1
|
Li ZF, Zhang JN, Tian S, Sun C, Ma Y, Ye ZX. Dual-Time-Point Radiomics for Prognosis Prediction in Colorectal Liver Metastasis Treated with Neoadjuvant Therapy Before Radical Resection: A Two-Center Study. Ann Surg Oncol 2025; 32:3516-3525. [PMID: 39907877 DOI: 10.1245/s10434-025-16941-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Optimal prognostic stratification for colorectal liver metastases (CRLM) patients undergoing surgery with neoadjuvant therapy (NAT) remains elusive. This study aimed to develop and validate dual-time-point radiomic models for CRLM prognosis prediction using pre- and post-NAT imaging features. METHODS Radiomic features were extracted from four MRI sequences in 100 cases of CRLM patients who underwent NAT and radical resection. RAD scores were generated, and clinical/pathologic variables were incorporated into uni- and multivariate Cox regression analyses to construct prognosis models. Time-ROC, time-C index, decision curve analysis (DCA), and calibration curves assessed the predictive performance of Fong score and pre- and post-NAT models for overall survival (OS) and disease-free survival (DFS) in a testing set. RESULTS The final models included four variables for OS and three variables for DFS. The post-NAT models outperformed the pre-NAT models in time-ROC, time-C index, calibration, and DCA analysis, except for the 1-year DFS area under the curve (AUC). The Fong score models underperformed. The post-NAT OS RAD score effectively stratified patients into prognostic subgroups. CONCLUSIONS The radiomic models incorporating pre- and post-NAT MRI features and clinical/pathologic variables effectively stratified CRLM patients prognositically. The post-NAT models demonstrated superior performance.
Collapse
Affiliation(s)
- Zhuo-Fu Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | - Jia-Ning Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | | | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China.
| |
Collapse
|
2
|
Thor M, Lee C, Sun L, Patel P, Apte A, Grkovski M, Shepherd AF, Gelblum DY, Wu AJ, Simone CB, Chaft JE, Rimner A, Gomez DR, Deasy JO, Shaverdian N. An 18F-FDG PET/CT and Mean Lung Dose Model to Predict Early Radiation Pneumonitis in Stage III Non-Small Cell Lung Cancer Patients Treated with Chemoradiation and Immunotherapy. J Nucl Med 2024; 65:520-526. [PMID: 38485270 PMCID: PMC10995528 DOI: 10.2967/jnumed.123.266965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/11/2024] [Indexed: 04/04/2024] Open
Abstract
Radiation pneumonitis (RP) that develops early (i.e., within 3 mo) (RPEarly) after completion of concurrent chemoradiation (cCRT) leads to treatment discontinuation and poorer survival for patients with stage III non-small cell lung cancer. Since no RPEarly risk model exists, we explored whether published RP models and pretreatment 18F-FDG PET/CT-derived features predict RPEarly Methods: One hundred sixty patients with stage III non-small cell lung cancer treated with cCRT and consolidative immunotherapy were analyzed for RPEarly Three published RP models that included the mean lung dose (MLD) and patient characteristics were examined. Pretreatment 18F-FDG PET/CT normal-lung SUV featured included the following: 10th percentile of SUV (SUVP10), 90th percentile of SUV (SUVP90), SUVmax, SUVmean, minimum SUV, and SD. Associations between models/features and RPEarly were assessed using area under the receiver-operating characteristic curve (AUC), P values, and the Hosmer-Lemeshow test (pHL). The cohort was randomly split, with similar RPEarly rates, into a 70%/30% derivation/internal validation subset. Results: Twenty (13%) patients developed RPEarly Predictors for RPEarly were MLD alone (AUC, 0.72; P = 0.02; pHL, 0.87), SUVP10, SUVP90, and SUVmean (AUC, 0.70-0.74; P = 0.003-0.006; pHL, 0.67-0.70). The combined MLD and SUVP90 model generalized in the validation subset and was deemed the final RPEarly model (RPEarly risk = 1/[1+e(- x )]; x = -6.08 + [0.17 × MLD] + [1.63 × SUVP90]). The final model refitted in the 160 patients indicated improvement over the published MLD-alone model (AUC, 0.77 vs. 0.72; P = 0.0001 vs. 0.02; pHL, 0.65 vs. 0.87). Conclusion: Patients at risk for RPEarly can be detected with high certainty by combining the normal lung's MLD and pretreatment 18F-FDG PET/CT SUVP90 This refined model can be used to identify patients at an elevated risk for premature immunotherapy discontinuation due to RPEarly and could allow for interventions to improve treatment outcomes.
Collapse
Affiliation(s)
- Maria Thor
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Chen Lee
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lian Sun
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Purvi Patel
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aditya Apte
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Annemarie F Shepherd
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Daphna Y Gelblum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Charles B Simone
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jamie E Chaft
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Narek Shaverdian
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| |
Collapse
|
3
|
Ruan D, Fang J, Teng X. Efficient 18F-fluorodeoxyglucose positron emission tomography/computed tomography-based machine learning model for predicting epidermal growth factor receptor mutations in non-small cell lung cancer. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2024; 68:70-83. [PMID: 35420272 DOI: 10.23736/s1824-4785.22.03441-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Beyond the human eye's limitations, radiomics provides more information that can be used for diagnosis. We develop a personalized and efficient model based on 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) to predict epidermal growth factor receptor (EGFR) mutations to help identify which non-small cell cancer (NSCLC) patients are candidates for EGFR-tyrosine kinase inhibitors (TKIs) therapy. METHODS We retrospectively included 100 patients with NSCLC and randomized them according to 70 patients in the training group and 30 patients in the validation group. The least absolute shrinkage and selection operator logistic regression (LLR) algorithm and support vector machine (SVM) classifier were used to build the models and predict whether EGFR is mutated or not. The predictive efficacy of the LLR algorithm-based model and the SVM classifier-based model was evaluated by plotting the receiver operating characteristic (ROC) curves and calculating the area under the curve (AUC). RESULTS The AUC, sensitivity and specificity of our radiomics model by LLR algorithm were 0.792, 0.967, and 0.600 for the training group and 0.643, 1.00, and 0.378 for the validation group, respectively, in predicting EGFR mutations. The AUC was 0.838 for the training group and 0.696 for the validation group after combining radiomics features with clinical features. The prediction results based on the SVM classifier showed that the validation group had the best performance when based on radial kernel function with AUC, sensitivity, and specificity of 0.741, 0.667, and 0.825, respectively. CONCLUSIONS Radiomics models based on 18F-FDG PET/CT modeled with different machine learning algorithms can improve the predictive efficacy of the models. Models that combine clinical features are more clinically valuable.
Collapse
Affiliation(s)
- Dan Ruan
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China -
| | - Janyao Fang
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| | - Xinyu Teng
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| |
Collapse
|
4
|
Thor M, Fitzgerald K, Apte A, Oh JH, Iyer A, Odiase O, Nadeem S, Yorke ED, Chaft J, Wu AJ, Offin M, Simone Ii CB, Preeshagul I, Gelblum DY, Gomez D, Deasy JO, Rimner A. Exploring published and novel pre-treatment CT and PET radiomics to stratify risk of progression among early-stage non-small cell lung cancer patients treated with stereotactic radiation. Radiother Oncol 2024; 190:109983. [PMID: 37926331 PMCID: PMC11233189 DOI: 10.1016/j.radonc.2023.109983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
PURPOSE Disease progression after definitive stereotactic body radiation therapy (SBRT) for early-stage non-small cell lung cancer (NSCLC) occurs in 20-40% of patients. Here, we explored published and novel pre-treatment CT and PET radiomics features to identify patients at risk of progression. MATERIALS/METHODS Published CT and PET features were identified and explored along with 15 other CT and PET features in 408 consecutively treated early-stage NSCLC patients having CT and PET < 3 months pre-SBRT (training/set-aside validation subsets: n = 286/122). Features were associated with progression-free survival (PFS) using bootstrapped Cox regression (Bonferroni-corrected univariate predictor: p ≤ 0.002) and only non-strongly correlated predictors were retained (|Rs|<0.70) in forward-stepwise multivariate analysis. RESULTS Tumor diameter and SUVmax were the two most frequently reported features associated with progression/survival (in 6/20 and 10/20 identified studies). These two features and 12 of the 15 additional features (CT: 6; PET: 6) were candidate PFS predictors. A re-fitted model including diameter and SUVmax presented with the best performance (c-index: 0.78; log-rank p-value < 0.0001). A model built with the two best additional features (CTspiculation1 and SUVentropy) had a c-index of 0.75 (log-rank p-value < 0.0001). CONCLUSIONS A re-fitted pre-treatment model using the two most frequently published features - tumor diameter and SUVmax - successfully stratified early-stage NSCLC patients by PFS after receiving SBRT.
Collapse
Affiliation(s)
- Maria Thor
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA.
| | - Kelly Fitzgerald
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | - Aditya Apte
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Aditi Iyer
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Otasowie Odiase
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | - Saad Nadeem
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Ellen D Yorke
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Jamie Chaft
- Department of Medicine, Memorial Sloan Kettering Cancer Center, USA
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, USA
| | - Charles B Simone Ii
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | | | - Daphna Y Gelblum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, USA
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, USA
| |
Collapse
|
5
|
Chen K, Hou L, Chen M, Li S, Shi Y, Raynor WY, Yang H. Predicting the Efficacy of SBRT for Lung Cancer with 18F-FDG PET/CT Radiogenomics. Life (Basel) 2023; 13:life13040884. [PMID: 37109413 PMCID: PMC10142286 DOI: 10.3390/life13040884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Purpose: to develop a radiogenomic model on the basis of 18F-FDG PET/CT radiomics and clinical-parameter EGFR for predicting PFS stratification in lung-cancer patients after SBRT treatment. Methods: A total of 123 patients with lung cancer who had undergone 18F-FDG PET/CT examination before SBRT from September 2014 to December 2021 were retrospectively analyzed. All patients’ PET/CT images were manually segmented, and the radiomic features were extracted. LASSO regression was used to select radiomic features. Logistic regression analysis was used to screen clinical features to establish the clinical EGFR model, and a radiogenomic model was constructed by combining radiomics and clinical EGFR. We used the receiver operating characteristic curve and calibration curve to assess the efficacy of the models. The decision curve and influence curve analysis were used to evaluate the clinical value of the models. The bootstrap method was used to validate the radiogenomic model, and the mean AUC was calculated to assess the model. Results: A total of 2042 radiomics features were extracted. Five radiomic features were related to the PFS stratification of lung-cancer patients with SBRT. T-stage and overall stages (TNM) were independent factors for predicting PFS stratification. AUCs under the ROC curve of the radiomics, clinical EGFR, and radiogenomic models were 0.84, 0.67, and 0.86, respectively. The calibration curve shows that the predicted value of the radiogenomic model was in good agreement with the actual value. The decision and influence curve showed that the model had high clinical application values. After Bootstrap validation, the mean AUC of the radiogenomic model was 0.850(95%CI 0.849–0.851). Conclusions: The radiogenomic model based on 18F-FDG PET/CT radiomics and clinical EGFR has good application value in predicting the PFS stratification of lung-cancer patients after SBRT treatment.
