1
|
Ceriani L, Milan L, Chauvie S, Zucca E. Understandings 18 FDG PET radiomics and its application to lymphoma. Br J Haematol 2025. [PMID: 40230306 DOI: 10.1111/bjh.20074] [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: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
The early identification of lymphoma patients who fail front-line treatment is crucial for optimizing disease management. Positron emission tomography, a well-established tool for staging and response evaluation in lymphoma, is typically assessed visually or semiquantitatively, leaving much of its latent information unexploited. Radiomic analysis, which employs mathematical descriptors, can enable the extraction of quantitative features from baseline images that correlate with the disease's biological characteristics. Emerging radiomic features such as metabolic tumour volume, total lesion glycolysis and markers of disease dissemination and metabolic heterogeneity are proving to be powerful prognostic biomarkers in lymphoma. Texture analysis, the most advanced area of radiomics, offers highly complex features that require further standardization and validation before being adopted as reliable biomarkers. Combining radiomic features with clinical risk factors and genomic data holds promising potential for improving clinical risk prediction. This review explores the current state of radiomic analysis, progress towards its standardization and its incorporation into clinical practice and trial designs. The integration of radiomic markers with circulating tumour DNA may provide a comprehensive approach to developing baseline and dynamic risk scores, facilitating the testing of novel treatments and advancing personalized treatment of aggressive lymphomas.
Collapse
Affiliation(s)
- Luca Ceriani
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Lisa Milan
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Stephane Chauvie
- Medical Physics Division, Santa Croce e Carlo Hospital, Cuneo, Italy
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Haematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Medical Oncology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Ende TVD, Kuijper SC, Widaatalla Y, Noortman WA, van Velden FHP, Woodruff HC, van der Pol Y, Moldovan N, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, de Geus-Oei LF, Lambin P, van Laarhoven HWM. Integrating Clinical Variables, Radiomics, and Tumor-derived Cell-Free DNA for Enhanced Prediction of Resectable Esophageal Adenocarcinoma Outcomes. Int J Radiat Oncol Biol Phys 2025; 121:963-974. [PMID: 39424077 DOI: 10.1016/j.ijrobp.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The value of integrating clinical variables, radiomics, and tumor-derived cell-free DNA (cfDNA) for the prediction of survival and response to chemoradiation of patients with resectable esophageal adenocarcinoma is not yet known. Our aim was to investigate if radiomics and cfDNA metrics combined with clinical variables can improve personalized predictions. METHODS AND MATERIALS A cohort of 111 patients with resectable esophageal adenocarcinoma from 2 centers treated with neoadjuvant chemoradiation therapy was used for exploratory retrospective analyses. Models combining the clinical variables of the SOURCE survival model with radiomic features and cfDNA were built using elastic net regression and internally validated using 5-fold cross-validation. Model performance for overall survival (OS) and time to progression (TTP) were evaluated with the C-index and the area under the curve for pathologic complete response. RESULTS The best-performing baseline models for OS and TTP were based on the combination of SOURCE-cfDNA that reached a C-index of 0.55 and 0.59 compared with 0.44 to 0.45 with SOURCE alone. The addition of restaging positron emission tomography radiomics to SOURCE was the most promising addition for predicting OS (C-index: 0.65) and TTP (C-index: 0.60). Baseline risk stratification was achieved for OS and TTP by combining SOURCE with radiomics or cfDNA, log-rank P < .01. The best-performing combination model for the prediction of pathologic complete response reached an area under the curve of 0.61 compared with 0.47 with SOURCE variables alone. CONCLUSIONS The addition of radiomics and cfDNA can improve the performance of an established survival model. External validity needs to be further assessed in future studies together with the optimization of radiomic pipelines.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Steven C Kuijper
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands; Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft., The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Jing F, Zhang X, Liu Y, Chen X, Zhao X, Chen X, Yuan H, Dai M, Wang N, Han J, Zhang J. Baseline 18F-FDG PET Radiomics Predicting Therapeutic Efficacy of Diffuse Large B-Cell Lymphoma after R-CHOP (-Like) Therapy. Cancer Biother Radiopharm 2025; 40:114-121. [PMID: 39230437 DOI: 10.1089/cbr.2024.0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
Objective: This study aimed to predict therapeutic efficacy among diffuse large B-cell lymphoma (DLBCL) after R-CHOP (-like) therapy using baseline 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) radiomics. Methods: A total of 239 patients with DLBCL were enrolled in this study, with 82 patients having refractory/relapsed disease. The radiomics signatures were developed using a stacking ensemble approach. The efficacy of the radiomics signatures, the National Comprehensive Cancer Network-International Prognostic Index (NCCN-IPI), conventional PET parameters model, and their combinations in assessing refractory/relapse risk were evaluated using receiver operating characteristic (ROC) curves, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), accuracy and decision curve analysis. Results: The stacking model, along with the integrated model that combines stacking with the NCCN-IPI and SDmax (the distance between the two lesions farthest apart, normalized to the patient's body surface area), showed remarkable predictive capabilities with a high area under the curve (AUC), sensitivity, specificity, PPV, NPV, accuracy, and significant net benefit of the AUC (NB-AUC). Although no significant differences were observed between the combined and stacking models in terms of the AUC in either the training cohort (AUC: 0.992 vs. 0.985, p = 0.139) or the testing cohort (AUC: 0.768 vs. 0.781, p = 0.668), the integrated model exhibited higher values for sensitivity, PPV, NPV, accuracy, and NB-AUC than the stacking model. Conclusion: Baseline PET radiomics could predict therapeutic efficacy in DLBCL after R-CHOP (-like) therapy, with improved predictive performance when incorporating clinical features and SDmax.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/diagnostic imaging
- Lymphoma, Large B-Cell, Diffuse/pathology
- Fluorodeoxyglucose F18/administration & dosage
- Male
- Female
- Vincristine/therapeutic use
- Rituximab/therapeutic use
- Doxorubicin/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Middle Aged
- Prednisone/therapeutic use
- Cyclophosphamide/therapeutic use
- Positron-Emission Tomography/methods
- Aged
- Adult
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Prognosis
- Treatment Outcome
- Radiopharmaceuticals
- Aged, 80 and over
- Young Adult
- Radiomics
Collapse
Affiliation(s)
- Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, People's Republic of China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, People's Republic of China
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
4
|
Jing F, Zhang X, Liu Y, Chen X, Zhao J, Zhao X, Chen X, Yuan H, Dai M, Wang N, Zhang Z, Zhang J. Baseline 18F-FDG PET/CT radiomics for prognosis prediction in diffuse large B cell lymphoma with extranodal involvement. Clin Transl Oncol 2025; 27:727-735. [PMID: 39083140 DOI: 10.1007/s12094-024-03633-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/19/2024] [Indexed: 02/01/2025]
Abstract
PURPOSE The objective of this investigation is to explore the capability of baseline 18F-FDG PET/CT radiomics to predict the prognosis of diffuse large B-cell lymphoma (DLBCL) with extranodal involvement (ENI). METHODS 126 patients diagnosed with DLBCL with ENI were included in the cohort. The least absolute shrinkage and selection operator (LASSO) Cox regression was utilized to refine the optimum subset from the 1328 features. Cox regression analyses were employed to discern significant clinical variables and conventional PET parameters, which were then employed with radiomics score to develop combined model for predicting both progression-free survival (PFS) and overall survival (OS). The fitness and the predictive capability of the models were assessed via the Akaike information criterion (AIC) and concordance index (C-index). RESULTS 62 patients experienced disease recurrence or progression and 28 patients ultimately died. The combined model exhibited a lower AIC value compared to the radiomics model and SDmax/clinical variables for both PFS (507.101 vs. 510.658 vs. 525.506) and OS (215.667 vs. 230.556 vs. 219.313), respectively. The C-indices of the combined model, radiomics model, and SDmax/clinical variables were 0.724, 0.704, and 0.615 for PFS, and 0.842, 0.744, and 0.792 for OS, respectively. Kaplan--Meier curves showed significantly higher rates of relapse and mortality among patients classified as high-risk compared to those classified as low-risk (all P < 0.05). CONCLUSIONS The combined model of clinical variables, conventional PET parameters, and baseline PET/CT radiomics features demonstrates a higher accuracy in predicting the prognosis of DLBCL with ENI.
