1
|
Wang TW, Wang CK, Hong JS, Lin YH, Wang SY, Lu CF, Wu YT. Prognostic power of radiomics in head and neck cancers: Insights from a meta-analysis. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 262:108683. [PMID: 40009959 DOI: 10.1016/j.cmpb.2025.108683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Prognostic modeling in head and neck cancers (HNC) has advanced with the integration of clinical factors and radiomic data from CT and MRI scans. However, previous reviews have not systematically evaluated the predictive performance of these models across different oncological endpoints or assessed factors affecting their generalizability. This study aims to fill this gap by providing a comprehensive analysis of prognostic models in HNC. METHODS Our systematic review and meta-analysis sourced data from PubMed, Embase, and Web of Science until August 30, 2023, shortlisting 16 studies. We concentrated on studies detailing HNC prognosis prediction through radiomics, which transparently tabulated performance metrics of c-index and utilized external validation sets. We excluded studies employing imaging techniques other than CT or MRI. Study quality was assessed using the QUIPS and RQS tools. Our meta-analysis comprised the radiomics prognosis model on all validation datasets, overall survival prediction with radiomics on all validation datasets, and overall survival prediction integrating clinical and radiomics data on external validation sets. All assessments adopted a random effects model. The research has been registered under CRD42023459049. RESULTS When evaluating by distinct endpoints, marked differences were observed. Delving deeper into the complexities of overall survival prediction, variables such as incorporation of clinical features and an enlarged training set were identified as major enhancers of the model's performance. Evaluating exclusively on external validation cohorts, purely clinical models demonstrated a prognostic strength of pooled 0.69 c-index for overall survival, in contrast to the 0.68 pooled c-index achieved by models rooted in radiomics. Combining both approaches elevated the pooled c-index to 0.76. It was clear that a blend of an expanded training dataset and features selected, coupled with the diversity in CT and MRI equipment and model counts, are pivotal in fortifying the model's resilience. CONCLUSION This systematic review and meta-analysis demonstrate that combining clinical and radiomic features significantly improves the predictive performance of prognostic models for overall survival in HNC. By systematically evaluating various endpoints and identifying key factors influencing model generalizability, our study fills a critical gap in the literature. These findings provide valuable insights for developing more accurate and personalized prognostic tools in HNC, guiding future research and enhancing clinical decision-making.
Collapse
Affiliation(s)
- Ting-Wei Wang
- Institute of Biophotonics, National Yang Ming Chiao Tung University, 155, Sec. 2, Li-Nong St. Beitou Dist., Taipei 112304, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Computer Science, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chih-Keng Wang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Otolaryngology-Head and Neck Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jia-Sheng Hong
- Institute of Biophotonics, National Yang Ming Chiao Tung University, 155, Sec. 2, Li-Nong St. Beitou Dist., Taipei 112304, Taiwan
| | - Yi-Hui Lin
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 407, Taiwan; College of Computer Science, National Yang Ming Chiao Tung University, Taiwan
| | - Shi-Yao Wang
- National Taiwan University, College of Medicine, Department of Dentistry, Taipei, Taiwan
| | - Chia-Fung Lu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, 155, Sec. 2, Li-Nong St. Beitou Dist., Taipei 112304, Taiwan; National Yang Ming Chiao Tung University, Brain Research Center, Taiwan; National Yang Ming Chiao Tung University, College Medical Device Innovation and Translation Center, Taiwan.
| |
Collapse
|
2
|
Zhou Y, Zhou XY, Xu YC, Ma XL, Tian R. Radiomics based on 18F-FDG PET for predicting treatment response and prognosis in newly diagnosed diffuse large B-cell lymphoma patients: do lesion selection and segmentation methods matter? Quant Imaging Med Surg 2025; 15:103-120. [PMID: 39839002 PMCID: PMC11744140 DOI: 10.21037/qims-24-585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/05/2024] [Indexed: 01/23/2025]
Abstract
Background Radiomics features extracted from baseline 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) scans have shown promising results in predicting the treatment response and outcome of diffuse large B-cell lymphoma (DLBCL) patients. This study aimed to assess the influence of lesion selection approaches and segmentation methods on the radiomics of DLBCL in terms of treatment response and prognosis prediction. Methods A total of 522 and 382 patients pathologically diagnosed with DLBCL were enrolled for complete regression and 2-year event-free survival prediction, respectively. Three lesion selection methods (largest or hottest lesion, patient level) and five segmentation methods (manual and four semiautomatic segmentations) were applied. A total of 112 radiomics features were extracted from the lesions and at the patient level. The feature selection was performed via random forest, and models were constructed via eXtreme Gradient Boosting. The performance of all the models was evaluated via the area under the curve (AUC), which was compared via the Delong test. Results The AUC values varied from 0.583 to 0.768 for the treatment response and prognosis prediction models on the basis of different lesion selection and segmentation methods. However, the prediction performance gap was not significant for each model (all P>0.05). The combined models (AUC =0.908 and 0.837 for treatment response and prognosis prediction, respectively) that incorporated radiomics and clinical features exhibited significant predictive superiority over the clinical models (AUC =0.622 and 0.636, respectively) and the international prognostic index model (AUC =0.623 for prognosis prediction) (all P<0.05). Conclusions Although there are differences in the selected radiomics features among lesion selection and segmentation methods, there is no substantial difference in the predictive power of each radiomics model. In addition, radiomics features have potential added value to clinical features.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xue-Yan Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yu-Chao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang, China
| | - Xue-Lei Ma
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Ortega C, Anconina R, Joshi S, Metser U, Prica A, Johnson S, Liu ZA, Keshavarzi S, Veit-Haibach P. Combination of FDG PET/CT radiomics and clinical parameters for outcome prediction in patients with non-Hodgkin's lymphoma. Nucl Med Commun 2024; 45:1039-1046. [PMID: 39412293 PMCID: PMC11537470 DOI: 10.1097/mnm.0000000000001895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/30/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE The purposes was to build model incorporating PET + computed tomography (CT) radiomics features from baseline PET/CT + clinical parameters to predict outcomes in patients with non-Hodgkin lymphomas. METHODS Cohort of 138 patients with complete clinical parameters and follow up times of 25.3 months recorded. Textural analysis of PET and manual correlating contouring in CT images analyzed using LIFE X software. Defined outcomes were overall survival (OS), disease free-survival, radiotherapy, and unfavorable response (defined as disease progression) assessed by end of therapy PET/CT or contrast CT. Univariable and multivariable analysis performed to assess association between PET, CT, and clinical. RESULTS Male ( P = 0.030), abnormal lymphocytes ( P = 0.030), lower value of PET entropy ( P = 0.030), higher value of SHAPE sphericity ( P = 0.002) were significantly associated with worse OS. Advanced stage (III or IV, P = 0.013), abnormal lymphocytes ( P = 0.032), higher value of CT gray-level run length matrix (GLRLM) LRLGE mean ( P = 0.010), higher value of PET gray-level co-occurrence matrix energy angular second moment ( P < 0.001), and neighborhood gray-level different matrix (NGLDM) busyness mean ( P < 0.001) were significant predictors of shorter DFS. Abnormal lymphocyte ( P = 0.033), lower value of CT NGLDM coarseness ( P = 0.082), and higher value of PET GLRLM gray-level nonuniformity zone mean ( P = 0.040) were significant predictors of unfavorable response to chemotherapy. Area under the curve for the three models (clinical alone, clinical + PET parameters, and clinical + PET + CT parameters) were 0.626, 0.716, and 0.759, respectively.