Collapse
Affiliation(s)
- Kuifei Chen
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Liqiao Hou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Meng Chen
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Shuling Li
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Yangyang Shi
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85724, USA
| | - William Y. Raynor
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Haihua Yang
- Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 317000, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou 317000, China
- Correspondence: or
| |
Collapse
|
6
|
Huang Y, Jiang X, Xu H, Zhang D, Liu LN, Xia YX, Xu DK, Wu HJ, Cheng G, Shi YH. Preoperative prediction of mediastinal lymph node metastasis in non-small cell lung cancer based on 18F-FDG PET/CT radiomics. Clin Radiol 2023; 78:8-17. [PMID: 36192203 DOI: 10.1016/j.crad.2022.08.140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/14/2022] [Accepted: 08/26/2022] [Indexed: 01/07/2023]
Abstract
AIM To establish and verify a 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT)-based radiomics nomogram to predict mediastinal lymph node metastasis (LNM) in non-small cell lung cancer (NSCLC) patients preoperatively. MATERIALS AND METHODS This retrospective study enrolled 155 NSCLC patients (primary cohort, n=93; validation cohort, n=62). For each patient, 2,704 radiomic features were extracted from the primary lung cancer regions. Four procedures including the Mann-Whitney U-test, Spearman's correlation analysis, minimum redundancy-maximum relevance (mRMR), and least absolute shrinkage and selection operator (LASSO) binary logistic regression were utilised for determining essential features and establishing a radiomics signature. After that, a nomogram was established. The nomogram's potential was assessed based on its discrimination, calibration, and clinical usefulness. The radiomics signature and nomogram predictive performances were evaluated with respect to the area under the receiver operating characteristic curve (AUC), specificity, accuracy, and sensitivity. RESULTS The radiomics signature composed of eight selected features had good discriminatory performance of LNM versus non-LNM groups an AUC of 0.851 and 0.826 in primary and validation cohorts, respectively. The nomogram also indicated good discrimination with an AUC of 0.869 and 0.847 in the primary and validation cohorts, respectively. Furthermore, good calibration was demonstrated utilising the nomogram. CONCLUSIONS An 18F-FDG PET/CT-based radiomics nomogram that integrates the radiomics signature and age was promoted to predict mediastinal LNM within NSCLC patients, which could potentially facilitate individualised therapy for mediastinal LNM before treatment. The nomogram was beneficial in clinical practice, as illustrated by decision curve analysis.
Collapse
Affiliation(s)
- Y Huang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - X Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - H Xu
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - D Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - L-N Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Y-X Xia
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - D-K Xu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - H-J Wu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - G Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Y-H Shi
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Akamatsu G, Tsutsui Y, Daisaki H, Mitsumoto K, Baba S, Sasaki M. A review of harmonization strategies for quantitative PET. Ann Nucl Med 2023; 37:71-88. [PMID: 36607466 PMCID: PMC9902332 DOI: 10.1007/s12149-022-01820-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
PET can reveal in vivo biological processes at the molecular level. PET-derived quantitative values have been used as a surrogate marker for clinical decision-making in numerous clinical studies and trials. However, quantitative values in PET are variable depending on technical, biological, and physical factors. The variability may have a significant impact on a study outcome. Appropriate scanner calibration and quality control, standardization of imaging protocols, and any necessary harmonization strategies are essential to make use of PET as a biomarker with low bias and variability. This review summarizes benefits, limitations, and remaining challenges for harmonization of quantitative PET, including whole-body PET in oncology, brain PET in neurology, PET/MR, and non-18F PET imaging. This review is expected to facilitate harmonization of quantitative PET and to promote the contribution of PET-derived biomarkers to research and development in medicine.
Collapse
Affiliation(s)
- Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan. .,Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Yuji Tsutsui
- Department of Radiological Science, Faculty of Health Science, Junshin Gakuen University, 1-1-1 Chikushigaoka, Minami-ku, Fukuoka, 815-8510 Japan
| | - Hiromitsu Daisaki
- Department of Radiological Technology, Gunma Prefectural College of Health Sciences, 323-1 Kamioki-machi, Maebashi, Gunma 371-0052 Japan
| | - Katsuhiko Mitsumoto
- Department of Clinical Radiology Service, Kyoto University Hospital, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Shingo Baba
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Masayuki Sasaki
- Department of Medical Quantum Science, Faculty of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| |
Collapse
|
8
|
Aide N, Weyts K, Lasnon C. Prediction of the Presence of Targetable Molecular Alteration(s) with Clinico-Metabolic 18 F-FDG PET Radiomics in Non-Asian Lung Adenocarcinoma Patients. Diagnostics (Basel) 2022; 12:diagnostics12102448. [PMID: 36292136 PMCID: PMC9601118 DOI: 10.3390/diagnostics12102448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate if combining clinical characteristics with pre-therapeutic 18 F-fluorodeoxyglucose (18 F-FDG) positron emission tomography (PET) radiomics could predict the presence of molecular alteration(s) in key molecular targets in lung adenocarcinoma. This non-interventional monocentric study included patients with newly diagnosed lung adenocarcinoma referred for baseline PET who had tumour molecular analyses. The data were randomly split into training and test datasets. LASSO regression with 100-fold cross-validation was performed, including sex, age, smoking history, AJCC cancer stage and 31 PET variables. In total, 109 patients were analysed, and it was found that 63 (57.8%) patients had at least one molecular alteration. Using the training dataset (n = 87), the model included 10 variables, namely age, sex, smoking history, AJCC stage, excessKustosis_HISTO, sphericity_SHAPE, variance_GLCM, correlation_GLCM, LZE_GLZLM, and GLNU_GLZLM. The ROC analysis for molecular alteration prediction using this model found an AUC equal to 0.866 (p < 0.0001). A cut-off value set to 0.48 led to a sensitivity of 90.6% and a positive likelihood ratio (LR+) value equal to 2.4. After application of this cut-off value in the unseen test dataset of patients (n = 22), the test presented a sensitivity equal to 90.0% and an LR+ value of 1.35. A clinico-metabolic 18 F-FDG PET phenotype allows the detection of key molecular target alterations with high sensitivity and negative predictive value. Hence, it opens the way to the selection of patients for molecular analysis.
Collapse
Affiliation(s)
- Nicolas Aide
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, 14000 Caen, France
| | - Kathleen Weyts
- Nuclear Medicine Department, Comprehensive Cancer Centre F. Baclesse, UNICANCER, 14000 Caen, France
| | - Charline Lasnon
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, 14000 Caen, France
- Nuclear Medicine Department, Comprehensive Cancer Centre F. Baclesse, UNICANCER, 14000 Caen, France
- Correspondence: ; Tel.: +33-261-455-268; Fax: +33-231-455-101
| |
Collapse
|
9
|
Impact of feature harmonization on radiogenomics analysis: Prediction of EGFR and KRAS mutations from non-small cell lung cancer PET/CT images. Comput Biol Med 2022; 142:105230. [DOI: 10.1016/j.compbiomed.2022.105230] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
|
10
|
Delgado Bolton RC, Aide N, Colletti PM, Ferrero A, Paez D, Skanjeti A, Giammarile F. EANM guideline on the role of 2-[ 18F]FDG PET/CT in diagnosis, staging, prognostic value, therapy assessment and restaging of ovarian cancer, endorsed by the American College of Nuclear Medicine (ACNM), the Society of Nuclear Medicine and Molecular Imaging (SNMMI) and the International Atomic Energy Agency (IAEA). Eur J Nucl Med Mol Imaging 2021; 48:3286-3302. [PMID: 34215923 DOI: 10.1007/s00259-021-05450-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/03/2021] [Indexed: 10/20/2022]
Abstract
In most patients with ovarian carcinoma, the diagnosis is reached when the disease is long past the initial stages, presenting already an advanced stage, and they usually have a very bad prognosis. Cytoreductive or debulking surgical procedures, platinum-based chemotherapy and targeted agents are key therapeutic elements. However, around 7 out of 10 patients present recurrent disease within 36 months from the initial diagnosis. The metastatic spread in ovarian cancer follows three pathways: contiguous dissemination across the peritoneum, dissemination through the lymphatic drainage and, although less importantly in this case, through the bloodstream. Radiological imaging, including ultrasound, CT and MRI, are the main imaging techniques in which management decisions are supported, CT being considered the best available technique for presurgical evaluation and staging purposes. Regarding 2-[18F]FDG PET/CT, the evidence available in the literature demonstrates efficacy in primary detection, disease staging and establishing the prognosis and especially for relapse detection. There is limited evidence when considering the evaluation of therapeutic response. This guideline summarizes the level of evidence and grade of recommendation for the clinical indications of 2-[18F]FDG PET/CT in each disease stage of ovarian carcinoma.
Collapse
Affiliation(s)
- Roberto C Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), La Rioja, Logroño, Spain.
| | - Nicolas Aide
- Department of Nuclear Medicine, Caen University Hospital, Caen, France.,INSERM U1086 ANTICIPE, Normandie Université, Caen, France
| | - Patrick M Colletti
- Department of Radiology, University of Southern California, Los Angeles, CA, USA
| | - Annamaria Ferrero
- Academic Division Gynaecology and Obstetrics, University of Torino, Mauriziano Hospital, Torino, Italy
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Andrea Skanjeti
- Department of Nuclear Medicine, Hospices Civils de Lyon, Université Claude Bernard, Lyon 1, Lyon, France
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, International Atomic Energy Agency (IAEA), Vienna, Austria.,Department of Nuclear Medicine, Centre Léon Bérard, Lyon, France
| |
Collapse
|
11
|
A Systematic Review of PET Textural Analysis and Radiomics in Cancer. Diagnostics (Basel) 2021; 11:diagnostics11020380. [PMID: 33672285 PMCID: PMC7926413 DOI: 10.3390/diagnostics11020380] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although many works have supported the utility of PET radiomics, several authors have raised concerns over the robustness and replicability of the results. This study aimed to perform a systematic review on the topic of PET radiomics and the used methodologies. Methods: PubMed was searched up to 15 October 2020. Original research articles based on human data specifying at least one tumor type and PET image were included, excluding those that apply only first-order statistics and those including fewer than 20 patients. Each publication, cancer type, objective and several methodological parameters (number of patients and features, validation approach, among other things) were extracted. Results: A total of 290 studies were included. Lung (28%) and head and neck (24%) were the most studied cancers. The most common objective was prognosis/treatment response (46%), followed by diagnosis/staging (21%), tumor characterization (18%) and technical evaluations (15%). The average number of patients included was 114 (median = 71; range 20–1419), and the average number of high-order features calculated per study was 31 (median = 26, range 1–286). Conclusions: PET radiomics is a promising field, but the number of patients in most publications is insufficient, and very few papers perform in-depth validations. The role of standardization initiatives will be crucial in the upcoming years.