Collapse
Affiliation(s)
- Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, 050000, Hebei, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jianqiang Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 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.
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
5
|
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
|
6
|
Liu C, Zhao J, Zhang H, Ni X. Diagnostic Value of 18F-FDG PET/CT Radiomics in Lymphoma: A Systematic Review and Meta-Analysis. Technol Cancer Res Treat 2025; 24:15330338251342860. [PMID: 40397102 DOI: 10.1177/15330338251342860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
IntroductionVarious machine learning models and features have been proposed for lymphoma diagnosis using 18F-fluorodeoxyglucose (18F-FDG) PET/CT radiomics. This research aimed to systematically evaluate the diagnostic value of 18F-FDG PET/CT radiomics in lymphoma by conducting a meta-analysis.MethodsData from published studies regarding the diagnosis of lymphoma using 18F-FDG PET/CT radiomics, from January 2010 to July 2024, were gathered from PubMed, Web of Science, and the Cochrane Library. Following their separate searches and screenings of the literature, two researchers extracted data and assessed the caliber of all the included studies. The quality assessment involved the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2), the Radiomics Quality Score (RQS), and the METhodological RadiomICs Score (METRICS). The meta-analysis was conducted by using RevMan 5.4.1, R 4.4.0, and Stata 17.0 software. Six meta-regressions were conducted on study performance, considering sample size, image modality, region of interest (ROI) selection, ROI segmentation, radiomics mode, and algorithms.ResultsIn total, 20 studies classified as type 2a or above according to the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD) statement were included for this systematic review and meta-analysis. The studies achieved an average RQS of 13 (ranging from 10 to 17), accounting for 36.1% of the total points. The average METRICS score was 69.3% (ranging from 54.8% to 80.9%). The quality category of the studies is mainly "good". The results of our meta-analysis showed that the pooled sensitivity (SEN), specificity (SPE), positive likelihood ratio, negative likelihood ratio and diagnostic odds ratio with 95% confidence interval (CI) were 0.82 (0.78, 0.88), 0.83 (0.76, 0.87), 4.7 (3.4, 6.6), 0.20 (0.15, 0.28) and 23 (13, 42), respectively. The area under the curve of the summary receiver operating characteristic curve was 0.90 (0.87, 0.92). The results of Spearman correlation analysis revealed no threshold effect among the studies (P = .423). Significant heterogeneity was observed among the studies (overall I2 = 83.7%; 95% CI: 76.0, 88.9; P < .01). Meta-regressions indicated that sample size and ROI selection contributed to the heterogeneity in SEN, while algorithms affected the heterogeneity in SPE (P < .05). Deeks' test confirmed there was no significant publication bias in all the included studies. The Fagan nomogram showed an absolute increase of 34% in the post-test probability following a positive test result.ConclusionThe results supported that 18F-FDG PET/CT radiomics has high diagnostic value for lymphoma. However, there is high heterogeneity among different studies. In the future, clinical practicality needs to be substantiated by more prospective studies with rigorous adherence to existing guidelines and multicentric validation.
Collapse
Affiliation(s)
- Chaoying Liu
- Department of Medical Equipment, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
| | - Jun Zhao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Heng Zhang
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
- Department of Radiotherapy, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Xinye Ni
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
- Department of Radiotherapy, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| |
Collapse
|
7
|
Seban RD, Champion L, De Moura A, Lerebours F, Loirat D, Pierga JY, Djerroudi L, Genevee T, Huchet V, Jehanno N, Bidard FC, Buvat I. Pre-treatment [18F]FDG PET/CT biomarkers for the prediction of antibody-drug conjugates efficacy in metastatic breast cancer. Eur J Nucl Med Mol Imaging 2025; 52:708-718. [PMID: 39373900 DOI: 10.1007/s00259-024-06929-x] [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: 07/19/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE This study aimed to evaluate the association between pretreatment [18F]FDG PET/CT-derived biomarkers and outcomes in metastatic breast cancer (mBC) patients treated with antibody-drug conjugates (ADCs) Sacituzumab Govitecan (SG) and Trastuzumab Deruxtecan (T-DXd). METHODS A retrospective bicentric analysis was conducted on triple-negative mBC (mTNBC) patients treated with SG and HER2-low mBC patients treated with T-DXd, who underwent [18F]FDG PET/CT scans before therapy. Key biomarkers, including maximum standardized uptake value (SUVmax), total metabolic tumor volume (TMTV) and maximum tumor dissemination (Dmax), were measured. Their prognostic value for progression-free survival (PFS) and overall survival (OS) was assessed using Cox models and Kaplan-Meier curves. RESULTS 128 patients were included: 71 mTNBC treated with SG and 57 HR-positive and -negative HER2-low mBC treated with T-DXd. Median follow-up was 12.9 months. In the SG cohort, median PFS and OS were 4.8 and 8.9 months, respectively. High Dmax (HR 2.1, 95% CI 1.1-4.3) and high TMTV (HR 2.9, 95% CI 1.2-6.6) were independently associated with shorter OS. In the T-DXd cohort, median PFS and OS were 5.8 and 9.0 months, respectively. High Dmax (HR 2.1, 95% CI 1.2-3.9) and high TMTV (HR 2.4, 95% CI 1.0-6.5) independently correlated with shorter PFS and shorter OS, respectively. CONCLUSION Pretreatment [18F]FDG PET/CT-derived biomarkers, namely TMTV and Dmax, have significant prognostic value in patients with mTNBC and HER2-low mBC treated with SG and T-DXd. These biomarkers improve prognostic prediction and may optimize treatment strategies, warranting their clinical use, but larger studies are needed to validate these findings.
Collapse
Affiliation(s)
- Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France.