Collapse
Affiliation(s)
- Claudia Ortega
- Department Medical Imaging, University Medical Imaging Toronto, University Health Network – Mount Sinai Hospital – Women College Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Reut Anconina
- Department of Diagnostic Imaging, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Sayali Joshi
- Department of Diagnostic Imaging, The Hospital for Sick Children
| | - Ur Metser
- Department Medical Imaging, University Medical Imaging Toronto, University Health Network – Mount Sinai Hospital – Women College Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Anca Prica
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, University of Toronto
| | - Sarah Johnson
- Department Medical Imaging, University Medical Imaging Toronto, University Health Network – Mount Sinai Hospital – Women College Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Zhihui Amy Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Sareh Keshavarzi
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Veit-Haibach
- Department Medical Imaging, University Medical Imaging Toronto, University Health Network – Mount Sinai Hospital – Women College Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Hasanabadi S, Aghamiri SMR, Abin AA, Abdollahi H, Arabi H, Zaidi H. Enhancing Lymphoma Diagnosis, Treatment, and Follow-Up Using 18F-FDG PET/CT Imaging: Contribution of Artificial Intelligence and Radiomics Analysis. Cancers (Basel) 2024; 16:3511. [PMID: 39456604 PMCID: PMC11505665 DOI: 10.3390/cancers16203511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Lymphoma, encompassing a wide spectrum of immune system malignancies, presents significant complexities in its early detection, management, and prognosis assessment since it can mimic post-infectious/inflammatory diseases. The heterogeneous nature of lymphoma makes it challenging to definitively pinpoint valuable biomarkers for predicting tumor biology and selecting the most effective treatment strategies. Although molecular imaging modalities, such as positron emission tomography/computed tomography (PET/CT), specifically 18F-FDG PET/CT, hold significant importance in the diagnosis of lymphoma, prognostication, and assessment of treatment response, they still face significant challenges. Over the past few years, radiomics and artificial intelligence (AI) have surfaced as valuable tools for detecting subtle features within medical images that may not be easily discerned by visual assessment. The rapid expansion of AI and its application in medicine/radiomics is opening up new opportunities in the nuclear medicine field. Radiomics and AI capabilities seem to hold promise across various clinical scenarios related to lymphoma. Nevertheless, the need for more extensive prospective trials is evident to substantiate their reliability and standardize their applications. This review aims to provide a comprehensive perspective on the current literature regarding the application of AI and radiomics applied/extracted on/from 18F-FDG PET/CT in the management of lymphoma patients.
Collapse
Affiliation(s)
- Setareh Hasanabadi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Seyed Mahmud Reza Aghamiri
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Ahmad Ali Abin
- Faculty of Computer Science and Engineering, Shahid Beheshti University, Tehran 1983969411, Iran;
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, 500 Odense, Denmark
- University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| |
Collapse
|
5
|
Qian C, Jiang C, Xie K, Ding C, Teng Y, Sun J, Gao L, Zhou Z, Ni X. Prognosis Prediction of Diffuse Large B-Cell Lymphoma in 18F-FDG PET Images Based on Multi-Deep-Learning Models. IEEE J Biomed Health Inform 2024; 28:4010-4023. [PMID: 38635387 DOI: 10.1109/jbhi.2024.3390804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL), a cancer of B cells, has been one of the most challenging and complicated diseases because of its considerable variation in clinical behavior, response to therapy, and prognosis. Radiomic features from medical images, such as PET images, have become one of the most valuable features for disease classification or prognosis prediction using learning-based methods. In this paper, a new flexible ensemble deep learning model is proposed for the prognosis prediction of the DLBCL in 18F-FDG PET images. This study proposes the multi-R-signature construction through selected pre-trained deep learning models for predicting progression-free survival (PFS) and overall survival (OS). The proposed method is trained and validated on two datasets from different imaging centers. Through analyzing and comparing the results, the prediction models, including Age, Ann abor stage, Bulky disease, SUVmax, TMTV, and multi-R-signature, achieve the almost best PFS prediction performance (C-index: 0.770, 95% CI: 0.705-0.834, with feature adding fusion method and C-index: 0.764, 95% CI: 0.695-0.832, with feature concatenate fusion method) and OS prediction (C-index: 0.770 (0.692-0.848) and 0.771 (0.694-0.849)) on the validation dataset. The developed multiparametric model could achieve accurate survival risk stratification of DLBCL patients. The outcomes of this study will be helpful for the early identification of high-risk DLBCL patients with refractory relapses and for guiding individualized treatment strategies.
Collapse
|
6
|
Chen M, Rong J, Zhao J, Teng Y, Jiang C, Chen J, Xu J. PET-based radiomic feature based on the cross-combination method for predicting the mid-term efficacy and prognosis in high-risk diffuse large B-cell lymphoma patients. Front Oncol 2024; 14:1394450. [PMID: 38903712 PMCID: PMC11188321 DOI: 10.3389/fonc.2024.1394450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Objectives This study aims to develop 7×7 machine-learning cross-combinatorial methods for selecting and classifying radiomic features used to construct Radiomics Score (RadScore) of predicting the mid-term efficacy and prognosis in high-risk patients with diffuse large B-cell lymphoma (DLBCL). Methods Retrospectively, we recruited 177 high-risk DLBCL patients from two medical centers between October 2012 and September 2022 and randomly divided them into a training cohort (n=123) and a validation cohort (n=54). We finally extracted 110 radiomic features along with SUVmax, MTV, and TLG from the baseline PET. The 49 features selection-classification pairs were used to obtain the optimal LASSO-LASSO model with 11 key radiomic features for RadScore. Logistic regression was employed to identify independent RadScore, clinical and PET factors. These models were evaluated using receiver operating characteristic (ROC) curves and calibration curves. Decision curve analysis (DCA) was conducted to assess the predictive power of the models. The prognostic power of RadScore was assessed using cox regression (COX) and Kaplan-Meier plots (KM). Results 177 patients (mean age, 63 ± 13 years,129 men) were evaluated. Multivariate analyses showed that gender (OR,2.760; 95%CI:1.196,6.368); p=0.017), B symptoms (OR,4.065; 95%CI:1.837,8.955; p=0.001), SUVmax (OR,2.619; 95%CI:1.107,6.194; p=0.028), and RadScore (OR,7.167; 95%CI:2.815,18.248; p<0.001) independently contributed to the risk factors for predicting mid-term outcome. The AUC values of the combined models in the training and validation groups were 0.846 and 0.724 respectively, outperformed the clinical model (0.714;0.556), PET based model (0.664; 0.589), NCCN-IPI model (0.523;0.406) and IPI model (0.510;0.412) in predicting mid-term treatment outcome. DCA showed that the combined model incorporating RadScore, clinical risk factors, and PET metabolic metrics has optimal net clinical benefit. COX indicated that the high RadScore group had worse prognosis and survival in progression-free survival (PFS) (HR, 2.1737,95%CI: 1.2983, 3.6392) and overall survival (OS) (HR,2.1356,95%CI: 1.2561, 3.6309) compared to the low RadScore group. KM survival analysis also showed the same prognosis prediction as Cox results. Conclusion The combined model incorporating RadScore, sex, B symptoms and SUVmax demonstrates a significant enhancement in predicting medium-term efficacy and prognosis in high-risk DLBCL patients. RadScore using 7×7 machine learning cross-combinatorial methods for selection and classification holds promise as a potential method for evaluating medium-term treatment outcome and prognosis in high-risk DLBCL patients.
