larue RTHM, Van De Voorde L, Berbée M, van Elmpt WJC, Dubois LJ, Panth KM, Peeters SGJA, Claessens A, Schreurs WMJ, Nap M, Warmerdam FARM, Erdkamp FLG, Sosef MN, Lambin P. A phase 1 'window-of-opportunity' trial testing evofosfamide (TH-302), a tumour-selective hypoxia-activated cytotoxic prodrug, with preoperative chemoradiotherapy in oesophageal adenocarcinoma patients. BMC Cancer 2016; 16:644. [PMID: 27535748 PMCID: PMC4989456 DOI: 10.1186/s12885-016-2709-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 08/11/2016] [Indexed: 01/03/2023] Open
Abstract
Background Neo-adjuvant chemoradiotherapy followed by surgery is the standard treatment with curative intent for oesophageal cancer patients, with 5-year overall survival rates up to 50 %. However, patients’ quality of life is severely compromised by oesophagectomy, and eventually many patients die due to metastatic disease. Most solid tumours, including oesophageal cancer, contain hypoxic regions that are more resistant to chemoradiotherapy. The hypoxia-activated prodrug evofosfamide works as a DNA-alkylating agent under these hypoxic conditions, which directly kills hypoxic cancer cells and potentially minimizes resistance to conventional therapy. This drug has shown promising results in several clinical studies when combined with chemotherapy. Therefore, in this phase I study we investigate the safety of evofosfamide added to the chemoradiotherapy treatment of oesophageal cancer. Methods/Design A phase I, non-randomized, single-centre, open-label, 3 + 3 trial with repeated hypoxia PET imaging, will test the safety of evofosfamide in combination with neo-adjuvant chemoradiotherapy in potentially resectable oesophageal adenocarcinoma patients. Investigated dose levels range from 120 mg/m2 to 340 mg/m2. Evofosfamide will be administered one week before the start of chemoradiotherapy (CROSS-regimen) and repeated weekly up to a total of six doses. PET/CT acquisitions with hypoxia tracer 18F-HX4 will be made before and after the first administration of evofosfamide, allowing early assessment of changes in hypoxia, accompanied with blood sampling to measure hypoxia blood biomarkers. Oesophagectomy will be performed according to standard clinical practice. Higher grade and uncommon non-haematological, haematological, and post-operative toxicities are the primary endpoints according to the CTCAEv4.0 and Clavien-Dindo classifications. Secondary endpoints are reduction in hypoxic fraction based on 18F-HX4 imaging, pathological complete response, histopathological negative circumferential resection margin (R0) rate, local and distant recurrence rate, and progression free and overall survival. Discussion This is the first clinical trial testing evofosfamide in combination with chemoradiotherapy. The primary objective is to determine the dose limiting toxicity of this combined treatment and herewith to define the maximum tolerated dose and recommended phase 2 dose for future clinical studies. The addition of non-invasive repeated hypoxia imaging (‘window-of-opportunity’) enables us to identify the biologically effective dose. We believe this approach could also be used for other hypoxia targeted drugs. Trial registration ClinicalTrials.gov Identifier: NCT02598687.