Collapse
|
12
|
Abstract
Carrying out large multicenter studies is one of the key goals to be achieved towards a faster transfer of the radiomics approach in the clinical setting. This requires large-scale radiomics data analysis, hence the need for integrating radiomic features extracted from images acquired in different centers. This is challenging as radiomic features exhibit variable sensitivity to differences in scanner model, acquisition protocols and reconstruction settings, which is similar to the so-called 'batch-effects' in genomics studies. In this review we discuss existing methods to perform data integration with the aid of reducing the unwanted variation associated with batch effects. We also discuss the future potential role of deep learning methods in providing solutions for addressing radiomic multicentre studies.
Collapse
Affiliation(s)
- R Da-Ano
- LaTiM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - D Visvikis
- LaTiM, INSERM, UMR 1101, Univ Brest, Brest, France
- equally contributed
| | - M Hatt
- LaTiM, INSERM, UMR 1101, Univ Brest, Brest, France
- equally contributed
| |
Collapse
|
13
|
van Timmeren JE, Cester D, Tanadini-Lang S, Alkadhi H, Baessler B. Radiomics in medical imaging-"how-to" guide and critical reflection. Insights Imaging 2020; 11:91. [PMID: 32785796 PMCID: PMC7423816 DOI: 10.1186/s13244-020-00887-2] [Citation(s) in RCA: 723] [Impact Index Per Article: 144.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Radiomics is a quantitative approach to medical imaging, which aims at enhancing the existing data available to clinicians by means of advanced mathematical analysis. Through mathematical extraction of the spatial distribution of signal intensities and pixel interrelationships, radiomics quantifies textural information by using analysis methods from the field of artificial intelligence. Various studies from different fields in imaging have been published so far, highlighting the potential of radiomics to enhance clinical decision-making. However, the field faces several important challenges, which are mainly caused by the various technical factors influencing the extracted radiomic features.The aim of the present review is twofold: first, we present the typical workflow of a radiomics analysis and deliver a practical "how-to" guide for a typical radiomics analysis. Second, we discuss the current limitations of radiomics, suggest potential improvements, and summarize relevant literature on the subject.
Collapse
Affiliation(s)
- Janita E van Timmeren
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Davide Cester
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Hatem Alkadhi
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Bettina Baessler
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
14
|
Shiri I, Hajianfar G, Sohrabi A, Abdollahi H, P Shayesteh S, Geramifar P, Zaidi H, Oveisi M, Rahmim A. Repeatability of radiomic features in magnetic resonance imaging of glioblastoma: Test-retest and image registration analyses. Med Phys 2020; 47:4265-4280. [PMID: 32615647 DOI: 10.1002/mp.14368] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To assess the repeatability of radiomic features in magnetic resonance (MR) imaging of glioblastoma (GBM) tumors with respect to test-retest, different image registration approaches and inhomogeneity bias field correction. METHODS We analyzed MR images of 17 GBM patients including T1- and T2-weighted images (performed within the same imaging unit on two consecutive days). For image segmentation, we used a comprehensive segmentation approach including entire tumor, active area of tumor, necrotic regions in T1-weighted images, and edema regions in T2-weighted images (test studies only; registration to retest studies is discussed next). Analysis included N3, N4 as well as no bias correction performed on raw MR images. We evaluated 20 image registration approaches, generated by cross-combination of four transformation and five cost function methods. In total, 714 images (17 patients × 2 images × ((4 transformations × 5 cost functions) + 1 test image) and 2856 segmentations (714 images × 4 segmentations) were prepared for feature extraction. Various radiomic features were extracted, including the use of preprocessing filters, specifically wavelet (WAV) and Laplacian of Gaussian (LOG), as well as discretization into fixed bin width and fixed bin count (16, 32, 64, 128, and 256), Exponential, Gradient, Logarithm, Square and Square Root scales. Intraclass correlation coefficients (ICC) were calculated to assess the repeatability of MRI radiomic features (high repeatability defined as ICC ≥ 95%). RESULTS In our ICC results, we observed high repeatability (ICC ≥ 95%) with respect to image preprocessing, different image registration algorithms, and test-retest analysis, for example: RLNU and GLNU from GLRLM, GLNU and DNU from GLDM, Coarseness and Busyness from NGTDM, GLNU and ZP from GLSZM, and Energy and RMS from first order. Highest fraction (percent) of repeatable features was observed, among registration techniques, for the method Full Affine transformation with 12 degrees of freedom using Mutual Information cost function (mean 32.4%), and among image processing methods, for the method Laplacian of Gaussian (LOG) with Sigma (2.5-4.5 mm) (mean 78.9%). The trends were relatively consistent for N4, N3, or no bias correction. CONCLUSION Our results showed varying performances in repeatability of MR radiomic features for GBM tumors due to test-retest and image registration. The findings have implications for appropriate usage in diagnostic and predictive models.
Collapse
Affiliation(s)
- Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, CH-1211, Switzerland
| | - Ghasem Hajianfar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Ahmad Sohrabi
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Abdollahi
- Department of Radiologic Sciences and Medical Physics, Faculty of Allied Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Sajad P Shayesteh
- Department of Physiology, Pharmacology and Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, CH-1211, Switzerland.,Geneva University Neurocenter, Geneva University, Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Mehrdad Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran.,Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Arman Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada.,Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| |
Collapse
|
15
|
Dercle L, Henry T, Carré A, Paragios N, Deutsch E, Robert C. Reinventing radiation therapy with machine learning and imaging bio-markers (radiomics): State-of-the-art, challenges and perspectives. Methods 2020; 188:44-60. [PMID: 32697964 DOI: 10.1016/j.ymeth.2020.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy is a pivotal cancer treatment that has significantly progressed over the last decade due to numerous technological breakthroughs. Imaging is now playing a critical role on deployment of the clinical workflow, both for treatment planning and treatment delivery. Machine-learning analysis of predefined features extracted from medical images, i.e. radiomics, has emerged as a promising clinical tool for a wide range of clinical problems addressing drug development, clinical diagnosis, treatment selection and implementation as well as prognosis. Radiomics denotes a paradigm shift redefining medical images as a quantitative asset for data-driven precision medicine. The adoption of machine-learning in a clinical setting and in particular of radiomics features requires the selection of robust, representative and clinically interpretable biomarkers that are properly evaluated on a representative clinical data set. To be clinically relevant, radiomics must not only improve patients' management with great accuracy but also be reproducible and generalizable. Hence, this review explores the existing literature and exposes its potential technical caveats, such as the lack of quality control, standardization, sufficient sample size, type of data collection, and external validation. Based upon the analysis of 165 original research studies based on PET, CT-scan, and MRI, this review provides an overview of new concepts, and hypotheses generating findings that should be validated. In particular, it describes evolving research trends to enhance several clinical tasks such as prognostication, treatment planning, response assessment, prediction of recurrence/relapse, and prediction of toxicity. Perspectives regarding the implementation of an AI-based radiotherapy workflow are presented.
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, USA
| | - Theophraste Henry
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alexandre Carré
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Eric Deutsch
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charlotte Robert
- Molecular Radiotherapy and Innovative Therapeutics, INSERM UMR1030, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France; Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
16
|
Lowe G, Spottiswoode B, Declerck J, Sullivan K, Sharif MS, Wong WL, Sanghera B. Positron emission tomography PET/CT harmonisation study of different clinical PET/CT scanners using commercially available software. BJR Open 2020; 2:20190035. [PMID: 33178963 PMCID: PMC7594895 DOI: 10.1259/bjro.20190035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 11/06/2022] Open
Abstract
Objectives: Harmonisation is the process whereby standardised uptake values from different scanners can be made comparable. This PET/CT pilot study aimed to evaluate the effectiveness of harmonisation of a modern scanner with image reconstruction incorporating resolution recovery (RR) with another vendor older scanner operated in two-dimensional (2D) mode, and for both against a European standard (EARL). The vendor-proprietary software EQ•PET was used, which achieves harmonisation with a Gaussian smoothing. A substudy investigated effect of RR on harmonisation. Methods: Phantom studies on each scanner were performed to optimise the smoothing parameters required to achieve successful harmonisation. 80 patients were retrospectively selected; half were imaged on each scanner. As proof of principle, a cohort of 10 patients was selected from the modern scanner subjects to study the effects of RR on harmonisation. Results: Before harmonisation, the modern scanner without RR adhered to EARL specification. Using the phantom data, filters were derived for optimal harmonisation between scanners and with and without RR as applicable, to the EARL standard. The 80-patient cohort did not reveal any statistically significant differences. In the 10-patient cohort SUVmax for RR > no RR irrespective of harmonisation but differences lacked statistical significance (one-way ANOVA F(3.36) = 0.37, p = 0.78). Bland-Altman analysis showed that harmonisation reduced the SUVmax ratio between RR and no RR to 1.07 (95% CI 0.96–1.18) with no outliers. Conclusions: EQ•PET successfully enabled harmonisation between modern and older scanners and against the EARL standard. Harmonisation reduces SUVmax and dependence on the use of RR in the modern scanner. Advances in knowledge: EQ•PET is feasible to harmonise different PET/CT scanners and reduces the effect of RR on SUVmax.
Collapse
Affiliation(s)
- Gerry Lowe
- Cancer Centre, Mount Vernon Hospital, London, UK
| | | | | | - Keith Sullivan
- Health Research Methods Unit, University of Hertfordshire, Hertfordshire, UK
| | - Mhd Saeed Sharif
- School of Arch., Comp. and Eng., University of East London, London, UK
| | - Wai-Lup Wong
- Paul Strickland Scanner Centre, Mount Vernon Hospital, London, UK
| | - Bal Sanghera
- Paul Strickland Scanner Centre, Mount Vernon Hospital, London, UK
| |
Collapse
|
17
|
Liu Q, Sun D, Li N, Kim J, Feng D, Huang G, Wang L, Song S. Predicting EGFR mutation subtypes in lung adenocarcinoma using 18F-FDG PET/CT radiomic features. Transl Lung Cancer Res 2020; 9:549-562. [PMID: 32676319 PMCID: PMC7354146 DOI: 10.21037/tlcr.2020.04.17] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Identification of epidermal growth factor receptor (EGFR) mutation types is crucial before tyrosine kinase inhibitors (TKIs) treatment. Radiomics is a new strategy to noninvasively predict the genetic status of cancer. In this study, we aimed to develop a predictive model based on 18F-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) radiomic features to identify the specific EGFR mutation subtypes. Methods We retrospectively studied 18F-FDG PET/CT images of 148 patients with isolated lung lesions, which were scanned in two hospitals with different CT scan setting (slice thickness: 3 and 5 mm, respectively). The tumor regions were manually segmented on PET/CT images, and 1,570 radiomic features (1,470 from CT and 100 from PET) were extracted from the tumor regions. Seven hundred and ninety-four radiomic features insensitive to different CT settings were first selected using the Mann white U test, and collinear features were further removed from them by recursively calculating the variation inflation factor. Then, multiple supervised machine learning models were applied to identify prognostic radiomic features through: (I) a multi-variate random forest to select features of high importance in discriminating different EGFR mutation status; (II) a logistic regression model to select features of the highest predictive value of the EGFR subtypes. The EGFR mutation predicting model was constructed from prognostic radiomic features using the popular Xgboost machine-learning algorithm and validated using 3-fold cross-validation. The performance of predicting model was analyzed using the receiver operating characteristic curve (ROC) and measured with the area under the curve (AUC). Results Two sets of prognostic radiomic features were found for specific EGFR mutation subtypes: 5 radiomic features for EGFR exon 19 deletions, and 5 radiomic features for EGFR exon 21 L858R missense. The corresponding radiomic predictors achieved the prediction accuracies of 0.77 and 0.92 in terms of AUC, respectively. Combing these two predictors, the overall model for predicting EGFR mutation positivity was also constructed, and the AUC was 0.87. Conclusions In our study, we established predictive models based on radiomic analysis of 18F-FDG PET/CT images. And it achieved a satisfying prediction power in the identification of EGFR mutation status as well as the certain EGFR mutation subtypes in lung cancer.