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Institut Curie, Paris Saclay University, 91400, Orsay, France.
| | - Laurence Champion
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Institut Curie, Paris Saclay University, 91400, Orsay, France
| | - Alexandre De Moura
- Department of Medical Oncology, Institut Curie, 92210, Saint-Cloud, France
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie, 92210, Saint-Cloud, France
| | - Delphine Loirat
- Department of Medical Oncology, Institut Curie, 75005, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, Université Paris Cité, 75005, Paris, France
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | | | - Thomas Genevee
- Department of Pharmacy, Institut Curie, 92210, Saint-Cloud, France
| | - Virginie Huchet
- Department of Nuclear Medicine, Institut Curie, 75005, Paris, France
| | - Nina Jehanno
- Department of Nuclear Medicine, Institut Curie, 75005, Paris, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Institut Curie, UVSQ/Paris-Saclay University, 92210, Saint-Cloud, France
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Irene Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Institut Curie, Paris Saclay University, 91400, Orsay, France
| |
Collapse
|
8
|
Triumbari EKA, Morland D, Gatta R, Boldrini L, De Summa M, Chiesa S, Cuccaro A, Maiolo E, Hohaus S, Annunziata S. The predictive power of 18F-FDG PET/CT two-lesions radiomics and conventional models in classical Hodgkin's Lymphoma: a comparative retrospectively-validated study. Ann Hematol 2025; 104:641-651. [PMID: 39808225 PMCID: PMC11868178 DOI: 10.1007/s00277-025-06190-8] [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: 08/28/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
In a previous preliminary study, radiomic features from the largest and the hottest lesion in baseline 18F-FDG PET/CT (bPET/CT) of classical Hodgkin's Lymphoma (cHL) predicted early response-to-treatment and prognosis. Aim of this large retrospectively-validated study is to evaluate the predictive role of two-lesions radiomics in comparison with other clinical and conventional PET/CT models. cHL patients with bPET/CT between 2010 and 2020 were retrospectively included and randomized into training-validation sets. Target lesions were: Lesion_A, with largest axial diameter (Dmax); Lesion_B, with highest SUVmax. Total-metabolic-tumor-volume (TMTV) was calculated and 212 radiomic features were extracted. PET/CT features were harmonized using ComBat across two scanners. Outcomes were progression-free-survival (PFS) and Deauville Score at interim PET/CT (DS). For each outcome, three predictive models and their combinations were trained and validated: - radiomic model "R"; - conventional PET/CT model "P"; - clinical model "C". 197 patients were included (training = 118; validation = 79): 38/197 (19%) patients had adverse events and 42/193 (22%) had DS ≥ 4. In the training phase, only one radiomic feature was selected for PFS prediction in model "R" (Lesion_B F_cm.corr, C-index 66.9%). Best "C" model combined stage and IPS (C-index 74.8%), while optimal "P" model combined TMTV and Dmax (C-index 63.3%). After internal validation, "C", "C + R", "R + P" and "C + R + P" significantly predicted PFS. The best validated model was "C + R" (C-index 66.3%). No model was validated for DS prediction. In this large retrospectively-validated study, a combination of baseline 18F-FDG PET/CT two-lesions radiomics and other conventional models showed an added prognostic power in patients with cHL. As single models, conventional clinical parameters maintain their prognostic power, while radiomics or conventional PET/CT alone seem to be sub-optimal to predict survival.
Collapse
Affiliation(s)
- Elizabeth Katherine Anna Triumbari
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - David Morland
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institut Godinot and CReSTIC EA 3804, Université de Reims Champagne-Ardenne, Reims, France
| | - Roberto Gatta
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Luca Boldrini
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco De Summa
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Medipass S.p.a. Integrative Service, Rome, Italy
| | - Silvia Chiesa
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Annarosa Cuccaro
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Maiolo
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefan Hohaus
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Annunziata
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Department of Radiology, Radiotherapy and Hematology, Unità di Medicina Nucleare, GSTeP Radiopharmacy, Fondazione Policlinico Universitario A.Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
9
|
Dabrowska-Iwanicka A, Nowakowski GS. DLBCL: who is high risk and how should treatment be optimized? Blood 2024; 144:2573-2582. [PMID: 37922443 DOI: 10.1182/blood.2023020779] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/05/2023] Open
Abstract
ABSTRACT Diffuse large B-cell lymphoma (DLBCL), not otherwise specified, is the most common subtype of large B-cell lymphoma, with differences in prognosis reflecting heterogeneity in the pathological, molecular, and clinical features. Current treatment standard is based on multiagent chemotherapy, including anthracycline and monoclonal anti-CD20 antibody, which leads to cure in 60% of patients. Recent years have brought new insights into lymphoma biology and have helped refine the risk groups. The results of these studies inspired the design of new clinical trials with targeted therapies and response-adapted strategies and allowed to identify groups of patients potentially benefiting from new agents. This review summarizes recent progress in identifying high-risk patients with DLBCL using clinical and biological prognostic factors assessed at diagnosis and during treatment in the front-line setting, as well as new treatment strategies with the application of targeted agents and immunotherapy, including response-adapted strategies.
Collapse
Affiliation(s)
- Anna Dabrowska-Iwanicka
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | |
Collapse
|
10
|
Sun Z, Yang T, Ding C, Shi Y, Cheng L, Jia Q, Tao W. Clinical scoring systems, molecular subtypes and baseline [ 18F]FDG PET/CT image analysis for prognosis of diffuse large B-cell lymphoma. Cancer Imaging 2024; 24:168. [PMID: 39696503 DOI: 10.1186/s40644-024-00810-8] [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: 02/26/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous hematological malignancy resulting in a range of outcomes, and the early prediction of these outcomes has important implications for patient management. Clinical scoring systems provide the most commonly used prognostic evaluation criteria, and the value of genetic testing has also been confirmed by in-depth research on molecular typing. [18F]-fluorodeoxyglucose positron emission tomography / computed tomography ([18F]FDG PET/CT) is an invaluable tool for predicting DLBCL progression. Conventional baseline image-based parameters and machine learning models have been used in prognostic FDG PET/CT studies of DLBCL; however, numerous studies have shown that combinations of baseline clinical scoring systems, molecular subtypes, and parameters and models based on baseline FDG PET/CT image may provide better predictions of patient outcomes and aid clinical decision-making in patients with DLBCL.
Collapse
Affiliation(s)
- Zhuxu Sun
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Tianshuo Yang
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Chongyang Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Luyi Cheng
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Qingshen Jia
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University, Tianjin, China
| | - Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China.
| |
Collapse
|
11
|
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
|
12
|
Czibor S, Csatlós Z, Fábián K, Piroska M, Györke T. Volumetric and textural analysis of PET/CT in patients with diffuse large B-cell lymphoma highlights the importance of novel MTVrate feature. Nucl Med Commun 2024; 45:931-937. [PMID: 39102514 PMCID: PMC11460743 DOI: 10.1097/mnm.0000000000001884] [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: 01/10/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVES To investigate the prognostic value of clinical, volumetric, and radiomics-based textural parameters in baseline [ 18 F]FDG-PET/CT scans of diffuse large B-cell lymphoma (DLBCL) patients. METHODS We retrospectively investigated baseline PET/CT scans and collected clinical data of fifty DLBCL patients. PET images were segmented semiautomatically to determine metabolic tumor volume (MTV), then the largest segmented lymphoma volume of interest (VOI) was used to extract first-, second-, and high-order textural features. A novel value, MTVrate was introduced as the quotient of the largest lesion's volume and total body MTV. Receiver operating characteristics (ROC) analyses were performed and 24-months progression-free survival (PFS) of low- and high-risk cohorts were compared by log-rank analyses. A machine learning algorithm was used to build a prognostic model from the available clinical, volumetric, and textural data based on logistic regression. RESULTS The area-under-the-curve (AUC) on ROC analysis was the highest of MTVrate at 0.74, followed by lactate-dehydrogenase, MTV, and skewness, with AUCs of 0.68, 0.63, and 0.55, respectively which parameters were also able to differentiate the PFS. A combined survival analysis including MTV and MTVrate identified a subgroup with particularly low PFS at 38%. In the machine learning-based model had an AUC of 0.83 and the highest relative importance was attributed to three textural features and both MTV and MTVrate as important predictors of PFS. CONCLUSION Individual evaluation of different biomarkers yielded only limited prognostic data, whereas a machine learning-based combined analysis had higher effectivity. MTVrate had the highest prognostic ability on individual analysis and, combined with MTV, it identified a patient group with particularly poor prognosis.