Collapse
Affiliation(s)
- Ruben T H M Larue
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Lien Van De Voorde
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Maaike Berbée
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Wouter J C van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ludwig J Dubois
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kranthi M Panth
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sarah G J A Peeters
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Cancer Research UK & Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Ann Claessens
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Wendy M J Schreurs
- Department of Nuclear Medicine, Zuyderland Medical Centre, Sittard-Geleen/Heerlen, The Netherlands
| | - Marius Nap
- Department of Pathology, Zuyderland Medical Centre, Sittard-Geleen/Heerlen, The Netherlands
| | - Fabiënne A R M Warmerdam
- Department of Medical Oncology, Zuyderland Medical Centre, Sittard-Geleen/Heerlen, The Netherlands
| | - Frans L G Erdkamp
- Department of Medical Oncology, Zuyderland Medical Centre, Sittard-Geleen/Heerlen, The Netherlands
| | - Meindert N Sosef
- Department of Surgery, Zuyderland Medical Centre, Sittard-Geleen/Heerlen, The Netherlands.,Surgical Collaborative Network Limburg, Limburg, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
33
|
Predictive value of repeated F-18 FDG PET/CT parameters changes during preoperative chemoradiotherapy to predict pathologic response and overall survival in locally advanced esophageal adenocarcinoma patients. Cancer Chemother Pharmacol 2016; 77:723-31. [PMID: 26891957 DOI: 10.1007/s00280-016-2988-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/06/2016] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The goal of the current study was to investigate the predictive and prognostic values of repeated F-18 FDG PET/CT parameter changes for prediction of complete pathologic response (pCR) in patients with adenocarcinoma of locally advanced esophageal cancer (LAEC) who received preoperative concurrent chemoradiotherapy (PCRT). METHODS A total 53 patients with LAEC patients were included in the current study. All patients were evaluated by F-18 FDG PET/CT before and during chemoradiotherapy. The percent changes (∆, %) in F-18 FDG PET/CT parameters were used to predict pCR and to calculate overall survival (OS). The predictive value for pCR of F-18 FDG PET/CT cutoff values was determined by ROC analysis. The prognostic significance was assessed using Kaplan-Meier analysis. RESULTS pCR occurred in 15 patients (28.3 %). When ΔSUVmax > 23.5 % was used as cutoff, the sensitivity and specificity of F-18 FDG PET/CT for prediction of pCR were 100 % and 52.6, respectively. The AUC was 0.750 (95 % CI; 0.612-0.859), and standard error (SE) was 0.0633 (p = 0.0002). ΔMTV resulted in 80 % sensitivity, 76.3 % specificity, and 0.731 AUC (95 % CI; 0.591-0.843, SE = 0.077, p = 0.0027) for cutoff values >25.5 %. When ΔTLG > 44.8 % was used as cutoff, the sensitivity and specificity of F-18 FDG PET/CT for prediction of pCR were 100 and 65.8 %, respectively. The AUC was 0.893 (95 % CI; 0.777-0.961), and SE was 0.0431 (p < 0.0001). Kaplan-Meier analysis showed that high ΔSUVmax, ΔMTV, and ΔTLG were associated with improved OS. CONCLUSION In conclusion, the current study shows the capability of the changes (Δ) in repeated F-18 FDG PET/CT parameters to predict the achievement of pCR during PCRT in LAEC patients. Among the parameters, the ΔSUVmax, ΔMTV, and ΔTLG were predictors for pCR and well associated with OS.
Collapse
|
34
|
Chang S, Kim SJ. Prediction of Recurrence and Mortality of Locally Advanced Esophageal Cancer Patients Using Pretreatment F-18 FDG PET/CT Parameters: Intratumoral Heterogeneity, SUV, and Volumetric Parameters. Cancer Biother Radiopharm 2016; 31:1-6. [DOI: 10.1089/cbr.2015.1932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Samuel Chang
- Department of Radiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Seong-Jang Kim
- Department of Nuclear Medicine, Pusan National University Hospital, Busan, Korea
- Medical Research Institute, Pusan National University, Busan, Korea
| |
Collapse
|
35
|
Parmar C, Grossmann P, Rietveld D, Rietbergen MM, Lambin P, Aerts HJWL. Radiomic Machine-Learning Classifiers for Prognostic Biomarkers of Head and Neck Cancer. Front Oncol 2015; 5:272. [PMID: 26697407 PMCID: PMC4668290 DOI: 10.3389/fonc.2015.00272] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION "Radiomics" extracts and mines a large number of medical imaging features in a non-invasive and cost-effective way. The underlying assumption of radiomics is that these imaging features quantify phenotypic characteristics of an entire tumor. In order to enhance applicability of radiomics in clinical oncology, highly accurate and reliable machine-learning approaches are required. In this radiomic study, 13 feature selection methods and 11 machine-learning classification methods were evaluated in terms of their performance and stability for predicting overall survival in head and neck cancer patients. METHODS Two independent head and neck cancer cohorts were investigated. Training cohort HN1 consisted of 101 head and neck cancer patients. Cohort HN2 (n = 95) was used for validation. A total of 440 radiomic features were extracted from the segmented tumor regions in CT images. Feature selection and classification methods were compared using an unbiased evaluation framework. RESULTS We observed that the three feature selection methods minimum redundancy maximum relevance (AUC = 0.69, Stability = 0.66), mutual information feature selection (AUC = 0.66, Stability = 0.69), and conditional infomax feature extraction (AUC = 0.68, Stability = 0.7) had high prognostic performance and stability. The three classifiers BY (AUC = 0.67, RSD = 11.28), RF (AUC = 0.61, RSD = 7.36), and NN (AUC = 0.62, RSD = 10.52) also showed high prognostic performance and stability. Analysis investigating performance variability indicated that the choice of classification method is the major factor driving the performance variation (29.02% of total variance). CONCLUSION Our study identified prognostic and reliable machine-learning methods for the prediction of overall survival of head and neck cancer patients. Identification of optimal machine-learning methods for radiomics-based prognostic analyses could broaden the scope of radiomics in precision oncology and cancer care.