Collapse
Affiliation(s)
- Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dazhen Sun
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Automation, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Nan Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jinman Kim
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Dagan Feng
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Gang Huang
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lisheng Wang
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Automation, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
18
|
Prigent K, Aide N. 18F-Fludeoxyglucose PET/Computed Tomography for Assessing Tumor Response to Immunotherapy and Detecting Immune-Related Side Effects: A Checklist for the PET Reader. PET Clin 2020; 15:1-10. [PMID: 31735296 DOI: 10.1016/j.cpet.2019.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
After a short summary of the biological basis of the immune checkpoint inhibitors used for the treatment of nonhematologic solid tumors, the issues of pseudoprogression, hyperprogression, and immune-related side effects are discussed as well as their implications for patient management. Recommendations are provided for performing 18F-Fludeoxyglucose PET scanning, assessing tumor response, and reporting immune-related side effects, with representative clinical cases.
Collapse
Affiliation(s)
- Kevin Prigent
- Nuclear Medicine Department, University Hospital, Caen, France; Service de Médecine Nucléaire, CHU de CAEN, Avenue Côte de Nacre, Caen, France
| | - Nicolas Aide
- Service de Médecine Nucléaire, CHU de CAEN, Avenue Côte de Nacre, Caen, France; University of Normandy, France.
| |
Collapse
|
19
|
Baseline 18F-FDG PET radiomic features as predictors of 2-year event-free survival in diffuse large B cell lymphomas treated with immunochemotherapy. Eur Radiol 2020; 30:4623-4632. [PMID: 32248365 DOI: 10.1007/s00330-020-06815-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVES To explore the prognostic value of positron emission tomography (PET) radiomic features in the field of diffuse large B cell lymphoma (DLBCL) treated with a first-line immunochemotherapy. METHODS One-hundred thirty-two patients newly diagnosed with DLBCL were retrospectively included. PET studies were reconstructed using an ordered subset expectation maximisation algorithm with point spread function modelling. The total metabolic tumour volume (MTV) was recorded for each patient, and the volume of interest structure of the largest target lesion was used to compute 18F-FDG textural parameters. Data was randomly split into training and validation datasets. Optimal cutoff values were determined by means of 2-year event-free survival (EFS) ROC analyses. Two-year EFS analyses were performed using Kaplan-Meier survival analyses and univariable and multivariable Cox regression models. RESULTS The median follow-up was 27 months, and the 2-year event-free survival (2y-EFS) was 77.3% in the entire population. ROC analyses for the 2y-EFS reached statistical significance for total MTV as well as four second-order metrics (homogeneity, contrast, correlation, dissimilarity) and five third-order metrics (LZE (Long-Zone Emphasis), LZLGE (Long-Zone Low-Grey Level Emphasis), LZHGE (Long-Zone High-Grey Level Emphasis), GLNU (Grey-Level Non-Uniformity) and ZP (Zone Percentage)). LZHGE displayed the highest ROC analysis accuracy (acc. = 0.76) and the best discriminant value on univariable Kaplan-Meier analysis (p < 0.0001, HR = 4.54). On multivariable analysis, including IPIaa, total MTV and LZHGE, LZHGE was the only independent predictor of 2y-EFS. These results were confirmed on the validation dataset. CONCLUSIONS Baseline 18F-FDG PET heterogeneity of the largest lymphoma lesion is a promising predictor of 2y-EFS in newly diagnosed DLBCL treated with immunochemotherapy. KEY POINTS •18F-FDG metabolic heterogeneity emerges as a new tool for survival prognostication of patients and has been explored in many solid tumours with promising results. • Baseline18F-FDG PET heterogeneity of the largest lymphoma lesion is an independent predictor of 2y-EFS in newly diagnosed DLBCL treated with immunochemotherapy. • DLBCL patients presenting with a heterogeneous tumour displayed a worse prognosis.
Collapse
|
20
|
Vaugier L, Ferrer L, Mengue L, Jouglar E. Radiomics for radiation oncologists: are we ready to go? BJR Open 2020; 2:20190046. [PMID: 33178967 PMCID: PMC7594896 DOI: 10.1259/bjro.20190046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Radiomics have emerged as an exciting field of research over the past few years, with very wide potential applications in personalised and precision medicine of the future. Radiomics-based approaches are still however limited in daily clinical practice in oncology. This review focus on how radiomics could be incorporated into the radiation therapy pipeline, and globally help the radiation oncologist, from the tumour diagnosis to follow-up after treatment. Radiomics could impact on all steps of the treatment pipeline, once the limitations in terms of robustness and reproducibility are overcome. Major ongoing efforts should be made to collect and share data in the most standardised manner possible.
Collapse
Affiliation(s)
- Loïg Vaugier
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Ludovic Ferrer
- Department of Medical Physics, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Laurence Mengue
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Emmanuel Jouglar
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| |
Collapse
|
21
|
Krarup MMK, Nygård L, Vogelius IR, Andersen FL, Cook G, Goh V, Fischer BM. Heterogeneity in tumours: Validating the use of radiomic features on 18F-FDG PET/CT scans of lung cancer patients as a prognostic tool. Radiother Oncol 2020; 144:72-78. [PMID: 31733491 DOI: 10.1016/j.radonc.2019.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/01/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
AIM The aim was to validate promising radiomic features (RFs)1 on 18F-flourodeoxyglucose positron emission tomography/computed tomography-scans (18F-FDG PET/CT) of non-small cell lung cancer (NSCLC) patients undergoing definitive chemo-radiotherapy. METHODS 18F-FDG PET/CT scans performed for radiotherapy (RT) planning were retrieved. Auto-segmentation with visual adaption was used to define the primary tumour on PET images. Six pre-selected prognostic and reproducible PET texture -and shape-features were calculated using texture respectively shape analysis. The correlation between these RFs and metabolic active tumour volume (MTV)3, gross tumour volume (GTV)4 and maximum and mean of standardized uptake value (SUV)5 was tested with a Spearman's Rank test. The prognostic value of RFs was tested in a univariate cox regression analysis and a multivariate cox regression analysis with GTV, clinical stage and histology. P-value ≤ 0.05 were considered significant. RESULTS Image analysis was performed for 233 patients: 145 males and 88 females, mean age of 65.7 and clinical stage II-IV. Mean GTV was 129.87 cm3 (SD 130.30 cm3). Texture and shape-features correlated more strongly to MTV and GTV compared to SUV-measurements. Four RFs predicted PFS in the univariate analysis. No RFs predicted PFS in the multivariate analysis, whereas GTV and clinical stage predicted PFS (p = 0.001 and p = 0.008 respectively). CONCLUSION The pre-selected RFs were insignificant in predicting PFS in combination with GTV, clinical stage and histology. These results might be due to variations in technical parameters. However, it is relevant to question whether RFs are stable enough to provide clinically useful information.
Collapse
Affiliation(s)
- Marie Manon Krebs Krarup
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Lotte Nygård
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Ivan Richter Vogelius
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark; Faculty of Health and Medical Sciences, Copenhagen University, Denmark.
| | - Flemming Littrup Andersen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Gary Cook
- PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom.
| | - Vicky Goh
- PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom.
| | - Barbara Malene Fischer
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen University Hospital, Denmark; PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom.
| |
Collapse
|
22
|
Mayerhoefer ME, Materka A, Langs G, Häggström I, Szczypiński P, Gibbs P, Cook G. Introduction to Radiomics. J Nucl Med 2020; 61:488-495. [PMID: 32060219 DOI: 10.2967/jnumed.118.222893] [Citation(s) in RCA: 927] [Impact Index Per Article: 185.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiomics is a rapidly evolving field of research concerned with the extraction of quantitative metrics-the so-called radiomic features-within medical images. Radiomic features capture tissue and lesion characteristics such as heterogeneity and shape and may, alone or in combination with demographic, histologic, genomic, or proteomic data, be used for clinical problem solving. The goal of this continuing education article is to provide an introduction to the field, covering the basic radiomics workflow: feature calculation and selection, dimensionality reduction, and data processing. Potential clinical applications in nuclear medicine that include PET radiomics-based prediction of treatment response and survival will be discussed. Current limitations of radiomics, such as sensitivity to acquisition parameter variations, and common pitfalls will also be covered.
Collapse
Affiliation(s)
- Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Andrzej Materka
- Institute of Electronics, Lodz University of Technology, Lodz, Poland
| | - Georg Langs
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ida Häggström
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Piotr Szczypiński
- Institute of Electronics, Lodz University of Technology, Lodz, Poland
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gary Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; and.,King's College London and Guy's and St. Thomas' PET Centre, St. Thomas' Hospital, London, United Kingdom
| |
Collapse
|
23
|
Ger RB, Meier JG, Pahlka RB, Gay S, Mumme R, Fuller CD, Li H, Howell RM, Layman RR, Stafford RJ, Zhou S, Mawlawi O, Court LE. Effects of alterations in positron emission tomography imaging parameters on radiomics features. PLoS One 2019; 14:e0221877. [PMID: 31487307 PMCID: PMC6728031 DOI: 10.1371/journal.pone.0221877] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/16/2019] [Indexed: 01/11/2023] Open
Abstract
Radiomics studies require large patient cohorts, which often include patients imaged using different imaging protocols. We aimed to determine the impact of variability in imaging protocol parameters and interscanner variability using a phantom that produced feature values similar to those of patients. Positron emission tomography (PET) scans of a Hoffman brain phantom were acquired on GE Discovery 710, Siemens mCT, and Philips Vereos scanners. A standard-protocol scan was acquired on each machine, and then each parameter that could be changed was altered individually. The phantom was contoured with 10 regions of interest (ROIs). Values for 45 features with 2 different preprocessing techniques were extracted for each image. To determine the impact of each parameter on the reliability of each radiomics feature, the intraclass correlation coefficient (ICC) was calculated with the ROIs as the subjects and the parameter values as the raters. For interscanner comparisons, we compared the standard deviation of each radiomics feature value from the standard-protocol images to the standard deviation of the same radiomics feature from PET scans of 224 patients with non-small cell lung cancer. When the pixel size was resampled prior to feature extraction, all features had good reliability (ICC > 0.75) for the field of view and matrix size. The time per bed position had excellent reliability (ICC > 0.9) on all features. When the filter cutoff was restricted to values below 6 mm, all features had good reliability. Similarly, when subsets and iterations were restricted to reasonable values used in clinics, almost all features had good reliability. The average ratio of the standard deviation of features on the phantom scans to that of the NSCLC patient scans was 0.73 using fixed-bin-width preprocessing and 0.92 using 64-level preprocessing. Most radiomics feature values had at least good reliability when imaging protocol parameters were within clinically used ranges. However, interscanner variability was about equal to interpatient variability; therefore, caution must be used when combining patients scanned on equipment from different vendors in radiomics data sets.