Collapse
Affiliation(s)
- Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| | | | - Krisztián Fábián
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
- Mediso Medical Imaging Systems, Budapest, Hungary
| | - Márton Piroska
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| |
Collapse
|
13
|
Ferrández MC, Golla SSV, Eertink JJ, Wiegers SE, Zwezerijnen GJC, Heymans MW, Lugtenburg PJ, Kurch L, Hüttmann A, Hanoun C, Dührsen U, Barrington SF, Mikhaeel NG, Ceriani L, Zucca E, Czibor S, Györke T, Chamuleau MED, Zijlstra JM, Boellaard R. Validation of an Artificial Intelligence-Based Prediction Model Using 5 External PET/CT Datasets of Diffuse Large B-Cell Lymphoma. J Nucl Med 2024; 65:1802-1807. [PMID: 39362767 DOI: 10.2967/jnumed.124.268191] [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: 06/06/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
The aim of this study was to validate a previously developed deep learning model in 5 independent clinical trials. The predictive performance of this model was compared with the international prognostic index (IPI) and 2 models incorporating radiomic PET/CT features (clinical PET and PET models). Methods: In total, 1,132 diffuse large B-cell lymphoma patients were included: 296 for training and 836 for external validation. The primary outcome was 2-y time to progression. The deep learning model was trained on maximum-intensity projections from PET/CT scans. The clinical PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, SUVpeak, age, and performance status. The PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, and SUVpeak Model performance was assessed using the area under the curve (AUC) and Kaplan-Meier curves. Results: The IPI yielded an AUC of 0.60 on all external data. The deep learning model yielded a significantly higher AUC of 0.66 (P < 0.01). For each individual clinical trial, the model was consistently better than IPI. Radiomic model AUCs remained higher for all clinical trials. The deep learning and clinical PET models showed equivalent performance (AUC, 0.69; P > 0.05). The PET model yielded the highest AUC of all models (AUC, 0.71; P < 0.05). Conclusion: The deep learning model predicted outcome in all trials with a higher performance than IPI and better survival curve separation. This model can predict treatment outcome in diffuse large B-cell lymphoma without tumor delineation but at the cost of a lower prognostic performance than with radiomics.
Collapse
Affiliation(s)
- Maria C Ferrández
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sandeep S V Golla
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn W Heymans
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lars Kurch
- Clinic and Polyclinic for Nuclear Medicine, Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sally F Barrington
- School of Biomedical Engineering and Imaging Sciences, King's College London and Guy's and St Thomas' PET Centre, King's Health Partners, King's College London, London, United Kingdom
| | - N George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Luca Ceriani
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Emanuele Zucca
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, Oncology Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; and
| | - Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Martine E D Chamuleau
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
14
|
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
|
15
|
Chen J, Lin F, Dai Z, Chen Y, Fan Y, Li A, Zhao C. Survival prediction in diffuse large B-cell lymphoma patients: multimodal PET/CT deep features radiomic model utilizing automated machine learning. J Cancer Res Clin Oncol 2024; 150:452. [PMID: 39382750 PMCID: PMC11464575 DOI: 10.1007/s00432-024-05905-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/21/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE We sought to develop an effective combined model for predicting the survival of patients with diffuse large B-cell lymphoma (DLBCL) based on the multimodal PET-CT deep features radiomics signature (DFR-signature). METHODS 369 DLBCL patients from two medical centers were included in this study. Their PET and CT images were fused to construct the multimodal PET-CT images using a deep learning fusion network. Then the deep features were extracted from those fused PET-CT images, and the DFR-signature was constructed through an Automated machine learning (AutoML) model. Combined with clinical indexes from the Cox regression analysis, we constructed a combined model to predict the progression-free survival (PFS) and the overall survival (OS) of patients. In addition, the combined model was evaluated in the concordance index (C-index) and the time-dependent area under the ROC curve (tdAUC). RESULTS A total of 1000 deep features were extracted to build a DFR-signature. Besides the DFR-signature, the combined model integrating metabolic and clinical factors performed best in terms of PFS and OS. For PFS, the C-indices are 0.784 and 0.739 in the training cohort and internal validation cohort, respectively. For OS, the C-indices are 0.831 and 0.782 in the training cohort and internal validation cohort. CONCLUSIONS DFR-signature constructed from multimodal images improved the classification accuracy of prognosis for DLBCL patients. Moreover, the constructed DFR-signature combined with NCCN-IPI exhibited excellent potential for risk stratification of DLBCL patients.
Collapse
Affiliation(s)
- Jianxin Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Fengyi Lin
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Zhaoyan Dai
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yu Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yawen Fan
- 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
| | - Chenyu Zhao
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| |
Collapse
|
16
|
Li M, Fang B, Gu H, Jiang Y. EQ-5D-5L and SF-6Dv2 health utilities scores of diffuse large B-cell lymphoma patients in China. Health Qual Life Outcomes 2024; 22:80. [PMID: 39300432 DOI: 10.1186/s12955-024-02297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND This study evaluates the health-related quality of life (HRQoL) of persons with diffuse large B-cell lymphoma (DLBCL) by using EQ-5D-5L and SF-6Dv2 and compares the measurement properties of the two instruments. METHOD DLBCL patients were identified via a patient group and were surveyed using web-based questionnaires. Demographic information, socioeconomic status (SES), clinical characteristics, and EQ-5D-5L and SF-6Dv2 responses were collected and statistically described. The association between the EQ-5D-5L and SF-6Dv2 dimensions were analyzed using the Spearman's correlation coefficient, whereas the correlation of the utility scores was evaluated using Pearson's correlation coefficient. The agreement between the responses of the two instruments were examined using a Bland-Altman (B-A) plot. A one-way analysis of variance (ANOVA) was performed to compare the utility scores across subgroups in different clinical states (a t-test was used if there were two subgroups). In addition, the graded response model (GRM) was used to describe the discrimination ability and difficulty characteristics of the dimensions in the two instruments. RESULTS In total, 582 valid responses were collected, among which 477 respondents were associated with initial-treatment and 105 respondents were relapsed/refractory (RR) patients. The mean (standard deviation [SD]) EQ-5D-5L and SF-6Dv2 utility scores of the DLBCL patients were 0.828 (0.222) and 0.641 (0.220), respectively. The correlation between the EQ-5D-5L and SF-6Dv2 dimensions ranged from 0.299 to 0.680, and the correlation between their utility scores was 0.787. The B-A plot demonstrated an acceptable but not strong agreement between EQ-5D-5L and SF-6Dv2 utility scores. The GRM model results indicated that all dimensions of each instrument were highly discriminating overall, but EQ-5D-5L had suboptimal discriminative power among patients with good health. CONCLUSION Both the EQ-5D-5L and SF-6Dv2 showed valid properties to assess the HRQoL of DLBCL patients. However, utility scores derived from the two instruments had substantial difference, thereby prohibiting the interchangeable use of utilities from the two instruments.