Collapse
Affiliation(s)
- Man Chen
- Department of Hematology, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jian Rong
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Jincheng Zhao
- Department of Hematology, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue Teng
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chong Jiang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Jingyan Xu
- Department of Hematology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Carlier T, Frécon G, Mateus D, Rizkallah M, Kraeber-Bodéré F, Kanoun S, Blanc-Durand P, Itti E, Le Gouill S, Casasnovas RO, Bodet-Milin C, Bailly C. Prognostic Value of 18F-FDG PET Radiomics Features at Baseline in PET-Guided Consolidation Strategy in Diffuse Large B-Cell Lymphoma: A Machine-Learning Analysis from the GAINED Study. J Nucl Med 2024; 65:156-162. [PMID: 37945379 DOI: 10.2967/jnumed.123.265872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
The results of the GA in Newly Diagnosed Diffuse Large B-Cell Lymphoma (GAINED) study demonstrated the success of an 18F-FDG PET-driven approach to allow early identification-for intensification therapy-of diffuse large B-cell lymphoma patients with a high risk of relapse. Besides, some works have reported the prognostic value of baseline PET radiomics features (RFs). This work investigated the added value of such biomarkers on survival of patients involved in the GAINED protocol. Methods: Conventional PET features and RFs were computed from 18F-FDG PET at baseline and extracted using different volume definitions (patient level, largest lesion, and hottest lesion). Clinical features and the consolidation treatment information were also considered in the model. Two machine-learning pipelines were trained with 80% of patients and tested on the remaining 20%. The training was repeated 100 times to highlight the test set variability. For the 2-y progression-free survival (PFS) outcome, the pipeline included a data augmentation and an elastic net logistic regression model. Results for different feature groups were compared using the mean area under the curve (AUC). For the survival outcome, the pipeline included a Cox univariate model to select the features. Then, the model included a split between high- and low-risk patients using the median of a regression score based on the coefficients of a penalized Cox multivariate approach. The log-rank test P values over the 100 loops were compared with a Wilcoxon signed-ranked test. Results: In total, 545 patients were included for the 2-y PFS classification and 561 for survival analysis. Clinical features alone, consolidation features alone, conventional PET features, and RFs extracted at patient level achieved an AUC of, respectively, 0.65 ± 0.07, 0.64 ± 0.06, 0.60 ± 0.07, and 0.62 ± 0.07 (0.62 ± 0.07 for the largest lesion and 0.54 ± 0.07 for the hottest). Combining clinical features with the consolidation features led to the best AUC (0.72 ± 0.06). Adding conventional PET features or RFs did not improve the results. For survival, the log-rank P values of the model involving clinical and consolidation features together were significantly smaller than all combined-feature groups (P < 0.007). Conclusion: The results showed that a concatenation of multimodal features coupled with a simple machine-learning model does not seem to improve the results in terms of 2-y PFS classification and PFS prediction for patient treated according to the GAINED protocol.
Collapse
Affiliation(s)
- Thomas Carlier
- Nantes Université, INSERM, CNRS, CRCINA, Université d'Angers, Nantes, France
- Nuclear Medicine Department, University Hospital, Nantes, France
| | - Gauthier Frécon
- Nantes Université, INSERM, CNRS, CRCINA, Université d'Angers, Nantes, France
- Nuclear Medicine Department, University Hospital, Nantes, France
| | - Diana Mateus
- Laboratoire des Sciences Numériques de Nantes, Ecole Centrale de Nantes, CNRS UMR 6004, Nantes, France
| | - Mira Rizkallah
- Laboratoire des Sciences Numériques de Nantes, Ecole Centrale de Nantes, CNRS UMR 6004, Nantes, France
| | - Françoise Kraeber-Bodéré
- Nantes Université, INSERM, CNRS, CRCINA, Université d'Angers, Nantes, France
- Nuclear Medicine Department, University Hospital, Nantes, France
| | - Salim Kanoun
- Nuclear Medicine, Georges-François Leclerc Center, Dijon, France
| | - Paul Blanc-Durand
- Nuclear Medicine, CHU Henri Mondor, Paris-Est University, Créteil, France
| | - Emmanuel Itti
- Nuclear Medicine, CHU Henri Mondor, Paris-Est University, Créteil, France
| | - Steven Le Gouill
- Haematology Department, University Hospital, Nantes, France; and
| | | | - Caroline Bodet-Milin
- Nantes Université, INSERM, CNRS, CRCINA, Université d'Angers, Nantes, France
- Nuclear Medicine Department, University Hospital, Nantes, France
| | - Clément Bailly
- Nantes Université, INSERM, CNRS, CRCINA, Université d'Angers, Nantes, France;
- Nuclear Medicine Department, University Hospital, Nantes, France
| |
Collapse
|
8
|
Jing F, Liu Y, Zhao X, Wang N, Dai M, Chen X, Zhang Z, Zhang J, Wang J, Wang Y. Baseline 18F-FDG PET/CT radiomics for prognosis prediction in diffuse large B cell lymphoma. EJNMMI Res 2023; 13:92. [PMID: 37884763 PMCID: PMC10603012 DOI: 10.1186/s13550-023-01047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma in adults. Standard treatment includes chemoimmunotherapy with R-CHOP or similar regimens. Despite treatment advancements, many patients with DLBCL experience refractory disease or relapse. While baseline 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) parameters have shown promise in predicting survival, they may not fully capture lesion heterogeneity. This study aimed to assess the prognostic value of baseline 18F-FDG PET radiomics features in comparison with clinical factors and metabolic parameters for assessing 2-year progression-free survival (PFS) and 5-year overall survival (OS) in patients with DLBCL. RESULTS A total of 201 patients with DLBCL were enrolled in this study, and 1328 radiomics features were extracted. The radiomics signatures, clinical factors, and metabolic parameters showed significant prognostic value for individualized prognosis prediction in patients with DLBCL. Radiomics signatures showed the lowest Akaike information criterion (AIC) value and highest Harrell's concordance index (C-index) value in comparison with clinical factors and metabolic parameters for both PFS (AIC: 571.688 vs. 596.040 vs. 576.481; C-index: 0.732 vs. 0.658 vs. 0.702, respectively) and OS (AIC: 339.843 vs. 363.671 vs. 358.412; C-index: 0.759 vs. 0.667 vs. 0.659, respectively). Statistically significant differences were observed in the area under the curve (AUC) values between the radiomics signatures and clinical factors for both PFS (AUC: 0.768 vs. 0.681, P = 0.017) and OS (AUC: 0.767 vs. 0.667, P = 0.023). For OS, the AUC of the radiomics signatures were significantly higher than those of metabolic parameters (AUC: 0.767 vs. 0.688, P = 0.007). However, for PFS, no significant difference was observed between the radiomics signatures and metabolic parameters (AUC: 0.768 vs. 0.756, P = 0.654). The combined model and the best-performing individual model (radiomics signatures) alone showed no significant difference for both PFS (AUC: 0.784 vs. 0.768, P = 0.163) or OS (AUC: 0.772 vs. 0.767, P = 0.403). CONCLUSIONS Radiomics signatures derived from PET images showed the high predictive power for progression in patients with DLBCL. The combination of radiomics signatures, clinical factors, and metabolic parameters may not significantly improve predictive value beyond that of radiomics signatures alone.