Collapse
Affiliation(s)
- Rachel B. Ger
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- * E-mail:
| | - Joseph G. Meier
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Raymond B. Pahlka
- Department of Radiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Skylar Gay
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Raymond Mumme
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Clifton D. Fuller
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Heng Li
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Rebecca M. Howell
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Rick R. Layman
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - R. Jason Stafford
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Shouhao Zhou
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Osama Mawlawi
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Laurence E. Court
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
24
|
Pfaehler E, van Sluis J, Merema BBJ, van Ooijen P, Berendsen RCM, van Velden FHP, Boellaard R. Experimental Multicenter and Multivendor Evaluation of the Performance of PET Radiomic Features Using 3-Dimensionally Printed Phantom Inserts. J Nucl Med 2019; 61:469-476. [PMID: 31420497 DOI: 10.2967/jnumed.119.229724] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/24/2019] [Indexed: 01/27/2023] Open
Abstract
The sensitivity of radiomic features to several confounding factors, such as reconstruction settings, makes clinical use challenging. To investigate the impact of harmonized image reconstructions on feature consistency, a multicenter phantom study was performed using 3-dimensionally printed phantom inserts reflecting realistic tumor shapes and heterogeneity uptakes. Methods: Tumors extracted from real PET/CT scans of patients with non-small cell lung cancer served as model for three 3-dimensionally printed inserts. Different heterogeneity pattern were realized by printing separate compartments that could be filled with different activity solutions. The inserts were placed in the National Electrical Manufacturers Association image-quality phantom and scanned various times. First, a list-mode scan was acquired and 5 statistically equal replicates were reconstructed. Second, the phantom was scanned 4 times on the same scanner. Third, the phantom was scanned on 6 PET/CT systems. All images were reconstructed using EANM Research Ltd. (EARL)-compliant and locally clinically preferred reconstructions. EARL-compliant reconstructions were performed without (EARL1) or with (EARL2) point-spread function. Images were analyzed with and without resampling to 2-mm cubic voxels. Images were discretized with a fixed bin width (FBW) of 0.25 and a fixed bin number (FBN) of 64. The intraclass correlation coefficient (ICC) of each scan setup was calculated and compared across reconstruction settings. An ICC above 0.75 was regarded as high. Results: The percentage of features yielding a high ICC was largest for the statistically equal replicates (70%-91% for FBN; 90%-96% for FBW discretization). For scans acquired on the same system, the percentage decreased, but most features still resulted in a high ICC (FBN, 52%-63%; FBW, 75%-85%). The percentage of features yielding a high ICC decreased more in the multicenter setting. In this case, the percentage of features yielding a high ICC was larger for images reconstructed with EARL-compliant reconstructions: for example, 40% for EARL1 and 60% for EARL2 versus 21% for the clinically preferred setting for FBW discretization. When discretized with FBW and resampled to isotropic voxels, this benefit was more pronounced. Conclusion: EARL-compliant reconstructions harmonize a wide range of radiomic features. FBW discretization and a sampling to isotropic voxels enhances the benefits of EARL-compliant reconstructions.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
| | - Bram B J Merema
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van Ooijen
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ralph C M Berendsen
- Department of Medical Physics, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Zwanenburg A. Radiomics in nuclear medicine: robustness, reproducibility, standardization, and how to avoid data analysis traps and replication crisis. Eur J Nucl Med Mol Imaging 2019; 46:2638-2655. [PMID: 31240330 DOI: 10.1007/s00259-019-04391-8] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/16/2022]
Abstract
Radiomics in nuclear medicine is rapidly expanding. Reproducibility of radiomics studies in multicentre settings is an important criterion for clinical translation. We therefore performed a meta-analysis to investigate reproducibility of radiomics biomarkers in PET imaging and to obtain quantitative information regarding their sensitivity to variations in various imaging and radiomics-related factors as well as their inherent sensitivity. Additionally, we identify and describe data analysis pitfalls that affect the reproducibility and generalizability of radiomics studies. After a systematic literature search, 42 studies were included in the qualitative synthesis, and data from 21 were used for the quantitative meta-analysis. Data concerning measurement agreement and reliability were collected for 21 of 38 different factors associated with image acquisition, reconstruction, segmentation and radiomics-specific processing steps. Variations in voxel size, segmentation and several reconstruction parameters strongly affected reproducibility, but the level of evidence remained weak. Based on the meta-analysis, we also assessed inherent sensitivity to variations of 110 PET image biomarkers. SUVmean and SUVmax were found to be reliable, whereas image biomarkers based on the neighbourhood grey tone difference matrix and most biomarkers based on the size zone matrix were found to be highly sensitive to variations, and should be used with care in multicentre settings. Lastly, we identify 11 data analysis pitfalls. These pitfalls concern model validation and information leakage during model development, but also relate to reporting and the software used for data analysis. Avoiding such pitfalls is essential for minimizing bias in the results and to enable reproduction and validation of radiomics studies.
Collapse
Affiliation(s)
- Alex Zwanenburg
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.
| |
Collapse
|
26
|
Forgács A, Béresová M, Garai I, Lassen ML, Beyer T, DiFranco MD, Berényi E, Balkay L. Impact of intensity discretization on textural indices of [ 18F]FDG-PET tumour heterogeneity in lung cancer patients. Phys Med Biol 2019; 64:125016. [PMID: 31108468 DOI: 10.1088/1361-6560/ab2328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Quantifying tumour heterogeneity from [18F]FDG-PET images promises benefits for treatment selection of cancer patients. Here, the calculation of texture parameters mandates an initial discretization step (binning) to reduce the number of intensity levels. Typically, three types of discrimination methods are used: lesion relative resampling (LRR) with fixed bin number, lesion absolute resampling (LAR) and absolute resampling (AR) with fixed bin widths. We investigated the effects of varying bin widths or bin number using 27 commonly cited local and regional texture indices (TIs) applied on lung tumour volumes. The data set were extracted from 58 lung cancer patients, with three different and robust tumour segmentation methods. In our cohort, the variations of the mean value as the function of the bin widths were similar for TIs calculated with LAR and AR quantification. The TI histograms calculated by LRR method showed distinct behaviour and its numerical values substantially effected by the selected bin number. The correlations of the AR and LAR based TIs demonstrated no principal differences between these methods. However, no correlation was found for the interrelationship between the TIs calculated by LRR and LAR (or AR) discretization method. Visual classification of the texture was also performed for each lesion. This classification analysis revealed that the parameters show statistically significant correlation with the visual score, if LAR or AR discretization method is considered, in contrast to LRR. Moreover, all the resulted tendencies were similar regardless the segmentation methods and the type of textural features involved in this work.
Collapse
Affiliation(s)
- Attila Forgács
- Scanomed Nuclear Medicine Center, Debrecen, Hungary. Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary. Author to whom any correspondence should be addressed
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Nie K, Al-Hallaq H, Li XA, Benedict SH, Sohn JW, Moran JM, Fan Y, Huang M, Knopp MV, Michalski JM, Monroe J, Obcemea C, Tsien CI, Solberg T, Wu J, Xia P, Xiao Y, El Naqa I. NCTN Assessment on Current Applications of Radiomics in Oncology. Int J Radiat Oncol Biol Phys 2019; 104:302-315. [PMID: 30711529 PMCID: PMC6499656 DOI: 10.1016/j.ijrobp.2019.01.087] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
Abstract
Radiomics is a fast-growing research area based on converting standard-of-care imaging into quantitative minable data and building subsequent predictive models to personalize treatment. Radiomics has been proposed as a study objective in clinical trial concepts and a potential biomarker for stratifying patients across interventional treatment arms. In recognizing the growing importance of radiomics in oncology, a group of medical physicists and clinicians from NRG Oncology reviewed the current status of the field and identified critical issues, providing a general assessment and early recommendations for incorporation in oncology studies.
Collapse
Affiliation(s)
- Ke Nie
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey.
| | - Hania Al-Hallaq
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - X Allen Li
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stanley H Benedict
- Department of Radiation Oncology, University of California-Davis, Sacramento, California
| | - Jason W Sohn
- Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Jean M Moran
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Yong Fan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mi Huang
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael V Knopp
- Division of Imaging Science, Department of Radiology, Ohio State University, Columbus, Ohio
| | - Jeff M Michalski
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - James Monroe
- Department of Radiation Oncology, St. Anthony's Cancer Center, St. Louis, Missouri
| | - Ceferino Obcemea
- Radiation Research Program, National Cancer Institute, Bethesda, Maryland
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Timothy Solberg
- Department of Radiation Oncology, University of California-San Francisco, San Francisco, California
| | - Jackie Wu
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Ping Xia
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Ying Xiao
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Issam El Naqa
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| |
Collapse
|
28
|
Ha S, Choi H, Paeng JC, Cheon GJ. Radiomics in Oncological PET/CT: a Methodological Overview. Nucl Med Mol Imaging 2019; 53:14-29. [PMID: 30828395 DOI: 10.1007/s13139-019-00571-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/27/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023] Open
Abstract
Radiomics is a medical imaging analysis approach based on computer-vision. Metabolic radiomics in particular analyses the spatial distribution patterns of molecular metabolism on PET images. Measuring intratumoral heterogeneity via image is one of the main targets of radiomics research, and it aims to build a image-based model for better patient management. The workflow of radiomics using texture analysis follows these steps: 1) imaging (image acquisition and reconstruction); 2) preprocessing (segmentation & quantization); 3) quantification (texture matrix design & texture feature extraction); and 4) analysis (statistics and/or machine learning). The parameters or conditions at each of these steps are effect on the results. In statistical testing or modeling, problems such as multiple comparisons, dependence on other variables, and high dimensionality of small sample size data should be considered. Standardization of methodology and harmonization of image quality are one of the most important challenges with radiomics methodology. Even though there are current issues in radiomics methodology, it is expected that radiomics will be clinically useful in personalized medicine for oncology.