Collapse
Affiliation(s)
- Mincai Li
- School of Public Health (Shenzhen), Sun Yat-Sen University, Room 533, West Wing of Medical Complex #1, Shenzhen, China
| | - Bingxue Fang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Room 533, West Wing of Medical Complex #1, Shenzhen, China
| | - Hongfei Gu
- Hongmian Cancers and Rare Disorders Charity Foundation of Guangzhou, Guangzhou, China
| | - Yawen Jiang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Room 533, West Wing of Medical Complex #1, Shenzhen, China.
| |
Collapse
|
17
|
Cui S, Xin W, Wang F, Shao X, Shao X, Niu R, Zhang F, Shi Y, Liu B, Gu W, Wang Y. Metabolic tumour area: a novel prognostic indicator based on 18F-FDG PET/CT in patients with diffuse large B-cell lymphoma in the R-CHOP era. BMC Cancer 2024; 24:895. [PMID: 39054508 PMCID: PMC11270790 DOI: 10.1186/s12885-024-12668-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND The metabolic tumour area (MTA) was found to be a promising predictor of prostate cancer. However, the role of MTA based on 18F-FDG PET/CT in diffuse large B-cell lymphoma (DLBCL) prognosis remains unclear. This study aimed to elucidate the prognostic significance of MTA and evaluate its incremental value to the National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI) for DLBCL patients treated with first-line R-CHOP regimens. METHODS A total of 280 consecutive patients with newly diagnosed DLBCL and baseline 18F-FDG PET/CT data were retrospectively evaluated. Lesions were delineated via a semiautomated segmentation method based on a 41% SUVmax threshold to estimate semiquantitative metabolic parameters such as total metabolic tumour volume (TMTV) and MTA. Receiver operating characteristic (ROC) curve analysis was used to determine the optimal cut-off values. Progression-free survival (PFS) and overall survival (OS) were the endpoints that were used to evaluate the prognosis. PFS and OS were estimated via Kaplan‒Meier curves and compared via the log-rank test. RESULTS Univariate analysis revealed that patients with high MTA, high TMTV and NCCN-IPI ≥ 4 were associated with inferior PFS and OS (P < 0.0001 for all). Multivariate analysis indicated that MTA remained an independent predictor of PFS and OS [hazard ratio (HR), 2.506; 95% confidence interval (CI), 1.337-4.696; P = 0.004; and HR, 1.823; 95% CI, 1.005-3.310; P = 0.048], whereas TMTV was not. Further analysis using the NCCN-IPI model as a covariate revealed that MTA and NCCN-IPI were still independent predictors of PFS (HR, 2.617; 95% CI, 1.494-4.586; P = 0.001; and HR, 2.633; 95% CI, 1.650-4.203; P < 0.0001) and OS (HR, 2.021; 95% CI, 1.201-3.401; P = 0.008; and HR, 3.869; 95% CI, 1.959-7.640; P < 0.0001; respectively). Furthermore, MTA was used to separate patients with high NCCN-IPI risk scores into two groups with significantly different outcomes. CONCLUSIONS Pre-treatment MTA based on 18F-FDG PET/CT and NCCN-IPI were independent predictor of PFS and OS in DLBCL patients treated with R-CHOP. MTA has additional predictive value for the prognosis of patients with DLBCL, especially in high-risk patients with NCCN-IPI ≥ 4. In addition, the combination of MTA and NCCN-IPI may be helpful in further improving risk stratification and guiding individualised treatment options. TRIAL REGISTRATION This research was retrospectively registered with the Ethics Committee of the Third Affiliated Hospital of Soochow University, and the registration number was approval No. 155 (approved date: 31 May 2022).
Collapse
Affiliation(s)
- Silu Cui
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
- Yangzhou University, Yangzhou, Jiangsu, China
| | - Wenchong Xin
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
- Department of Nuclear Medicine, Linyi People's Hospital, Linyi, Shandong, China
| | - Fei Wang
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China.
| | - Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Rong Niu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Feifei Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Yunmei Shi
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Bao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
18
|
Knaup H, Weindler J, van Heek L, Voltin CA, Fuchs M, Borchmann P, Dietlein M, Kobe C, Roth K. PET/CT Reconstruction and Its Impact on [Measures of] Metabolic Tumor Volume. Acad Radiol 2024; 31:3020-3025. [PMID: 38155023 DOI: 10.1016/j.acra.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
RATIONALE AND OBJECTIVES In oncological imaging, the use of metabolic tumor volume (MTV) for further prognostic differentiation and the development of risk adapted strategies appears promising. The aim of this analysis was to evaluate ultra-high definition (UHD) and ordered subset expectation maximization (OSEM) PET/CT reconstructions for their potential impact on different methods of MTV measurement. MATERIALS AND METHODS We analyzed positron emission tomography combined with computed tomography (PET/CT) scans of 40 Hodgkin lymphoma patients before first-line treatment who had undergone fluorodeoxyglucose (FDG) PET/CT. The MTVs were determined taking an SUV of 4.0 (MTV4.0) as a fixed threshold or 41% of the single hottest voxel (MTV41%) as an adaptive threshold for automated lymphoma delineation in both UHD and OSEM reconstructions. We then compared the absolute and relative differences between MTV4.0 and MTV41% in UHD and OSEM reconstructions. The relative distribution of MTV4.0 and MTV41% in relation to the reconstruction method applied was recorded and respective differences were tested for statistical significance using the paired sample t-test. RESULTS A comparison of MTV4.0 and MTV41% showed smaller relative and absolute differences in MTV between different reconstruction settings for the MTV4.0 method. Conversely, the absolute as well as the relative differences between MTVs obtained from different reconstructions settings were significantly greater when the MTV41% method was applied (p < 0001). CONCLUSION MTV4.0 brings higher robustness between different reconstruction settings, while with MTV41% the deviation between volumes obtained with different reconstruction settings is greater. For clinical routine and for multicenter settings, the MTV4.0 therefore appears most promising.
Collapse
Affiliation(s)
- Henry Knaup
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Jasmin Weindler
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Lutz van Heek
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Conrad-Amadeus Voltin
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Michael Fuchs
- German Hodgkin Study Group, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany (M.F., P.B.)
| | - Peter Borchmann
- German Hodgkin Study Group, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany (M.F., P.B.)