Collapse
Affiliation(s)
- Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China.
| | - Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| | - Yingchen Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
9
|
Zhou Y, Zhang B, Han J, Dai N, Jia T, Huang H, Deng S, Sang S. Development of a radiomic-clinical nomogram for prediction of survival in patients with diffuse large B-cell lymphoma treated with chimeric antigen receptor T cells. J Cancer Res Clin Oncol 2023; 149:11549-11560. [PMID: 37395846 DOI: 10.1007/s00432-023-05038-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND In our current work, an 18F-FDG PET/CT radiomics-based model was developed to assess the progression-free survival (PFS) and overall survival (OS) of patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) who received chimeric antigen receptor (CAR)-T cell therapy. METHODS A total of 61 DLBCL cases receiving 18F-FDG PET/CT before CAR-T cell infusion were included in the current analysis, and these patients were randomly assigned to a training cohort (n = 42) and a validation cohort (n = 19). Radiomic features from PET and CT images were obtained using LIFEx software, and radiomics signatures (R-signatures) were then constructed by choosing the optimal parameters according to their PFS and OS. Subsequently, the radiomics model and clinical model were constructed and validated. RESULTS The radiomics model that integrated R-signatures and clinical risk factors showed superior prognostic performance compared with the clinical models in terms of both PFS (C-index: 0.710 vs. 0.716; AUC: 0.776 vs. 0.712) and OS (C-index: 0.780 vs. 0.762; AUC: 0.828 vs. 0.728). For validation, the C-index of the two approaches was 0.640 vs. 0.619 and 0.676 vs. 0.699 for predicting PFS and OS, respectively. Moreover, the AUC was 0.886 vs. 0.635 and 0.778 vs. 0.705, respectively. The calibration curves indicated good agreement, and the decision curve analysis suggested that the net benefit of radiomics models was higher than that of clinical models. CONCLUSIONS PET/CT-derived R-signature could be a potential prognostic biomarker for R/R DLBCL patients undergoing CAR-T cell therapy. Moreover, the risk stratification could be further enhanced when the PET/CT-derived R-signature was combined with clinical factors.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jiangqin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Na Dai
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Tongtong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haiwen Huang
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
10
|
Travaini LL, Botta F, Derenzini E, Lo Presti G, Ferrari ME, Airò Farulla LS, Radice T, Mazzara S, Tarella C, Pileri S, Raimondi S, Ceci F. [ 18 F]-FDG PET radiomic model as prognostic biomarker in diffuse large B-cell lymphoma. Hematol Oncol 2023; 41:674-682. [PMID: 37209024 DOI: 10.1002/hon.3171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/13/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023]
Abstract
To evaluate the association between radiomic features (RFs) extracted from 18 F-FDG PET/CT (18 F-FDG-PET) with progression-free survival (PFS) and overall survival (OS) in diffuse large-B-cell lymphoma (DLBCL) patients eligible to first-line chemotherapy. DLBCL patients who underwent 18 F-FDG-PET prior to first-line chemotherapy were retrospectively analyzed. RFs were extracted from the lesion showing the highest uptake. A radiomic score to predict PFS and OS was obtained by multivariable Elastic Net Cox model. Radiomic univariate model, clinical and combined clinical-radiomic multivariable models to predict PFS and OS were obtained. 112 patients were analyzed. Median follow-up was 34.7 months (Inter-Quartile Range (IQR) 11.3-66.3 months) for PFS and 41.1 (IQR 18.4-68.9) for OS. Radiomic score resulted associated with PFS and OS (p < 0.001), outperforming conventional PET parameters. C-index (95% CI) for PFS prediction were 0.67 (0.58-0.76), 0.81 (0.75-0.88) and 0.84 (0.77-0.91) for clinical, radiomic and combined clinical-radiomic model, respectively. C-index for OS were 0.77 (0.66-0.89), 0.84 (0.76-0.91) and 0.90 (0.81-0.98). In the Kaplan-Meier analysis (low-IPI vs. high-IPI), the radiomic score was significant predictor of PFS (p < 0.001). The radiomic score was an independent prognostic biomarker of survival in DLBCL patients. The extraction of RFs from baseline 18 F-FDG-PET might be proposed in DLBCL to stratify high-risk versus low-risk patients of relapse after first-line therapy, especially in low-IPI patients.
Collapse
Affiliation(s)
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Enrico Derenzini
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Giuliana Lo Presti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | - Lighea Simona Airò Farulla
- Division of Nuclear Medicine, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Tommaso Radice
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Saveria Mazzara
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Corrado Tarella
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Stefano Pileri
- Haemolymphopathology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Zanoni L, Bezzi D, Nanni C, Paccagnella A, Farina A, Broccoli A, Casadei B, Zinzani PL, Fanti S. PET/CT in Non-Hodgkin Lymphoma: An Update. Semin Nucl Med 2023; 53:320-351. [PMID: 36522191 DOI: 10.1053/j.semnuclmed.2022.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022]
Abstract
Non-Hodgkin lymphomas represents a heterogeneous group of lymphoproliferative disorders characterized by different clinical courses, varying from indolent to highly aggressive. 18F-FDG-PET/CT is the current state-of-the-art diagnostic imaging, for the staging, restaging and evaluation of response to treatment in lymphomas with avidity for 18F-FDG, despite it is not routinely recommended for surveillance. PET-based response criteria (using five-point Deauville Score) are nowadays uniformly applied in FDG-avid lymphomas. In this review, a comprehensive overview of the role of 18F-FDG-PET in Non-Hodgkin lymphomas is provided, at each relevant point of patient management, particularly focusing on recent advances on diffuse large B-cell lymphoma and follicular lymphoma, with brief updates also on other histotypes (such as marginal zone, mantle cell, primary mediastinal- B cell lymphoma and T cell lymphoma). PET-derived semiquantitative factors useful for patient stratification and prognostication and emerging radiomics research are also presented.