Collapse
Affiliation(s)
- Seunggyun Ha
- 1Radiation Medicine Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- 2Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hongyoon Choi
- 2Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jin Chul Paeng
- 2Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Gi Jeong Cheon
- 1Radiation Medicine Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- 2Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- 3Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
29
|
Aide N, Salomon T, Blanc-Fournier C, Grellard JM, Levy C, Lasnon C. Implications of reconstruction protocol for histo-biological characterisation of breast cancers using FDG-PET radiomics. EJNMMI Res 2018; 8:114. [PMID: 30594961 PMCID: PMC6311169 DOI: 10.1186/s13550-018-0466-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/10/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The aim of this study is to determine if the choice of the 18F-FDG-PET protocol, especially matrix size and reconstruction algorithm, is of importance to discriminate between immunohistochemical subtypes (luminal versus non-luminal) in breast cancer with textural features (TFs). PROCEDURES Forty-seven patients referred for breast cancer staging in the framework of a prospective study were reviewed as part of an ancillary study. In addition to standard PET imaging (PSFWholeBody), a high-resolution breast acquisition was performed and reconstructed with OSEM and PSF (OSEMbreast/PSFbreast). PET standard metrics and TFs were extracted. For each reconstruction protocol, a prediction model for tumour classification was built using a random forests method. Spearman coefficients were used to seek correlation between PET metrics. RESULTS PSFWholeBody showed lower numbers of voxels within VOIs than OSEMbreast and PSFbreast with median (interquartile range) equal to 130 (43-271), 316 (167-1042), 367 (107-1221), respectively (p < 0.0001). Therefore, using LifeX software, 28 (59%), 46 (98%) and 42 (89%) patients were exploitable with PSFWholeBody, OSEMbreast and PSFbreast, respectively. On matched comparisons, PSFbreast reconstruction presented better abilities than PSFwholeBody and OSEMbreast for the classification of luminal versus non-luminal breast tumours with an accuracy reaching 85.7% as compared to 67.8% for PSFwholeBody and 73.8% for OSEMbreast. PSFbreast accuracy, sensitivity, specificity, PPV and NPV were equal to 85.7%, 94.3%, 42.9%, 89.2%, 60.0%, respectively. Coarseness and ZLNU were found to be main variables of importance, appearing in all three prediction models. Coarseness was correlated with SUVmax on PSFwholeBody images (ρ = - 0.526, p = 0.005), whereas it was not on OSEMbreast (ρ = - 0.183, p = 0.244) and PSFbreast (ρ = - 0.244, p = 0.119) images. Moreover, the range of its values was higher on PSFbreast images as compared to OSEMbreast, especially in small lesions (MTV < 3 ml). CONCLUSIONS High-resolution breast PET acquisitions, applying both small-voxel matrix and PSF modelling, appeared to improve the characterisation of breast tumours.
Collapse
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, University Hospital, Caen, France.,INSERM 1199 ANTICIPE, Normandy University, Caen, France
| | | | | | - Jean-Michel Grellard
- Biostatistics and Clinical Research Unit, François Baclesse Cancer Centre, Caen, France
| | - Christelle Levy
- Breast Cancer Unit, François Baclesse Cancer Centre, Caen, France
| | - Charline Lasnon
- INSERM 1199 ANTICIPE, Normandy University, Caen, France. .,Nuclear Medicine Department, François Baclesse Cancer Centre, 3 Avenue du Général Harris, BP 45026 Cedex 5, 14076, Caen, France.
| |
Collapse
|
30
|
Zhuang M, García DV, Kramer GM, Frings V, Smit EF, Dierckx R, Hoekstra OS, Boellaard R. Variability and Repeatability of Quantitative Uptake Metrics in 18F-FDG PET/CT of Non-Small Cell Lung Cancer: Impact of Segmentation Method, Uptake Interval, and Reconstruction Protocol. J Nucl Med 2018; 60:600-607. [PMID: 30389824 DOI: 10.2967/jnumed.118.216028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022] Open
Abstract
There is increased interest in various new quantitative uptake metrics beyond SUV in oncologic PET/CT studies. The purpose of this study was to investigate the variability and test-retest ratio (TRT) of metabolically active tumor volume (MATV) measurements and several other new quantitative metrics in non-small cell lung cancer using 18F-FDG PET/CT with different segmentation methods, user interactions, uptake intervals, and reconstruction protocols. Methods: Ten patients with advanced non-small cell lung cancer received 2 series of 2 whole-body 18F-FDG PET/CT scans at 60 min after injection and at 90 min after injection. PET data were reconstructed with 4 different protocols. Eight segmentation methods were applied to delineate lesions with and without a tumor mask. MATV, SUVmax, SUVmean, total lesion glycolysis, and intralesional heterogeneity features were derived. Variability and repeatability were evaluated using a generalized-estimating-equation statistical model with Bonferroni adjustment for multiple comparisons. The statistical model, including interaction between uptake interval and reconstruction protocol, was applied individually to the data obtained from each segmentation method. Results: Without masking, none of the segmentation methods could delineate all lesions correctly. MATV was affected by both uptake interval and reconstruction settings for most segmentation methods. Similar observations were obtained for the uptake metrics SUVmax, SUVmean, total lesion glycolysis, homogeneity, entropy, and zone percentage. No effect of uptake interval was observed on TRT metrics, whereas the reconstruction protocol affected the TRT of SUVmax Overall, segmentation methods showing poor quantitative performance in one condition showed better performance in other (combined) conditions. For some metrics, a clear statistical interaction was found between the segmentation method and both uptake interval and reconstruction protocol. Conclusion: All segmentation results need to be reviewed critically. MATV and other quantitative uptake metrics, as well as their TRT, depend on segmentation method, uptake interval, and reconstruction protocol. To obtain quantitative reliable metrics, with good TRT performance, the optimal segmentation method depends on local imaging procedure, the PET/CT system, or reconstruction protocol. Rigid harmonization of imaging procedure and PET/CT performance will be helpful in mitigating this variability.
Collapse
Affiliation(s)
- Mingzan Zhuang
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,The Key Laboratory of Digital Signal and Image Processing of Guangdong Province, Shantou University, Shantou, China
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Virginie Frings
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - E F Smit
- Department of Pulmonary Disease, VU University Medical Center, Amsterdam, The Netherlands
| | - Rudi Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands .,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
31
|
Aide N, Hicks RJ, Le Tourneau C, Lheureux S, Fanti S, Lopci E. FDG PET/CT for assessing tumour response to immunotherapy : Report on the EANM symposium on immune modulation and recent review of the literature. Eur J Nucl Med Mol Imaging 2018; 46:238-250. [PMID: 30291373 PMCID: PMC6267687 DOI: 10.1007/s00259-018-4171-4] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
This paper follows the immunotherapy symposium held during the European Association of Nuclear Medicine (EANM) 2017 Annual Congress. The biological basis of the immune checkpoint inhibitors and the drugs most frequently used for the treatment of solid tumours are reviewed. The issues of pseudoprogression (frequency, timeline), hyperprogression and immune-related side effects are discussed, as well as their implications for patient management. A review of the recent literature on the use of FDG PET for assessment of immunotherapy is presented, and recommendations are provided for assessing tumour response and reporting immune-related side effects with FDG PET based on published data and experts' experience. Representative clinical cases are also discussed.
Collapse
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, Caen University Hospital, Caen, France. .,Normandie University, Caen, France. .,INSERM 1086 ANTICIPE, Normandie University, Caen, France. .,EANM Oncology Committee, Vienna, Austria.
| | - Rodney J Hicks
- Centre for Molecular Imaging, Department of Cancer Imaging, Peter MacCallum Cancer, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Christophe Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France.,INSERM U900 Research Unit, Saint-Cloud, France
| | - Stéphanie Lheureux
- Princess Margaret Cancer Centre, Department of Medical Oncology, University of Toronto, Toronto, ON, Canada
| | - Stefano Fanti
- EANM Oncology Committee, Vienna, Austria.,Nuclear Medicine, Policlinico S. Orsola, Università di Bologna, Bologna, Italy
| | - Egesta Lopci
- EANM Oncology Committee, Vienna, Austria.,Nuclear Medicine Department, Humanitas Clinical and Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
32
|
Lovinfosse P, Visvikis D, Hustinx R, Hatt M. FDG PET radiomics: a review of the methodological aspects. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0292-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Traverso A, Wee L, Dekker A, Gillies R. Repeatability and Reproducibility of Radiomic Features: A Systematic Review. Int J Radiat Oncol Biol Phys 2018; 102:1143-1158. [PMID: 30170872 PMCID: PMC6690209 DOI: 10.1016/j.ijrobp.2018.05.053] [Citation(s) in RCA: 534] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 12/15/2022]
Abstract
Purpose: An ever-growing number of predictive models used to inform clinical decision making have included quantitative, computer-extracted imaging biomarkers, or “radiomic features.” Broadly generalizable validity of radiomics-assisted models may be impeded by concerns about reproducibility. We offer a qualitative synthesis of 41 studies that specifically investigated the repeatability and reproducibility of radiomic features, derived from a systematic review of published peer-reviewed literature. Methods and Materials: The PubMed electronic database was searched using combinations of the broad Haynes and Ingui filters along with a set of text words specific to cancer, radiomics (including texture analyses), reproducibility, and repeatability. This review has been reported in compliance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. From each full-text article, information was extracted regarding cancer type, class of radiomic feature examined, reporting quality of key processing steps, and statistical metric used to segregate stable features. Results: Among 624 unique records, 41 full-text articles were subjected to review. The studies primarily addressed non-small cell lung cancer and oropharyngeal cancer. Only 7 studies addressed in detail every methodologic aspect related to image acquisition, preprocessing, and feature extraction. The repeatability and reproducibility of radiomic features are sensitive at various degrees to processing details such as image acquisition settings, image reconstruction algorithm, digital image preprocessing, and software used to extract radiomic features. First-order features were overall more reproducible than shape metrics and textural features. Entropy was consistently reported as one of the most stable first-order features. There was no emergent consensus regarding either shape metrics or textural features; however, coarseness and contrast appeared among the least reproducible. Conclusions: Investigations of feature repeatability and reproducibility are currently limited to a small number of cancer types. Reporting quality could be improved regarding details of feature extraction software, digital image manipulation (preprocessing), and the cutoff value used to distinguish stable features. We offer a qualitative synthesis of 41 studies that specifically investigated the repeatability and reproducibility of radiomic features. The repeatability and reproducibility of radiomic features are sensitive at various degrees to image quality and to software used to extract radiomic features. Investigations of feature repeatability and reproducibility are currently limited to a small number of cancer types. No consensus was found regarding the most repeatable and reproducible features with respect to different settings.