Collapse
Affiliation(s)
- Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Radiation Oncology (MAASTRO), Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute , Boston, MA , USA
| | - Derek Rietveld
- Department of Radiation Oncology, VU University Medical Center , Amsterdam , Netherlands
| | - Michelle M Rietbergen
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center , Amsterdam , Netherlands
| | - Philippe Lambin
- Radiation Oncology (MAASTRO), Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute , Boston, MA , USA
| |
Collapse
|
36
|
Grau C, Overgaard J, Høyer M, Tanderup K, Lindegaard JC, Muren LP. Biology-guided adaptive radiotherapy (BiGART) is progressing towards clinical reality. Acta Oncol 2015; 54:1245-50. [PMID: 26390238 DOI: 10.3109/0284186x.2015.1076992] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Cai Grau
- a Department of Oncology , Aarhus University Hospital , Aarhus , Denmark
| | - Jens Overgaard
- b Department of Experimental Clinical Oncology , Aarhus University Hospital , Aarhus , Denmark
| | - Morten Høyer
- a Department of Oncology , Aarhus University Hospital , Aarhus , Denmark
| | - Kari Tanderup
- a Department of Oncology , Aarhus University Hospital , Aarhus , Denmark
- c Department of Medical Physics , Aarhus University Hospital , Aarhus , Denmark
| | | | - Ludvig Paul Muren
- a Department of Oncology , Aarhus University Hospital , Aarhus , Denmark
- c Department of Medical Physics , Aarhus University Hospital , Aarhus , Denmark
| |
Collapse
|
37
|
Panth KM, Leijenaar RT, Carvalho S, Lieuwes NG, Yaromina A, Dubois L, Lambin P. Is there a causal relationship between genetic changes and radiomics-based image features? An in vivo preclinical experiment with doxycycline inducible GADD34 tumor cells. Radiother Oncol 2015; 116:462-6. [DOI: 10.1016/j.radonc.2015.06.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/28/2015] [Accepted: 06/09/2015] [Indexed: 12/25/2022]
|
38
|
Leijenaar RTH, Nalbantov G, Carvalho S, van Elmpt WJC, Troost EGC, Boellaard R, Aerts HJWL, Gillies RJ, Lambin P. The effect of SUV discretization in quantitative FDG-PET Radiomics: the need for standardized methodology in tumor texture analysis. Sci Rep 2015; 5:11075. [PMID: 26242464 PMCID: PMC4525145 DOI: 10.1038/srep11075] [Citation(s) in RCA: 303] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 05/13/2015] [Indexed: 12/16/2022] Open
Abstract
FDG-PET-derived textural features describing intra-tumor heterogeneity are increasingly investigated as imaging biomarkers. As part of the process of quantifying heterogeneity, image intensities (SUVs) are typically resampled into a reduced number of discrete bins. We focused on the implications of the manner in which this discretization is implemented. Two methods were evaluated: (1) RD, dividing the SUV range into D equally spaced bins, where the intensity resolution (i.e. bin size) varies per image; and (2) RB, maintaining a constant intensity resolution B. Clinical feasibility was assessed on 35 lung cancer patients, imaged before and in the second week of radiotherapy. Forty-four textural features were determined for different D and B for both imaging time points. Feature values depended on the intensity resolution and out of both assessed methods, RB was shown to allow for a meaningful inter- and intra-patient comparison of feature values. Overall, patients ranked differently according to feature values–which was used as a surrogate for textural feature interpretation–between both discretization methods. Our study shows that the manner of SUV discretization has a crucial effect on the resulting textural features and the interpretation thereof, emphasizing the importance of standardized methodology in tumor texture analysis.