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Carsten Kobe
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.).
| | - Katrin Roth
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| |
Collapse
|
19
|
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
|
20
|
Horvat N, Papanikolaou N, Koh DM. Radiomics Beyond the Hype: A Critical Evaluation Toward Oncologic Clinical Use. Radiol Artif Intell 2024; 6:e230437. [PMID: 38717290 PMCID: PMC11294952 DOI: 10.1148/ryai.230437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
Radiomics is a promising and fast-developing field within oncology that involves the mining of quantitative high-dimensional data from medical images. Radiomics has the potential to transform cancer management, whereby radiomics data can be used to aid early tumor characterization, prognosis, risk stratification, treatment planning, treatment response assessment, and surveillance. Nevertheless, certain challenges have delayed the clinical adoption and acceptability of radiomics in routine clinical practice. The objectives of this report are to (a) provide a perspective on the translational potential and potential impact of radiomics in oncology; (b) explore frequent challenges and mistakes in its derivation, encompassing study design, technical requirements, standardization, model reproducibility, transparency, data sharing, privacy concerns, quality control, as well as the complexity of multistep processes resulting in less radiologist-friendly interfaces; (c) discuss strategies to overcome these challenges and mistakes; and (d) propose measures to increase the clinical use and acceptability of radiomics, taking into account the different perspectives of patients, health care workers, and health care systems. Keywords: Radiomics, Oncology, Cancer Management, Artificial Intelligence © RSNA, 2024.
Collapse
Affiliation(s)
- Natally Horvat
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY (N.H.); Department of Radiology, University of São Paulo, São Paulo, Brazil (N.H.); Computational Clinical Imaging Group, Champalimaud Foundation, Portugal (N.P.); and Department of Radiology, Royal Marsden Hospital, Downs Rd, Sutton SM2 5PT, United Kingdom (N.P., D.M.K.)
| | - Nikolaos Papanikolaou
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY (N.H.); Department of Radiology, University of São Paulo, São Paulo, Brazil (N.H.); Computational Clinical Imaging Group, Champalimaud Foundation, Portugal (N.P.); and Department of Radiology, Royal Marsden Hospital, Downs Rd, Sutton SM2 5PT, United Kingdom (N.P., D.M.K.)
| | - Dow-Mu Koh
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY (N.H.); Department of Radiology, University of São Paulo, São Paulo, Brazil (N.H.); Computational Clinical Imaging Group, Champalimaud Foundation, Portugal (N.P.); and Department of Radiology, Royal Marsden Hospital, Downs Rd, Sutton SM2 5PT, United Kingdom (N.P., D.M.K.)
| |
Collapse
|
21
|
Haider SP, Zeevi T, Sharaf K, Gross M, Mahajan A, Kann BH, Judson BL, Prasad ML, Burtness B, Aboian M, Canis M, Reichel CA, Baumeister P, Payabvash S. Impact of 18F-FDG PET Intensity Normalization on Radiomic Features of Oropharyngeal Squamous Cell Carcinomas and Machine Learning-Generated Biomarkers. J Nucl Med 2024; 65:803-809. [PMID: 38514087 PMCID: PMC11927063 DOI: 10.2967/jnumed.123.266637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
We aimed to investigate the effects of 18F-FDG PET voxel intensity normalization on radiomic features of oropharyngeal squamous cell carcinoma (OPSCC) and machine learning-generated radiomic biomarkers. Methods: We extracted 1,037 18F-FDG PET radiomic features quantifying the shape, intensity, and texture of 430 OPSCC primary tumors. The reproducibility of individual features across 3 intensity-normalized images (body-weight SUV, reference tissue activity ratio to lentiform nucleus of brain and cerebellum) and the raw PET data was assessed using an intraclass correlation coefficient (ICC). We investigated the effects of intensity normalization on the features' utility in predicting the human papillomavirus (HPV) status of OPSCCs in univariate logistic regression, receiver-operating-characteristic analysis, and extreme-gradient-boosting (XGBoost) machine-learning classifiers. Results: Of 1,037 features, a high (ICC ≥ 0.90), medium (0.90 > ICC ≥ 0.75), and low (ICC < 0.75) degree of reproducibility across normalization methods was attained in 356 (34.3%), 608 (58.6%), and 73 (7%) features, respectively. In univariate analysis, features from the PET normalized to the lentiform nucleus had the strongest association with HPV status, with 865 of 1,037 (83.4%) significant features after multiple testing corrections and a median area under the receiver-operating-characteristic curve (AUC) of 0.65 (interquartile range, 0.62-0.68). Similar tendencies were observed in XGBoost models, with the lentiform nucleus-normalized model achieving the numerically highest average AUC of 0.72 (SD, 0.07) in the cross validation within the training cohort. The model generalized well to the validation cohorts, attaining an AUC of 0.73 (95% CI, 0.60-0.85) in independent validation and 0.76 (95% CI, 0.58-0.95) in external validation. The AUCs of the XGBoost models were not significantly different. Conclusion: Only one third of the features demonstrated a high degree of reproducibility across intensity-normalization techniques, making uniform normalization a prerequisite for interindividual comparability of radiomic markers. The choice of normalization technique may affect the radiomic features' predictive value with respect to HPV. Our results show trends that normalization to the lentiform nucleus may improve model performance, although more evidence is needed to draw a firm conclusion.
Collapse
Affiliation(s)
- Stefan P Haider
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany;
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Tal Zeevi
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Kariem Sharaf
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Moritz Gross
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
- Charité Center for Diagnostic and Interventional Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Amit Mahajan
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Benjamin H Kann
- Department of Radiation Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Benjamin L Judson
- Division of Otolaryngology, Yale School of Medicine, New Haven, Connecticut
| | - Manju L Prasad
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut; and
| | - Barbara Burtness
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut
| | - Mariam Aboian
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Martin Canis
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | | |
Collapse
|
22
|
Philip MM, Watts J, Moeini SNM, Musheb M, McKiddie F, Welch A, Nath M. Comparison of semi-automatic and manual segmentation methods for tumor delineation on head and neck squamous cell carcinoma (HNSCC) positron emission tomography (PET) images. Phys Med Biol 2024; 69:095005. [PMID: 38530298 DOI: 10.1088/1361-6560/ad37ea] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/26/2024] [Indexed: 03/27/2024]
Abstract
Objective. Accurate and reproducible tumor delineation on positron emission tomography (PET) images is required to validate predictive and prognostic models based on PET radiomic features. Manual segmentation of tumors is time-consuming whereas semi-automatic methods are easily implementable and inexpensive. This study assessed the reliability of semi-automatic segmentation methods over manual segmentation for tumor delineation in head and neck squamous cell carcinoma (HNSCC) PET images.Approach. We employed manual and six semi-automatic segmentation methods (just enough interaction (JEI), watershed, grow from seeds (GfS), flood filling (FF), 30% SUVmax and 40%SUVmax threshold) using 3D slicer software to extract 128 radiomic features from FDG-PET images of 100 HNSCC patients independently by three operators. We assessed the distributional properties of all features and considered 92 log-transformed features for subsequent analysis. For each paired comparison of a feature, we fitted a separate linear mixed effect model using the method (two levels; manual versus one semi-automatic method) as a fixed effect and the subject and the operator as the random effects. We estimated different statistics-the intraclass correlation coefficient agreement (aICC), limits of agreement (LoA), total deviation index (TDI), coverage probability (CP) and coefficient of individual agreement (CIA)-to evaluate the agreement between the manual and semi-automatic methods.Main results. Accounting for all statistics across 92 features, the JEI method consistently demonstrated acceptable agreement with the manual method, with median values of aICC = 0.86, TDI = 0.94, CP = 0.66, and CIA = 0.91.Significance. This study demonstrated that JEI method is a reliable semi-automatic method for tumor delineation on HNSCC PET images.