Collapse
Affiliation(s)
- Alberto Traverso
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| | - Leonard Wee
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
34
|
Radiomics in Nuclear Medicine Applied to Radiation Therapy: Methods, Pitfalls, and Challenges. Int J Radiat Oncol Biol Phys 2018; 102:1117-1142. [PMID: 30064704 DOI: 10.1016/j.ijrobp.2018.05.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023]
Abstract
Radiomics is a recent area of research in precision medicine and is based on the extraction of a large variety of features from medical images. In the field of radiation oncology, comprehensive image analysis is crucial to personalization of treatments. A better characterization of local heterogeneity and the shape of the tumor, depicting individual cancer aggressiveness, could guide dose planning and suggest volumes in which a higher dose is needed for better tumor control. In addition, noninvasive imaging features that could predict treatment outcome from baseline scans could help the radiation oncologist to determine the best treatment strategies and to stratify patients as at low risk or high risk of recurrence. Nuclear medicine molecular imaging reflects information regarding biological processes in the tumor thanks to a wide range of radiotracers. Many studies involving 18F-fluorodeoxyglucose positron emission tomography suggest an added value of radiomics compared with the use of conventional PET metrics such as standardized uptake value for both tumor diagnosis and prediction of recurrence or treatment outcome. However, these promising results should not hide technical difficulties that still currently prevent the approach from being widely studied or clinically used. These difficulties mostly pertain to the variability of the imaging features as a function of the acquisition device and protocol, the robustness of the models with respect to that variability, and the interpretation of the radiomic models. Addressing the impact of the variability in acquisition and reconstruction protocols is needed, as is harmonizing the radiomic feature calculation methods, to ensure the reproducibility of studies in a multicenter context and their implementation in a clinical workflow. In this review, we explain the potential impact of positron emission tomography radiomics for radiation therapy and underline the various aspects that need to be carefully addressed to make the most of this promising approach.
Collapse
|
35
|
Prediction of cervical cancer recurrence using textural features extracted from 18F-FDG PET images acquired with different scanners. Oncotarget 2018; 8:43169-43179. [PMID: 28574816 PMCID: PMC5522136 DOI: 10.18632/oncotarget.17856] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Objectives To identify an imaging signature predicting local recurrence for locally advanced cervical cancer (LACC) treated by chemoradiation and brachytherapy from baseline 18F-FDG PET images, and to evaluate the possibility of gathering images from two different PET scanners in a radiomic study. Methods 118 patients were included retrospectively. Two groups (G1, G2) were defined according to the PET scanner used for image acquisition. Eleven radiomic features were extracted from delineated cervical tumors to evaluate: (i) the predictive value of features for local recurrence of LACC, (ii) their reproducibility as a function of the scanner within a hepatic reference volume, (iii) the impact of voxel size on feature values. Results Eight features were statistically significant predictors of local recurrence in G1 (p < 0.05). The multivariate signature trained in G2 was validated in G1 (AUC=0.76, p<0.001) and identified local recurrence more accurately than SUVmax (p=0.022). Four features were significantly different between G1 and G2 in the liver. Spatial resampling was not sufficient to explain the stratification effect. Conclusion This study showed that radiomic features could predict local recurrence of LACC better than SUVmax. Further investigation is needed before applying a model designed using data from one PET scanner to another.
Collapse
|
36
|
Aide N, Lasnon C, Damaj G. Combining baseline TMTV and gene profiling for a better risk stratification in diffuse large B cell lymphoma. Eur J Nucl Med Mol Imaging 2018; 45:677-679. [PMID: 29455312 DOI: 10.1007/s00259-018-3966-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, Caen University Hospital, Caen, France. .,Normandie University, Caen, France. .,INSERM 1086 ANTICIPE, Normandie University, Caen, France.
| | - Charline Lasnon
- INSERM 1086 ANTICIPE, Normandie University, Caen, France.,Nuclear Medicine Department, François Baclesse Cancer Centre, Caen, France
| | - Gandhi Damaj
- Normandie University, Caen, France.,Haematology Institute, Caen University Hospital, Caen, France.,MICAH INSERM U1245, Rouen University, Rouen, France
| |
Collapse
|
37
|
Orlhac F, Boughdad S, Philippe C, Stalla-Bourdillon H, Nioche C, Champion L, Soussan M, Frouin F, Frouin V, Buvat I. A Postreconstruction Harmonization Method for Multicenter Radiomic Studies in PET. J Nucl Med 2018; 59:1321-1328. [PMID: 29301932 DOI: 10.2967/jnumed.117.199935] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/03/2017] [Indexed: 12/14/2022] Open
Abstract
Several reports have shown that radiomic features are affected by acquisition and reconstruction parameters, thus hampering multicenter studies. We propose a method that, by removing the center effect while preserving patient-specific effects, standardizes features measured from PET images obtained using different imaging protocols. Methods: Pretreatment 18F-FDG PET images of patients with breast cancer were included. In one nuclear medicine department (department A), 63 patients were scanned on a time-of-flight PET/CT scanner, and 16 lesions were triple-negative (TN). In another nuclear medicine department (department B), 74 patients underwent PET/CT on a different brand of scanner and a different reconstruction protocol, and 15 lesions were TN. The images from department A were smoothed using a gaussian filter to mimic data from a third department (department A-S). The primary lesion was segmented to obtain a lesion volume of interest (VOI), and a spheric VOI was set in healthy liver tissue. Three SUVs and 6 textural features were computed in all VOIs. A harmonization method initially described for genomic data was used to estimate the department effect based on the observed feature values. Feature distributions in each department were compared before and after harmonization. Results: In healthy liver tissue, the distributions significantly differed for 4 of 9 features between departments A and B and for 6 of 9 between departments A and A-S (P < 0.05, Wilcoxon test). After harmonization, none of the 9 feature distributions significantly differed between 2 departments (P > 0.1). The same trend was observed in lesions, with a realignment of feature distributions between the departments after harmonization. Identification of TN lesions was largely enhanced after harmonization when the cutoffs were determined on data from one department and applied to data from the other department. Conclusion: The proposed harmonization method is efficient at removing the multicenter effect for textural features and SUVs. The method is easy to use, retains biologic variations not related to a center effect, and does not require any feature recalculation. Such harmonization allows for multicenter studies and for external validation of radiomic models or cutoffs and should facilitate the use of radiomic models in clinical practice.
Collapse
Affiliation(s)
- Fanny Orlhac
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Sarah Boughdad
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France.,Department of Nuclear Medicine, Institut Curie-René Huguenin, Saint-Cloud, France
| | - Cathy Philippe
- NeuroSpin/UNATI, CEA, Université Paris-Saclay, Gif-sur-Yvette, France; and
| | | | - Christophe Nioche
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Laurence Champion
- Department of Nuclear Medicine, Institut Curie-René Huguenin, Saint-Cloud, France
| | - Michaël Soussan
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France.,Department of Nuclear Medicine, AP-HP, Hôpital Avicenne, Bobigny, France
| | - Frédérique Frouin
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Vincent Frouin
- NeuroSpin/UNATI, CEA, Université Paris-Saclay, Gif-sur-Yvette, France; and
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, CEA-SHFJ, INSERM, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| |
Collapse
|
38
|
Aide N, Talbot M, Fruchart C, Damaj G, Lasnon C. Diagnostic and prognostic value of baseline FDG PET/CT skeletal textural features in diffuse large B cell lymphoma. Eur J Nucl Med Mol Imaging 2017; 45:699-711. [PMID: 29214417 PMCID: PMC5978926 DOI: 10.1007/s00259-017-3899-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/23/2017] [Indexed: 12/20/2022]
Abstract
Purpose Our purpose was to evaluate the diagnostic and prognostic value of skeletal textural features (TFs) on baseline FDG PET in diffuse large B cell lymphoma (DLBCL) patients. Methods Eighty-two patients with DLBCL who underwent a bone marrow biopsy (BMB) and a PET scan between December 2008 and December 2015 were included. Two readers blinded to the BMB results visually assessed PET images for bone marrow involvement (BMI) in consensus, and a third observer drew a volume of interest (VOI) encompassing the axial skeleton and the pelvis, which was used to assess skeletal TFs. ROC analysis was used to determine the best TF able to diagnose BMI among four first-order, six second-order and 11 third-order metrics, which was then compared for diagnosis and prognosis in disease-free patients (BMB−/PET-) versus patients considered to have BMI (BMB+/PET-, BMB−/PET+, and BMB+/PET+). Results Twenty-two out of 82 patients (26.8%) had BMI: 13 BMB−/PET+, eight BMB+/PET+ and one BMB+/PET-. Among the nine BMB+ patients, one had discordant BMI identified by both visual and TF PET assessment. ROC analysis showed that SkewnessH, a first-order metric, was the best parameter for identifying BMI with sensitivity and specificity of 81.8% and 81.7%, respectively. SkewnessH demonstrated better discriminative power over BMB and PET visual analysis for patient stratification: hazard ratios (HR), 3.78 (P = 0.02) versus 2.81 (P = 0.06) for overall survival (OS) and HR, 3.17 (P = 0.03) versus 1.26 (P = 0.70) for progression-free survival (PFS). In multivariate analysis accounting for IPI score, bulky status, haemoglobin and SkewnessH, the only independent predictor of OS was the IPI score, while the only independent predictor of PFS was SkewnessH. Conclusion The better discriminative power of skeletal heterogeneity for risk stratification compared to BMB and PET visual analysis in the overall population, and more specifically in BMB−/PET- patients, suggests that it can be useful to identify diagnostically overlooked BMI.