Collapse
Affiliation(s)
- Lucia Zanoni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Davide Bezzi
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Paccagnella
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy; Nuclear Medicine Unit, AUSL Romagna, Cesena, Italy
| | - Arianna Farina
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Alessandro Broccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Pant S, Kang SR, Lee M, Phuc PS, Yang HJ, Yang DH. Survival Prediction Using Transformer-Based Categorical Feature Representation in the Treatment of Diffuse Large B-Cell Lymphoma. Healthcare (Basel) 2023; 11:healthcare11081171. [PMID: 37108006 PMCID: PMC10137756 DOI: 10.3390/healthcare11081171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a common and aggressive subtype of lymphoma, and accurate survival prediction is crucial for treatment decisions. This study aims to develop a robust survival prediction strategy to integrate various risk factors effectively, including clinical risk factors and Deauville scores in positron-emission tomography/computed tomography at different treatment stages using a deep-learning-based approach. We conduct a multi-institutional study on 604 DLBCL patients' clinical data and validate the model on 220 patients from an independent institution. We propose a survival prediction model using transformer architecture and a categorical-feature-embedding technique that can handle high-dimensional and categorical data. Comparison with deep-learning survival models such as DeepSurv, CoxTime, and CoxCC based on the concordance index (C-index) and the mean absolute error (MAE) demonstrates that the categorical features obtained using transformers improved the MAE and the C-index. The proposed model outperforms the best-performing existing method by approximately 185 days in terms of the MAE for survival time estimation on the testing set. Using the Deauville score obtained during treatment resulted in a 0.02 improvement in the C-index and a 53.71-day improvement in the MAE, highlighting its prognostic importance. Our deep-learning model could improve survival prediction accuracy and treatment personalization for DLBCL patients.
Collapse
Affiliation(s)
- Sudarshan Pant
- Department of Artificial Intelligence Convergence, Chonnam National University, Buk-gu, Gwangju 61186, Republic of Korea
| | - Sae-Ryung Kang
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea
| | - Minhee Lee
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea
| | - Pham-Sy Phuc
- Department of Artificial Intelligence Convergence, Chonnam National University, Buk-gu, Gwangju 61186, Republic of Korea
| | - Hyung-Jeong Yang
- Department of Artificial Intelligence Convergence, Chonnam National University, Buk-gu, Gwangju 61186, Republic of Korea
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea
| |
Collapse
|
13
|
Deng H, Zhou Y, Lu W, Chen W, Yuan Y, Li L, Shu H, Zhang P, Ye X. Development and validation of nomograms by radiomic features on ultrasound imaging for predicting overall survival in patients with primary nodal diffuse large B-cell lymphoma. Front Oncol 2022; 12:991948. [PMID: 36568168 PMCID: PMC9768489 DOI: 10.3389/fonc.2022.991948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives To develop and validate a nomogram to predict the overall survival (OS) of patients with primary nodal diffuse large B-cell lymphoma(N-DLBCL) based on radiomic features and clinical features. Materials and methods A retrospective analysis was performed on 145 patients confirmed with N-DLBCL and they were randomly assigned to training set(n=78), internal validation set(n=33), external validation set(n=34). First, a clinical model (model 1) was established according to clinical features and ultrasound (US) results. Then, based on the radiomics features extracted from conventional ultrasound images, a radiomic signature was constructed (model 2), and the radiomics score (Rad-Score) was calculated. Finally, a comprehensive model was established (model 3) combined with Rad-score and clinical features. Receiver operating characteristic (ROC) curves were employed to evaluate the performance of model 1, model 2 and model 3. Based on model 3, we plotted a nomogram. Calibration curves were used to test the effectiveness of the nomogram, and decision curve analysis (DCA) was used to asset the nomogram in clinical use. Results According to multivariate analysis, 3 clinical features and Rad-score were finally selected to construct the model 3, which showed better predictive value for OS in patients with N-DLBCL than mode 1 and model 2 in training (AUC,0. 891 vs. 0.779 vs.0.756), internal validation (AUC, 0.868 vs. 0.713, vs.0.756) and external validation (AUC, 914 vs. 0.866, vs.0.789) sets. Decision curve analysis demonstrated that the nomogram based on model 3 was more clinically useful than the other two models. Conclusion The developed nomogram is a useful tool for precisely analyzing the prognosis of N-DLBCL patients, which could help clinicians in making personalized survival predictions and assessing individualized clinical options.
Collapse
Affiliation(s)
- Hongyan Deng
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yasu Zhou
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjuan Lu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenqin Chen
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ya Yuan
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hua Shu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pingyang Zhang
- Department of Cardiovascular Ultrasound, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xinhua Ye, ; Pingyang Zhang,
| | - Xinhua Ye
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xinhua Ye, ; Pingyang Zhang,
| |
Collapse
|
14
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 2, Infradiaphragmatic Cancers, Blood Malignancies, Melanoma and Musculoskeletal Cancers. Diagnostics (Basel) 2022; 12:diagnostics12061330. [PMID: 35741139 PMCID: PMC9222024 DOI: 10.3390/diagnostics12061330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
The objective of this review was to summarize published radiomics studies dealing with infradiaphragmatic cancers, blood malignancies, melanoma, and musculoskeletal cancers, and assess their quality. PubMed database was searched from January 1990 to February 2022 for articles performing radiomics on PET imaging of at least 1 specified tumor type. Exclusion criteria includd: non-oncological studies; supradiaphragmatic tumors; reviews, comments, cases reports; phantom or animal studies; technical articles without a clinically oriented question; studies including <30 patients in the training cohort. The review database contained PMID, first author, year of publication, cancer type, number of patients, study design, independent validation cohort and objective. This database was completed twice by the same person; discrepant results were resolved by a third reading of the articles. A total of 162 studies met inclusion criteria; 61 (37.7%) studies included >100 patients, 13 (8.0%) were prospective and 61 (37.7%) used an independent validation set. The most represented cancers were esophagus, lymphoma, and cervical cancer (n = 24, n = 24 and n = 19 articles, respectively). Most studies focused on 18F-FDG, and prognostic and response to treatment objectives. Although radiomics and artificial intelligence are technically challenging, new contributions and guidelines help improving research quality over the years and pave the way toward personalized medicine.
Collapse
Affiliation(s)
- David Morland
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
- Correspondence:
| | - Elizabeth Katherine Anna Triumbari
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
15
|
Jiang C, Huang X, Li A, Teng Y, Ding C, Chen J, Xu J, Zhou Z. Radiomics signature from [ 18F]FDG PET images for prognosis predication of primary gastrointestinal diffuse large B cell lymphoma. Eur Radiol 2022; 32:5730-5741. [PMID: 35298676 DOI: 10.1007/s00330-022-08668-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To investigate the prognostic value of PET radiomics feature in the prognosis of patients with primary gastrointestinal diffuse large B cell lymphoma (PGI-DLBCL) treated with R-CHOP-like regimen. METHODS A total of 140 PGI-DLBCL patients who underwent pre-therapy [18F] FDG PET/CT were enrolled in this retrospective analysis. PET radiomics features obtained from patients in the training cohort were subjected to three machine learning methods and Pearson's correlation test for feature selection. Support vector machine (SVM) was used to build a radiomics signature classifier associated with progression-free survival (PFS) and overall survival (OS). A multivariate Cox proportional hazards regression model was established to predict survival outcomes. RESULTS A total of 1421 PET radiomics features were extracted and reduced to 5 features to build a radiomics signature which was significantly associated with PFS and OS (p < 0.05). The combined model incorporating radiomics signatures, metabolic metrics, and clinical risk factors showed high C-indices in both the training (PFS: 0.825, OS: 0.834) and validation sets (PFS: 0.831, OS: 0.877). Decision curve analysis (DCA) demonstrated that the combined models achieved the most net benefit across a wider reasonable range of threshold probabilities for predicting PFS and OS. CONCLUSION The newly developed radiomics signatures obtained by the ensemble strategy were independent predictors of PFS and OS for PGI-DLBCL patients. Moreover, the combined model with clinical and metabolic factors was able to predict patient prognosis and may enable personalized treatment decision-making. KEY POINTS • Radiomics signatures generated from the optimal radiomics feature set from the [18F]FDG PET images can predict the survival of PGI-DLBCL patients. • The optimal radiomics feature set is constructed by integrating the feature selection outputs of LASSO, RF, Xgboost, and PC methods. • Combined models incorporating radiomics signatures from18F-FDG PET images, metabolic parameters, and clinical factors outperformed clinical models, and NCCN-IPI.