Collapse
Affiliation(s)
- Ralph T H Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - Georgi Nalbantov
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - Sara Carvalho
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - Wouter J C van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - Esther G C Troost
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Hugo J W L Aerts
- 1] Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands [2] Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC+), Maastricht, the Netherlands
| |
Collapse
|
39
|
Arnesen MR, Knudtsen IS, Rekstad BL, Eilertsen K, Dale E, Bruheim K, Helland Å, Løndalen AM, Hellebust TP, Malinen E. Dose painting by numbers in a standard treatment planning system using inverted dose prescription maps. Acta Oncol 2015. [PMID: 26213311 DOI: 10.3109/0284186x.2015.1061690] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Dose painting by numbers (DPBN) is a method to deliver an inhomogeneous tumor dose voxel-by-voxel with a prescription based on biological medical images. However, planning of DPBN is not supported by commercial treatment planning systems (TPS) today. Here, a straightforward method for DPBN with a standard TPS is presented. MATERIAL AND METHODS DPBN tumor dose prescription maps were generated from (18)F-FDG-PET images applying a linear relationship between image voxel value and dose. An inverted DPBN prescription map was created and imported into a standard TPS where it was defined as a mock pre-treated dose. Using inverse optimization for the summed dose, a planned DPBN dose distribution was created. The procedure was tested in standard TPS for three different tumor cases; cervix, lung and head and neck. The treatment plans were compared to the prescribed DPBN dose distribution by three-dimensional (3D) gamma analysis and quality factors (QFs). Delivery of the DPBN plans was assessed with portal dosimetry (PD). RESULTS Maximum tumor doses of 149%, 140% and 151% relative to the minimum tumor dose were prescribed for the cervix, lung and head and neck case, respectively. DPBN distributions were well achieved within the tumor whilst normal tissue doses were within constraints. Generally, high gamma pass rates (> 89% at 2%/2 mm) and low QFs (< 2.6%) were found. PD showed that all DPBN plans could be successfully delivered. CONCLUSIONS The presented methodology enables the use of currently available TPSs for DPBN planning and delivery and may therefore pave the way for clinical implementation.