Collapse
Affiliation(s)
- Mahima Merin Philip
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Jessica Watts
- National Health Service Grampian, Aberdeen AB15 6RE, United Kingdom
| | | | - Mohammed Musheb
- National Health Service Highland, Inverness IV2 3BW, United Kingdom
| | - Fergus McKiddie
- National Health Service Grampian, Aberdeen AB15 6RE, United Kingdom
| | - Andy Welch
- Institute of Education in Healthcare and Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Mintu Nath
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| |
Collapse
|
23
|
Draye-Carbonnier S, Camus V, Becker S, Tonnelet D, Lévêque E, Zduniak A, Jardin F, Tilly H, Vera P, Decazes P. Prognostic value of the combination of volume, massiveness and fragmentation parameters measured on baseline FDG pet in high-burden follicular lymphoma. Sci Rep 2024; 14:8033. [PMID: 38580734 PMCID: PMC10997640 DOI: 10.1038/s41598-024-58412-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
The prognostic value of radiomic quantitative features measured on pre-treatment 18F-FDG PET/CT was investigated in patients with follicular lymphoma (FL). We conducted a retrospective study of 126 FL patients (grade 1-3a) diagnosed between 2006 and 2020. A dozen of PET/CT-derived features were extracted via a software (Oncometer3D) from baseline 18F-FDG PET/CT images. The receiver operating characteristic (ROC) curve, Kaplan-Meier method and Cox analysis were used to assess the prognostic factors for progression of disease within 24 months (POD24) and progression-free survival at 24 months. Four different clusters were identified among the twelve PET parameters analyzed: activity, tumor burden, fragmentation-massiveness and dispersion. On ROC analyses, TMTV, the total metabolic tumor volume, had the highest AUC (0.734) followed by medPCD, the median distance between the centroid of the tumors and their periphery (AUC: 0.733). Patients with high TMTV (HR = 4.341; p < 0.001), high Tumor Volume Surface Ratio (TVSR) (HR = 3.204; p < 0.003) and high medPCD (HR = 4.507; p < 0.001) had significantly worse prognosis in both Kaplan-Meier and Cox univariate analyses. Furthermore, a synergistic effect was observed in Kaplan-Meier and Cox analyses combining these three PET/CT-derived parameters (HR = 12.562; p < 0.001). Having two or three high parameters among TMTV, TVSR and medPCD was able to predict POD24 status with a specificity of 68% and a sensitivity of 75%. TMTV, TVSR and baseline medPCD are strong prognostic factors in FL and their combination better predicts disease prognosis.
Collapse
Affiliation(s)
| | - V Camus
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Université de Rouen, IRIB, Rouen, France
| | - S Becker
- Department of Nuclear Medicine, Centre Henri Becquerel, Rouen, France
- QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - D Tonnelet
- Department of Nuclear Medicine, Centre Henri Becquerel, Rouen, France
| | - E Lévêque
- Department of Statistics and Clinical Research Unit, Centre Henri Becquerel, Rouen, France
| | - A Zduniak
- Department of Hematology, Centre Henri Becquerel, Rouen, France
| | - F Jardin
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Université de Rouen, IRIB, Rouen, France
| | - H Tilly
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Université de Rouen, IRIB, Rouen, France
| | - P Vera
- Department of Nuclear Medicine, Centre Henri Becquerel, Rouen, France
- QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - P Decazes
- Department of Nuclear Medicine, Centre Henri Becquerel, Rouen, France.
- QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France.
| |
Collapse
|
24
|
Aksu A, Küçüker KA, Solmaz Ş, Turgut B. A different perspective on PET/CT before treatment in patients with Hodgkin lymphoma: importance of volumetric and dissemination parameters. Ann Hematol 2024; 103:813-822. [PMID: 37964021 DOI: 10.1007/s00277-023-05547-1] [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: 10/02/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023]
Abstract
The aim of this study is to investigate the role of the combination of volumetric and dissemination parameters obtained from pretreatment 18-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) in predicting the interim response and progression status in patients with Hodgkin lymphoma (HL). Pretreatment PET/CT images of HL patients were analyzed with LIFEx software, and volumes of interest (VOIs) were drawn with a fixed SUV 4.0 threshold. MTV, SUVmax, and TLG values were obtained from each VOI. Total MTV (tMTV) was calculated by summing the MTV values in all VOIs, and similarly, total TLG (tTLG) was obtained by summing the TLG values. The distance between the centers of the lesions was noted as Dmax, and the distance between the outermost voxels of the lesions as DmaxVox. tMTV/DmaxVox was calculated by dividing the tMTV value by the DmaxVox value, and tTLG/DmaxVox was calculated by dividing the tTLG value by the DmaxVox value. The correlation of pretreatment PET parameters with response groups (complete/poor) and relapse/progression status (stable/progressive) was statistically evaluated. A total of 52 patients were included in the study. Bulky disease, tMTV, tTLG, and tMTV/DmaxVox values were found to be significantly higher in the poor response group. tMTV > 190.60 ml was found to be the only prognostic factor predicting interim PET response. The tMTV/DmaxVox and tTLG/DmaxVox showed statistically significant differences between the groups with and without progression. tMTV/DmaxVox > 7.70 was found to be the only prognostic factor in predicting relapse/progression. The evaluation of tumor burden and dissemination together in 18F-FDG PET/CT before treatment in patients with HL can help us to predict the results of the patients.
Collapse
Affiliation(s)
- Ayşegül Aksu
- Department of Nuclear Medicine, Atatürk Training and Research Hospital, İzmir Kâtip Çelebi University, İzmir, Turkey.