Collapse
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, Caen University Hospital, Caen, France.,Normandie University, Caen, France.,INSERM 1086 ANTICIPE, Normandie University, Caen, France
| | | | | | - Gandhi Damaj
- Haematology Institute, Caen University Hospital, Caen, France
| | - Charline Lasnon
- INSERM 1086 ANTICIPE, Normandie University, Caen, France. .,Nuclear Medicine Department, François Baclesse Cancer Centre, Caen, France.
| |
Collapse
|
39
|
Lapa P, Marques M, Isidoro J, Barata F, Costa G, de Lima J. 18 F-FDG PET/CT in lung cancer. The added value of quantification. Rev Esp Med Nucl Imagen Mol 2017. [DOI: 10.1016/j.remnie.2017.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
40
|
Lapa P, Oliveiros B, Marques M, Isidoro J, Alves FC, Costa JMN, Costa G, de Lima JP. Metabolic tumor burden quantified on [ 18F]FDG PET/CT improves TNM staging of lung cancer patients. Eur J Nucl Med Mol Imaging 2017; 44:2169-2178. [PMID: 28785842 DOI: 10.1007/s00259-017-3789-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/19/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE The purpose of our study was to test a new staging algorithm, combining clinical TNM staging (cTNM) with whole-body metabolic active tumor volume (MATV-WB), with the goal of improving prognostic ability and stratification power. METHODS Initial staging [18F]FDG PET/CT of 278 non-small cell lung cancer (NSCLC) patients, performed between January/2011 and April/2016, 74(26.6%) women, 204(73.4%) men; aged 34-88 years (mean ± SD:66 ± 10), was retrospectively evaluated, and MATV-WB was quantified. Each patient's follow-up time was recorded: 0.7-83.6 months (mean ± SD:25.1 ± 20.3). RESULTS MATV-WB was an independent and statistically-significant predictor of overall survival (p < 0.001). The overall survival predictive ability of MATV-WB (C index: mean ± SD = 0.7071 ± 0.0009) was not worse than cTNM (C index: mean ± SD = 0.7031 ± 0.007) (Z = -0.143, p = 0.773). Estimated mean survival times of 56.3 ± 3.0 (95%CI:50.40-62.23) and 21.7 ± 2.2 months (95%CI:17.34-25.98) (Log-Rank = 77.48, p < 0.001), one-year survival rate of 86.8% and of 52.8%, and five-year survival rate of 53.6% and no survivors, were determined, respectively, for patients with MATV-WB < 49.5 and MATV-WB ≥ 49.5. Patients with MATV-WB ≥ 49.5 had a mortality risk 2.9-5.8 times higher than those with MATV-WB < 49.5 (HR = 4.12, p < 0.001). MATV-WB cutoff points were also determined for each cTNM stage: 23.7(I), 49.5(II), 52(III), 48.8(IV) (p = 0.029, p = 0.227, p = 0.025 and p = 0.001, respectively). At stages I, III and IV there was a statistically-significant difference in the estimated mean overall survival time between groups of patients defined by the cutoff points (p = 0.007, p = 0.004 and p < 0.001, respectively). At stage II (p = 0.365), there was a clinically-significant difference of about 12 months between the groups. In all cTNM stages, patients with MATV-WB ≥ cutoff points had lower survival rates. Combined clinical TNM-PET staging (cTNM-P) was then tested: Stage I < 23.7; Stage I ≥ 23.7; Stage II < 49.5; Stage II ≥ 49.5; Stage III < 52; Stage III ≥ 52; Stage IV < 48.8; Stage IV ≥ 48.8. cTNM-P staging presented a superior overall survival predictive ability (C index = 0.730) compared with conventional cTNM staging (C index = 0.699) (Z = -4.49, p < 0.001). CONCLUSION cTNM-P staging has superior prognostic value compared with conventional cTNM staging, and allows better stratification of NSCLC patients.
Collapse
Affiliation(s)
- Paula Lapa
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
| | - Bárbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Institute for Biomedical Imaging and Life Sciences, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Margarida Marques
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Technology and Information Systems Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Jorge Isidoro
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Filipe Caseiro Alves
- Radiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - J M Nascimento Costa
- University Oncology Clinic, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Gracinda Costa
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - João Pedroso de Lima
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health-ICNAS, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
41
|
Ben Bouallègue F, Tabaa YA, Kafrouni M, Cartron G, Vauchot F, Mariano-Goulart D. Association between textural and morphological tumor indices on baseline PET-CT and early metabolic response on interim PET-CT in bulky malignant lymphomas. Med Phys 2017; 44:4608-4619. [DOI: 10.1002/mp.12349] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Fayçal Ben Bouallègue
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Yassine Al Tabaa
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Marilyne Kafrouni
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Guillaume Cartron
- Haematology Department; Saint Eloi University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Fabien Vauchot
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Denis Mariano-Goulart
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
- U1046 INSERM - UMR9214 CNRS; CHU Arnaud de Villeneuve; 371 Avenue du Doyen Giraud 34295 Montpellier Cedex 5 France
| |
Collapse
|
42
|
Aide N, Lasnon C, Veit-Haibach P, Sera T, Sattler B, Boellaard R. EANM/EARL harmonization strategies in PET quantification: from daily practice to multicentre oncological studies. Eur J Nucl Med Mol Imaging 2017; 44:17-31. [PMID: 28623376 PMCID: PMC5541084 DOI: 10.1007/s00259-017-3740-2] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/24/2017] [Indexed: 01/18/2023]
Abstract
Quantitative positron emission tomography/computed tomography (PET/CT) can be used as diagnostic or prognostic tools (i.e. single measurement) or for therapy monitoring (i.e. longitudinal studies) in multicentre studies. Use of quantitative parameters, such as standardized uptake values (SUVs), metabolic active tumor volumes (MATVs) or total lesion glycolysis (TLG), in a multicenter setting requires that these parameters be comparable among patients and sites, regardless of the PET/CT system used. This review describes the motivations and the methodologies for quantitative PET/CT performance harmonization with emphasis on the EANM Research Ltd. (EARL) Fluorodeoxyglucose (FDG) PET/CT accreditation program, one of the international harmonization programs aiming at using FDG PET as a quantitative imaging biomarker. In addition, future accreditation initiatives will be discussed. The validation of the EARL accreditation program to harmonize SUVs and MATVs is described in a wide range of tumor types, with focus on therapy assessment using either the European Organization for Research and Treatment of Cancer (EORTC) criteria or PET Evaluation Response Criteria in Solid Tumors (PERCIST), as well as liver-based scales such as the Deauville score. Finally, also presented in this paper are the results from a survey across 51 EARL-accredited centers reporting how the program was implemented and its impact on daily routine and in clinical trials, harmonization of new metrics such as MATV and heterogeneity features.
Collapse
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, University Hospital, Caen, France.
- Inserm U1086 ANTICIPE, Caen University, Caen, France.
| | - Charline Lasnon
- Inserm U1086 ANTICIPE, Caen University, Caen, France
- Nuclear Medicine Department, François Baclesse Cancer Centre, Caen, France
| | - Patrick Veit-Haibach
- Department of Nuclear Medicine and Department of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
- Joint Department Medical Imaging, University Health Network, University of Toronto, Toronto, Canada
| | - Terez Sera
- Nuclear Medicine Department, University of Szeged, Szeged, Hungary
| | - Bernhard Sattler
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103, Leipzig, Germany
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Lapa P, Marques M, Isidoro J, Barata F, Costa G, de Lima JP. 18F-FDG PET/CT in lung cancer. The added value of quantification. Rev Esp Med Nucl Imagen Mol 2017; 36:342-349. [PMID: 28566260 DOI: 10.1016/j.remn.2017.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To test a software application for the quantification of metabolic heterogeneity and to evaluate its superiority in relation to visual interpretation. To investigate if a quantitative analysis adds information to the interpretation of 18F-FDG-PET/CT. MATERIAL AND METHODS The study analyzed 215 patients with a 18F-FDG-PET/CT done for the initial staging of lung cancer between March 2011 and December 2015. The study included 57 (26.5%) women and 158 (73.5%) men, with ages ranging from 34 to 88 years (mean±SD: 67.23±10.04). There were 82 surgical stages (I, II, IIIA), and 133 non-surgical stages (IIIB, IV). The primary tumour was analyzed quantitatively by obtaining the following parameters: SUVmax, metabolic active tumour volume (MATV), total lesion glycolysis (TLG), and the entropy heterogeneity index (ET). Heterogeneity was assessed visually. Death dates and/or the follow-up time were registered, ranging from 0.70 to 67.60 months (mean±SD: 23.20±17.68). RESULTS In multivariate analysis, ET emerged as a better predictor of survival than visual analysis of heterogeneity that was not statistically significant. The C-index determination demonstrated that all quantitative parameters were statistically-significant predictors of survival. Cut-offs were obtained in order to compare survival times. A multivariate analysis was performed. In the total population, the best predictor was the TNM stage, but MATV, ET, and male gender were statistically significant and independent predictors of survival. In stages without surgical indication, the best predictor was the TNM stage, but the MATV and male gender were statistically significant and independent predictors of survival. In the surgical stages, ET was the only statistically significant and independent predictor of survival. CONCLUSIONS Quantification adds prognostic information to the visual analysis of 18F-FDG-PET/CT.
Collapse
Affiliation(s)
- P Lapa
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
| | - M Marques
- Technology and Information Systems Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - J Isidoro
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - F Barata
- Lung Diseases Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - G Costa
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - J P de Lima
- Nuclear Medicine Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Institute of Nuclear Sciences Applied to Health-ICNAS, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
44
|
Deroose CM, Stroobants S, Liu Y, Shankar LK, Bourguet P. Using PET for therapy monitoring in oncological clinical trials: challenges ahead. Eur J Nucl Med Mol Imaging 2017; 44:32-40. [DOI: 10.1007/s00259-017-3689-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 11/24/2022]
|
45
|
Scalco E, Rizzo G. Texture analysis of medical images for radiotherapy applications. Br J Radiol 2017; 90:20160642. [PMID: 27885836 PMCID: PMC5685100 DOI: 10.1259/bjr.20160642] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/27/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
The high-throughput extraction of quantitative information from medical images, known as radiomics, has grown in interest due to the current necessity to quantitatively characterize tumour heterogeneity. In this context, texture analysis, consisting of a variety of mathematical techniques that can describe the grey-level patterns of an image, plays an important role in assessing the spatial organization of different tissues and organs. For these reasons, the potentiality of texture analysis in the context of radiotherapy has been widely investigated in several studies, especially for the prediction of the treatment response of tumour and normal tissues. Nonetheless, many different factors can affect the robustness, reproducibility and reliability of textural features, thus limiting the impact of this technique. In this review, an overview of the most recent works that have applied texture analysis in the context of radiotherapy is presented, with particular focus on the assessment of tumour and tissue response to radiations. Preliminary, the main factors that have an influence on features estimation are discussed, highlighting the need of more standardized image acquisition and reconstruction protocols and more accurate methods for region of interest identification. Despite all these limitations, texture analysis is increasingly demonstrating its ability to improve the characterization of intratumour heterogeneity and the prediction of clinical outcome, although prospective studies and clinical trials are required to draw a more complete picture of the full potential of this technique.
Collapse
Affiliation(s)
- Elisa Scalco
- Institute of Molecular Bioimaging and Physiology (IBFM), Italian
National Research Council (CNR), Milan, Italy
| | - Giovanna Rizzo
- Institute of Molecular Bioimaging and Physiology (IBFM), Italian
National Research Council (CNR), Milan, Italy
| |
Collapse
|
46
|
Hatt M, Tixier F, Visvikis D, Cheze Le Rest C. Radiomics in PET/CT: More Than Meets the Eye? J Nucl Med 2016; 58:365-366. [PMID: 27811126 DOI: 10.2967/jnumed.116.184655] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 01/07/2023] Open
Affiliation(s)
- Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, IBSAM, Brest, France; and
| | - Florent Tixier
- Academic Department of Nuclear Medicine, CHU Poitiers, Poitiers, France
| | - Dimitris Visvikis
- LaTIM, INSERM, UMR 1101, University of Brest, IBSAM, Brest, France; and
| | | |
Collapse
|