Collapse
Affiliation(s)
- Chong Jiang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Xiangjun Huang
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Ang Li
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yue Teng
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Chongyang Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jianxin Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Jingyan Xu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321, Zhongshan Road, Nanjing City, Jiangsu Province, 210008, China.
| | - Zhengyang Zhou
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| |
Collapse
|
16
|
Jiang C, Li A, Teng Y, Huang X, Ding C, Chen J, Xu J, Zhou Z. Optimal PET-based radiomic signature construction based on the cross-combination method for predicting the survival of patients with diffuse large B-cell lymphoma. Eur J Nucl Med Mol Imaging 2022; 49:2902-2916. [PMID: 35146578 DOI: 10.1007/s00259-022-05717-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE To develop and externally validate models incorporating a PET radiomics signature (R-signature) obtained by the cross-combination method for predicting the survival of patients with diffuse large B-cell lymphoma (DLBCL). METHODS A total of 383 patients with DLBCL from two medical centres between 2011 and 2019 were included. The cross-combination method was used on three types of PET radiomics features from the training cohort to generate 49 feature selection-classification candidates based on 7 different machine learning models. The R-signature was then built by selecting the optimal candidates based on their progression-free survival (PFS) and overall survival (OS). Cox regression analysis was used to develop the survival prediction models. The calibration, discrimination, and clinical utility of the models were assessed and externally validated. RESULTS The R-signatures determined by 12 and 31 radiomics features were significantly associated with PFS and OS, respectively (P<0.05). The combined models that incorporated R-signatures, metabolic metrics, and clinical risk factors exhibited significant prognostic superiority over the clinical models, PET-based models, and the National Comprehensive Cancer Network International Prognostic Index in terms of both PFS (C-index: 0.801 vs. 0.732 vs. 0.785 vs. 0.720, respectively) and OS (C-index: 0.807 vs. 0.740 vs. 0.773 vs. 0.726, respectively). For external validation, the C-indices were 0.758 vs. 0.621 vs. 0.732 vs. 0.673 and 0.794 vs. 0.696 vs. 0.781 vs. 0.708 in the PFS and OS analyses, respectively. The calibration curves showed good consistency, and the decision curve analysis supported the clinical utility of the combined model. CONCLUSION The R-signature could be used as a survival predictor for DLBCL, and its combination with clinical factors may allow for accurate risk stratification.
Collapse
Affiliation(s)
- Chong Jiang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Ang Li
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yue Teng
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Xiangjun Huang
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Chongyang Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jianxin Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Jingyan Xu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| | - Zhengyang Zhou
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| |
Collapse
|
17
|
Zhou Y, Li J, Zhang X, Jia T, Zhang B, Dai N, Sang S, Deng S. Prognostic Value of Radiomic Features of 18F-FDG PET/CT in Patients With B-Cell Lymphoma Treated With CD19/CD22 Dual-Targeted Chimeric Antigen Receptor T Cells. Front Oncol 2022; 12:834288. [PMID: 35198451 PMCID: PMC8858981 DOI: 10.3389/fonc.2022.834288] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022] Open
Abstract
ObjectiveIn the present study, we aimed to evaluate the prognostic value of PET/CT-derived radiomic features for patients with B-cell lymphoma (BCL), who were treated with CD19/CD22 dual-targeted chimeric antigen receptor (CAR) T cells. Moreover, we explored the relationship between baseline radiomic features and the occurrence probability of cytokine release syndrome (CRS).MethodsA total of 24 BCL patients who received 18F-FDG PET/CT before CAR T-cell infusion were enrolled in the present study. Radiomic features from PET and CT images were extracted using LIFEx software, and the least absolute shrinkage and selection operator (LASSO) regression was used to select the most useful predictive features of progression-free survival (PFS) and overall survival (OS). Receiver operating characteristic curves, Cox proportional hazards model, and Kaplan-Meier curves were conducted to assess the potential prognostic value.ResultsContrast extracted from neighbourhood grey-level different matrix (NGLDM) was an independent predictor of PFS (HR = 15.16, p = 0.023). MYC and BCL2 double-expressor (DE) was of prognostic significance for PFS (HR = 7.02, p = 0.047) and OS (HR = 10.37, p = 0.041). The combination of NGLDM_ContrastPET and DE yielded three risk groups with zero (n = 7), one (n = 11), or two (n = 6) factors (p < 0.0001 and p = 0.0004, for PFS and OS), respectively. The PFS was 85.7%, 63.6%, and 0%, respectively, and the OS was 100%, 90.9%, and 16.7%, respectively. Moreover, there was no significant association between PET/CT variables and CRS.ConclusionsIn conclusion, radiomic features extracted from baseline 18F-FDG PET/CT images in combination with genomic factors could predict the survival outcomes of BCL patients receiving CAR T-cell therapy.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jihui Li
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Changshu No. 2 People’s Hospital, Changshu, China
| | - Tongtong Jia
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Na Dai
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shibiao Sang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Shengming Deng, ; Shibiao Sang,
| | - Shengming Deng
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
- *Correspondence: Shengming Deng, ; Shibiao Sang,
| |
Collapse
|
18
|
Jiang H, Li A, Ji Z, Tian M, Zhang H. Role of Radiomics-Based Baseline PET/CT Imaging in Lymphoma: Diagnosis, Prognosis, and Response Assessment. Mol Imaging Biol 2022; 24:537-549. [PMID: 35031945 DOI: 10.1007/s11307-022-01703-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
Radiomic analysis provides information on the underlying tumour heterogeneity in lymphoma, reflecting the real-time evolution of malignancy. 2-Deoxy-2-[18F] fluoro-D-glucose positron emission tomography ([18F] FDG PET/CT) imaging is recommended before, during, and at the end of treatment for almost all lymphoma patients. This methodology offers high specificity and sensitivity, which can aid in accurate staging and assist in prompt treatment. Pretreatment [18F] FDG PET/CT-based radiomics facilitates improved diagnostic ability, guides individual treatment regimens, and boosts outcome prognosis based on heterogeneity as well as the biological, pathological, and metabolic status of the lymphoma. This technique has attracted considerable attention given its numerous applications in medicine. In the current review, we will briefly describe the basic radiomics workflow and types of radiomic features. Details of current applications of baseline [18F] FDG PET/CT-based radiomics in lymphoma will be discussed, such as differential diagnosis from other primary malignancies, diagnosis of bone marrow involvement, and response and prognostic prediction. We will also describe how this technique provides a unique noninvasive platform to assess tumour heterogeneity. Newly emerging PET radiotracers and multimodality technology will improve diagnostic specificity and further clarify tumor biology and even genetic variations in lymphoma, potentially promoting the development of precision medicine.