Collapse
Affiliation(s)
- Marius Røthe Arnesen
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
- b Department of Physics , University of Oslo , Oslo , Norway
| | - Ingerid Skjei Knudtsen
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
- b Department of Physics , University of Oslo , Oslo , Norway
| | - Bernt Louni Rekstad
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
| | - Karsten Eilertsen
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
| | - Einar Dale
- c Department of Oncology , Oslo University Hospital , Oslo , Norway
| | - Kjersti Bruheim
- c Department of Oncology , Oslo University Hospital , Oslo , Norway
| | - Åslaug Helland
- c Department of Oncology , Oslo University Hospital , Oslo , Norway
| | - Ayca Muftuler Løndalen
- d Department of Radiology and Nuclear Medicine , Oslo University Hospital , Oslo , Norway
| | - Taran Paulsen Hellebust
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
- b Department of Physics , University of Oslo , Oslo , Norway
| | - Eirik Malinen
- a Department of Medical Physics , The Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway
- b Department of Physics , University of Oslo , Oslo , Norway
| |
Collapse
|
40
|
Toma-Dasu I, Uhrdin J, Lazzeroni M, Carvalho S, van Elmpt W, Lambin P, Dasu A. Evaluating tumor response of non-small cell lung cancer patients with ¹⁸F-fludeoxyglucose positron emission tomography: potential for treatment individualization. Int J Radiat Oncol Biol Phys 2015; 91:376-84. [PMID: 25636761 DOI: 10.1016/j.ijrobp.2014.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/19/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To assess early tumor responsiveness and the corresponding effective radiosensitivity for individual patients with non-small cell lung cancer (NSCLC) based on 2 successive (18)F-fludeoxyglucose positron emission tomography (FDG-PET) scans. METHODS AND MATERIALS Twenty-six NSCLC patients treated in Maastricht were included in the study. Fifteen patients underwent sequential chemoradiation therapy, and 11 patients received concomitant chemoradiation therapy. All patients were imaged with FDG before the start and during the second week of radiation therapy. The sequential images were analyzed in relation to the dose delivered until the second image. An operational quantity, effective radiosensitivity, αeff, was determined at the voxel level. Correlations were sought between the average αeff or the fraction of negative αeff values and the overall survival at 2 years. Separate analyses were performed for the primary gross target volume (GTV), the lymph node GTV, and the clinical target volumes (CTVs). RESULTS Patients receiving sequential treatment could be divided into responders and nonresponders, using a threshold for the average αeff of 0.003 Gy(-1) in the primary GTV, with a sensitivity of 75% and a specificity of 100% (P<.0001). Choosing the fraction of negative αeff as a criterion, the threshold 0.3 also had a sensitivity of 75% and a specificity of 100% (P<.0001). Good prognostic potential was maintained for patients receiving concurrent chemotherapy. For lymph node GTV, the correlation had low statistical significance. A cross-validation analysis confirmed the potential of the method. CONCLUSIONS Evaluation of the early response in NSCLC patients showed that it is feasible to determine a threshold value for effective radiosensitivity corresponding to good response. It also showed that a threshold value for the fraction of negative αeff could also be correlated with poor response. The proposed method, therefore, has potential to identify candidates for more aggressive strategies to increase the rate of local control and also avoid exposing to unnecessary aggressive therapies the majority of patients responding to standard treatment.
Collapse
Affiliation(s)
- Iuliana Toma-Dasu
- Medical Radiation Physics, Stockholm University and Karolinska Institutet, Stockholm, Sweden.
| | | | - Marta Lazzeroni
- Medical Radiation Physics, Karolinska Institutet, Stockholm, Sweden
| | - Sara Carvalho
- Department of Radiation Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Alexandru Dasu
- Department of Radiation Physics and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
Thorwarth D. Functional imaging for radiotherapy treatment planning: current status and future directions-a review. Br J Radiol 2015; 88:20150056. [PMID: 25827209 PMCID: PMC4628531 DOI: 10.1259/bjr.20150056] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years, radiotherapy (RT) has been subject to a number of technological innovations. Today, RT is extremely flexible, allowing irradiation of tumours with high doses, whilst also sparing normal tissues from doses. To make use of these additional degrees of freedom, integration of functional image information may play a key role (i) for better staging and tumour detection, (ii) for more accurate RT target volume delineation, (iii) to assess functional information about biological characteristics and individual radiation resistance and (iv) to apply personalized dose prescriptions. In this article, we discuss the current status and future directions of different clinically available functional imaging modalities; CT, MRI, positron emission tomography (PET) as well as the hybrid imaging techniques PET/CT and PET/MRI and their potential for individualized RT.
Collapse
Affiliation(s)
- D Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
42
|
Muren LP, Teräs M, Knuuti J. NACP 2014 and the Turku PET symposium: the interaction between therapy and imaging. Acta Oncol 2014; 53:993-6. [PMID: 25141819 DOI: 10.3109/0284186x.2014.941073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Ludvig P Muren
- Department of Medical Physics, Aarhus University and Aarhus University Hospital , Aarhus , Denmark
| | | | | |
Collapse
|