Collapse
Affiliation(s)
- Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ang Li
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Zhongyou Ji
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, 8 Hangzhou, Hangzhou, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, 8 Hangzhou, Hangzhou, China. .,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
PET imaging of lymphomas. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00047-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
20
|
Radiomic Features of 18F-FDG PET in Hodgkin Lymphoma Are Predictive of Outcomes. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:6347404. [PMID: 34887712 PMCID: PMC8629643 DOI: 10.1155/2021/6347404] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/10/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
Purpose In the present study, we aimed to investigate whether the radiomic features of baseline 18F-FDG PET can predict the prognosis of Hodgkin lymphoma (HL). Methods A total 65 HL patients (training cohort: n = 49; validation cohort: n = 16) were retrospectively enrolled in the present study. A total of 47 radiomic features were extracted from pretreatment PET images. The least absolute shrinkage and selection operator (LASSO) regression was used to select the most useful prognostic features in the training cohort. The distance between the two lesions that were the furthest apart (Dmax) was recorded. The receiver operating characteristic (ROC) curve, Kaplan–Meier method, and Cox proportional hazards model were used to assess the prognostic factors. Results Long-zone high gray-level emphasis extracted from a gray-level zone-length matrix (LZHGEGLZLM) (HR = 9.007; p=0.044) and Dmax (HR = 3.641; p=0.048) were independently correlated with 2-year progression-free survival (PFS). A prognostic stratification model was established based on both risk predictors, which could distinguish three risk categories for PFS (p=0.0002). The 2-year PFS was 100.0%, 64.7%, and 33.3%, respectively. Conclusions LZHGEGLZLM and Dmax were independent prognostic factors for survival outcomes. Besides, we proposed a prognostic stratification model that could further improve the risk stratification of HL patients.
Collapse
|
21
|
Diagnostic value of baseline 18FDG PET/CT skeletal textural features in follicular lymphoma. Sci Rep 2021; 11:23812. [PMID: 34893676 PMCID: PMC8664828 DOI: 10.1038/s41598-021-03278-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/17/2021] [Indexed: 01/06/2023] Open
Abstract
At present, 18F-fluorodesoxyglucose (18FDG) positron emission tomography (PET)/computed tomography (CT) cannot be used to omit a bone marrow biopsy (BMB) among initial staging procedures in follicular lymphoma (FL). The additional diagnostic value of skeletal textural features on baseline 18FDG-PET/CT in diffuse large B-cell lymphoma (DLBCL) patients has given promising results. The aim of this study is to evaluate the value of 18FDG-PET/CT radiomics for the diagnosis of bone marrow involvement (BMI) in FL patients. This retrospective bicentric study enrolled newly diagnosed FL patients addressed for baseline 18FDG PET/CT. For visual assessment, examinations were considered positive in cases of obvious bone focal uptakes. For textural analysis, the skeleton volumes of interest (VOIs) were automatically extracted from segmented CT images and analysed using LifeX software. BMB and visual assessment were taken as the gold standard: BMB −/PET − patients were considered as bone-NEGATIVE patients, whereas BMB +/PET −, BMB −/PET + and BMB +/PET + patients were considered bone-POSITIVE patients. A LASSO regression algorithm was used to select features of interest and to build a prediction model. Sixty-six consecutive patients were included: 36 bone-NEGATIVE (54.5%) and 30 bone-POSITIVE (45.5%). The LASSO regression found variance_GLCM, correlation_GLCM, joint entropy_GLCM and busyness_NGLDM to have nonzero regression coefficients. Based on ROC analysis, a cut-off equal to − 0.190 was found to be optimal for the diagnosis of BMI using PET pred.score. The corresponding sensitivity, specificity, PPV and NPV values were equal to 70.0%, 83.3%, 77.8% and 76.9%, respectively. When comparing the ROC AUCs with using BMB alone, visual PET assessment or PET pred.score, a significant difference was found between BMB versus visual PET assessments (p = 0.010) but not between BMB and PET pred.score assessments (p = 0.097). Skeleton texture analysis is worth exploring to improve the performance of 18FDG-PET/CT for the diagnosis of BMI at baseline in FL patients.
Collapse
|
22
|
Ceriani L, Milan L, Cascione L, Gritti G, Dalmasso F, Esposito F, Pirosa MC, Schär S, Bruno A, Dirnhofer S, Giovanella L, Hayoz S, Mamot C, Rambaldi A, Chauvie S, Zucca E. Generation and validation of a PET radiomics model that predicts survival in diffuse large B cell lymphoma treated with R-CHOP14: A SAKK 38/07 trial post-hoc analysis. Hematol Oncol 2021; 40:11-21. [PMID: 34714558 DOI: 10.1002/hon.2935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
Functional parameters from positron emission tomography (PET) seem promising biomarkers in various lymphoma subtypes. This study investigated the prognostic value of PET radiomics in diffuse large B-cell lymphoma (DLBCL) patients treated with R-CHOP given either every 14 (testing set) or 21 days (validation set). Using the PyRadiomics Python package, 107 radiomics features were extracted from baseline PET scans of 133 patients enrolled in the Swiss Group for Clinical Cancer Research 38/07 prospective clinical trial (SAKK 38/07) [ClinicalTrial.gov identifier: NCT00544219]. The international prognostic indices, the main clinical parameters and standard PET metrics, together with 52 radiomics uncorrelated features (selected using the Spearman correlation test) were included in a least absolute shrinkage and selection operator (LASSO) Cox regression to assess their impact on progression-free (PFS), cause-specific (CSS), and overall survival (OS). A linear combination of the resulting parameters generated a prognostic radiomics score (RS) whose area under the curve (AUC) was calculated by receiver operating characteristic analysis. The RS efficacy was validated in an independent cohort of 107 DLBCL patients. LASSO Cox regression identified four radiomics features predicting PFS in SAKK 38/07. The derived RS showed a significant capability to foresee PFS in both testing (AUC, 0.709; p < 0.001) and validation (AUC, 0.706; p < 0.001) sets. RS was significantly associated also with CSS and OS in testing (CSS: AUC, 0.721; p < 0.001; OS: AUC, 0.740; p < 0.001) and validation (CSS: AUC, 0.763; p < 0.0001; OS: AUC, 0.703; p = 0.004) sets. The RS allowed risk classification of patients with significantly different PFS, CSS, and OS in both cohorts showing better predictive accuracy respect to clinical international indices. PET-derived radiomics may improve the prediction of outcome in DLBCL patients.
Collapse
Affiliation(s)
- Luca Ceriani
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Faculty of Biomedical Sciences, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Lisa Milan
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Luciano Cascione
- Faculty of Biomedical Sciences, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.,SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Giuseppe Gritti
- Hematology Unit, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Fabiana Esposito
- Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Maria Cristina Pirosa
- Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Sämi Schär
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Andrea Bruno
- Department of Nuclear Medicine, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Stephan Dirnhofer
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Luca Giovanella
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stefanie Hayoz
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Christoph Mamot
- Division of Oncology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Alessandro Rambaldi
- Hematology Unit, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stephane Chauvie
- Medical Physics Unit, Santa Croce e Carlo Hospital, Cuneo, Italy
| | - Emanuele Zucca
- Faculty of Biomedical Sciences, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.,Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,Department of Medical Oncology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Al Tabaa Y, Bailly C, Kanoun S. FDG-PET/CT in Lymphoma: Where Do We Go Now? Cancers (Basel) 2021; 13:cancers13205222. [PMID: 34680370 PMCID: PMC8533807 DOI: 10.3390/cancers13205222] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 01/06/2023] Open
Abstract
18F-fluorodeoxyglucose positron emission tomography combined with computed tomography (FDG-PET/CT) is an essential part of the management of patients with lymphoma at staging and response evaluation. Efforts to standardize PET acquisition and reporting, including the 5-point Deauville scale, have enabled PET to become a surrogate for treatment success or failure in common lymphoma subtypes. This review summarizes the key clinical-trial evidence that supports PET-directed personalized approaches in lymphoma but also points out the potential place of innovative PET/CT metrics or new radiopharmaceuticals in the future.
Collapse
Affiliation(s)
- Yassine Al Tabaa
- Scintidoc Nuclear Medicine Center, 25 rue de Clémentville, 34070 Montpellier, France
- Correspondence:
| | - Clement Bailly
- CRCINA, INSERM, CNRS, Université d’Angers, Université de Nantes, 44093 Nantes, France;
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
| | - Salim Kanoun
- Nuclear Medicine Department, Institute Claudius Regaud, 31100 Toulouse, France;
- Cancer Research Center of Toulouse (CRCT), Team 9, INSERM UMR 1037, 31400 Toulouse, France
| |
Collapse
|
24
|
Zhang X, Chen L, Jiang H, He X, Feng L, Ni M, Ma M, Wang J, Zhang T, Wu S, Zhou R, Jin C, Zhang K, Qian W, Chen Z, Zhuo C, Zhang H, Tian M. A novel analytic approach for outcome prediction in diffuse large B-cell lymphoma by [ 18F]FDG PET/CT. Eur J Nucl Med Mol Imaging 2021; 49:1298-1310. [PMID: 34651227 PMCID: PMC8921097 DOI: 10.1007/s00259-021-05572-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/22/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE This study aimed to develop a novel analytic approach based on 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography ([18F]FDG PET/CT) radiomic signature (RS) and International Prognostic Index (IPI) to predict the progression-free survival (PFS) and overall survival (OS) of patients with diffuse large B-cell lymphoma (DLBCL). METHODS We retrospectively enrolled 152 DLBCL patients and divided them into a training cohort (n = 100) and a validation cohort (n = 52). A total of 1245 radiomic features were extracted from the total metabolic tumor volume (TMTV) and the metabolic bulk volume (MBV) of pre-treatment PET/CT images. The least absolute shrinkage and selection operator (LASSO) algorithm was applied to develop the RS. Cox regression analysis was used to construct hybrid nomograms based on different RS and clinical variables. The performances of hybrid nomograms were evaluated using the time-dependent receiver operator characteristic (ROC) curve and the Hosmer-Lemeshow test. The clinical utilities of prediction nomograms were determined via decision curve analysis. The predictive efficiency of different RS, clinical variables, and hybrid nomograms was compared. RESULTS The RS and IPI were identified as independent predictors of PFS and OS, and were selected to construct hybrid nomograms. Both TMTV- and MBV-based hybrid nomograms had significantly higher values of area under the curve (AUC) than IPI in training and validation cohorts (all P < 0.05), while no significant difference was found between TMTV- and MBV-based hybrid nomograms (P > 0.05). The Hosmer-Lemeshow test showed that both TMTV- and MBV-based hybrid nomograms calibrated well in the training and validation cohorts (all P > 0.05). Decision curve analysis indicated that hybrid nomograms had higher net benefits than IPI. CONCLUSION The hybrid nomograms combining RS with IPI could significantly improve survival prediction in DLBCL. Radiomic analysis on MBV may serve as a potential approach for prognosis assessment in DLBCL. TRIAL REGISTRATION NCT04317313. Registered March 16, 2020. Public site: https://clinicaltrials.gov/ct2/show/NCT04317313.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Lin Chen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xuexin He
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Liu Feng
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Miaoqi Ni
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Mindi Ma
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Teng Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Kai Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zexin Chen
- Department of Clinical Epidemiology & Biostatistics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhuo
- College of Information Science & Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China. .,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
25
|
|
26
|
Prognostic Value of Combing Primary Tumor and Nodal Glycolytic-Volumetric Parameters of 18F-FDG PET in Patients with Non-Small Cell Lung Cancer and Regional Lymph Node Metastasis. Diagnostics (Basel) 2021; 11:diagnostics11061065. [PMID: 34207763 PMCID: PMC8228685 DOI: 10.3390/diagnostics11061065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
We investigated whether the combination of primary tumor and nodal 18F-FDG PET parameters predict survival outcomes in patients with nodal metastatic non-small cell lung cancer (NSCLC) without distant metastasis. We retrospectively extracted pre-treatment 18F-FDG PET parameters from 89 nodal-positive NSCLC patients (stage IIB–IIIC). The Cox proportional hazard model was used to identify independent prognosticators of overall survival (OS) and progression-free survival (PFS). We devised survival stratification models based on the independent prognosticators and compared the model to the American Joint Committee on Cancer (AJCC) staging system using Harrell’s concordance index (c-index). Our results demonstrated that total TLG (the combination of primary tumor and nodal total lesion glycolysis) and age were independent risk factors for unfavorable OS (p < 0.001 and p = 0.001) and PFS (both p < 0.001), while the Eastern Cooperative Oncology Group scale independently predicted poor OS (p = 0.022). Our models based on the independent prognosticators outperformed the AJCC staging system (c-index = 0.732 versus 0.544 for OS and c-index = 0.672 versus 0.521 for PFS, both p < 0.001). Our results indicate that incorporating total TLG with clinical factors may refine risk stratification in nodal metastatic NSCLC patients and may facilitate tailored therapeutic strategies in this patient group.
Collapse
|
27
|
Michalet M, Azria D, Tardieu M, Tibermacine H, Nougaret S. Radiomics in radiation oncology for gynecological malignancies: a review of literature. Br J Radiol 2021; 94:20210032. [PMID: 33882246 DOI: 10.1259/bjr.20210032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Radiomics is the extraction of a significant number of quantitative imaging features with the aim of detecting information in correlation with useful clinical outcomes. Features are extracted, after delineation of an area of interest, from a single or a combined set of imaging modalities (including X-ray, US, CT, PET/CT and MRI). Given the high dimensionality, the analytical process requires the use of artificial intelligence algorithms. Firstly developed for diagnostic performance in radiology, it has now been translated to radiation oncology mainly to predict tumor response and patient outcome but other applications have been developed such as dose painting, prediction of side-effects, and quality assurance. In gynecological cancers, most studies have focused on outcomes of cervical cancers after chemoradiation. This review highlights the role of this new tool for the radiation oncologists with particular focus on female GU oncology.
Collapse
Affiliation(s)
- Morgan Michalet
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Univ Montpellier, Montpellier, France.,INSERM U1194 IRCM, Montpellier, France
| | - David Azria
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Univ Montpellier, Montpellier, France.,INSERM U1194 IRCM, Montpellier, France
| | | | | | | |
Collapse
|