| | - Kadir Alper Küçüker
- Department of Nuclear Medicine, Atatürk Training and Research Hospital, İzmir Kâtip Çelebi University, İzmir, Turkey
| | - Şerife Solmaz
- Department of Hematology, Atatürk Training and Research Hospital, İzmir Kâtip Çelebi University, İzmir, Turkey
| | - Bülent Turgut
- Department of Nuclear Medicine, Atatürk Training and Research Hospital, İzmir Kâtip Çelebi University, İzmir, Turkey
| |
Collapse
|
25
|
Girum KB, Cottereau AS, Vercellino L, Rebaud L, Clerc J, Casasnovas O, Morschhauser F, Thieblemont C, Buvat I. Tumor Location Relative to the Spleen Is a Prognostic Factor in Lymphoma Patients: A Demonstration from the REMARC Trial. J Nucl Med 2024; 65:313-319. [PMID: 38071535 PMCID: PMC10858380 DOI: 10.2967/jnumed.123.266322] [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: 07/12/2023] [Revised: 10/23/2023] [Indexed: 02/03/2024] Open
Abstract
Baseline [18F]FDG PET/CT radiomic features can improve the survival prediction in patients with diffuse large B-cell lymphoma (DLBCL). The purpose of this study was to investigate whether characterizing tumor locations relative to the spleen location in baseline [18F]FDG PET/CT images predicts survival in patients with DLBCL and improves the predictive value of total metabolic tumor volume (TMTV) and age-adjusted international prognostic index (IPI). Methods: This retrospective study included 301 DLBCL patients from the REMARC (NCT01122472) cohort. Physicians delineated the tumor regions, whereas the spleen was automatically segmented using an open-access artificial intelligence algorithm. We systematically measured the distance between the centroid of the spleen and all other lesions, defining the SD of these distances as the lesion spread (SpreadSpleen). We calculated the maximum distance between the spleen and another lesion (Dspleen) for each patient and normalized it with the body surface area, resulting in standardized Dspleen (sDspleen). The predictive value of each PET/CT feature for progression-free survival (PFS) and overall survival (OS) was evaluated through univariate and multivariate time-dependent Cox models and Kaplan-Meier analysis. Results: In total, 282 patients (mean age, 68.33 ± 5.41 y; 164 men) were evaluated. The artificial intelligence algorithm successfully segmented the spleen in 96% of the patients. SpreadSpleen, Dspleen, and sDspleen were correlated neither with TMTV (Pearson ρ < 0.23) nor with IPI (Pearson ρ < 0.15). When median values were used as the cutoff, SpreadSpleen, Dspleen, and sDspleen all significantly classified patients into 2 risk groups for PFS and OS (P < 0.001). They complemented TMTV and IPI to classify the patients into 3 risk groups for PFS and OS (P < 0.001). Integrating SpreadSpleen, Dspleen, or sDspleen into a Cox model on the basis of TMTV, IPI, and TMTV combined with IPI significantly improved the concordance index for PFS and OS (P < 0.05). Conclusion: Baseline PET/CT features that characterize tumor spread and dissemination relative to the spleen strongly predicted survival in patients with DLBCL. Integrating these features with TMTV and IPI further improved survival prediction.
Collapse
Affiliation(s)
- Kibrom B Girum
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France
| | - Anne-Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, Paris Descartes University, Paris, France
| | | | - Louis Rebaud
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France
- Research and Clinical Collaborations, Siemens Medical Solutions USA, Knoxville, Tennessee
| | - Jérôme Clerc
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, Paris Descartes University, Paris, France
| | | | - Franck Morschhauser
- Research Group on Injectable Forms and Associated Technologies, Department of Hematology, Claude Huriez Hospital, University Lille, Lille, France; and
| | | | - Irène Buvat
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France;
| |
Collapse
|
26
|
Alderuccio JP, Reis IM, Hamadani M, Nachiappan M, Leslom S, Kahl BS, Ai WZ, Radford J, Solh M, Ardeshna KM, Hess BT, Lunning MA, Zinzani PL, Stathis A, Carlo-Stella C, Lossos IS, Caimi PF, Han S, Yang F, Kuker RA, Moskowitz CH. PET/CT Biomarkers Enable Risk Stratification of Patients with Relapsed/Refractory Diffuse Large B-cell Lymphoma Enrolled in the LOTIS-2 Clinical Trial. Clin Cancer Res 2024; 30:139-149. [PMID: 37855688 PMCID: PMC10872617 DOI: 10.1158/1078-0432.ccr-23-1561] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/11/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE Significant progress has occurred in developing quantitative PET/CT biomarkers in diffuse large B-cell lymphoma (DLBCL). Total metabolic tumor volume (MTV) is the most extensively studied, enabling assessment of FDG-avid tumor burden associated with outcomes. However, prior studies evaluated the outcome of cytotoxic chemotherapy or chimeric antigen receptor T-cell therapy without data on recently approved FDA agents. Therefore, we aimed to assess the prognosis of PET/CT biomarkers in patients treated with loncastuximab tesirine. EXPERIMENTAL DESIGN We centrally reviewed screening PET/CT scans of patients with relapsed/refractory DLBCL enrolled in the LOTIS-2 (NCT03589469) study. MTV was obtained by computing individual volumes using the SUV ≥4.0 threshold. Other PET/CT metrics, clinical factors, and the International Metabolic Prognostic Index (IMPI) were evaluated. Logistic regression was used to assess the association between biomarkers and treatment response. Cox regression was used to determine the effect of biomarkers on time-to-event outcomes. We estimated biomarker prediction as continuous and binary variables defined by cutoff points. RESULTS Across 138 patients included in this study, MTV with a cutoff point of 96 mL was the biomarker associated with the highest predictive performance in univariable and multivariable models to predict failure to achieve complete metabolic response (OR, 5.42; P = 0.002), progression-free survival (HR, 2.68; P = 0.002), and overall survival (HR, 3.09; P < 0.0001). IMPI demonstrated an appropriate performance, however, not better than MTV alone. CONCLUSIONS Pretreatment MTV demonstrated robust risk stratification, with those patients demonstrating high MTV achieving lower responses and survival to loncastuximab tesirine in relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Isildinha M. Reis
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mehdi Hamadani
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Muthiah Nachiappan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Salman Leslom
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Brad S. Kahl
- Washington University, St. Louis, MO, United States
| | - Weiyun Z. Ai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - John Radford
- NIHR Clinical Research Facility, University of Manchester and the Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Melhem Solh
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, United States
| | - Kirit M. Ardeshna
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Brian T. Hess
- Medical University of South Carolina, Charleston, SC, United States
| | - Matthew A. Lunning
- University of Nebraska Medical Center- Fred and Pamela Buffett Cancer Center, Omaha, NE, United States
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and Department of Oncology and Hematology, Humanitas Research Hospital–IRCCS, Milano, Italy
| | - Izidore S. Lossos
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Paolo F. Caimi
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States
| | - Sunwoo Han
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Fei Yang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Russ A. Kuker
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Craig H. Moskowitz
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
27
|
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
|
28
|
Marchal E, Palard-Novello X, Lhomme F, Meyer ME, Manson G, Devillers A, Marolleau JP, Houot R, Girard A. Baseline [ 18F]FDG PET features are associated with survival and toxicity in patients treated with CAR T cells for large B cell lymphoma. Eur J Nucl Med Mol Imaging 2024; 51:481-489. [PMID: 37721580 DOI: 10.1007/s00259-023-06427-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/04/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T cells have established themselves as an effective treatment for refractory or relapsed large B cell lymphoma (LBCL). Recently, the sDmax, which corresponds to the distance separating the two farthest lesions standardized by the patient's body surface area, has appeared as a prognostic factor in LBCL. This study aimed to identify [18F]FDG-PET biomarkers associated with prognosis and predictive of adverse events in patients treated with CAR T cells. METHODS Patients were retrospectively included from two different university hospitals. They were being treated with CAR T cells for LBCL and underwent [18F]FDG-PET just before CAR T cell infusion. Lesions were segmented semi-automatically with a threshold of 41% of the maximal uptake. In addition to clinico-biological features, sDmax, total metabolic tumor volume (TMTV), SUVmax, and uptake intensity of healthy lymphoid organs and liver were collected. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. The occurrence of adverse events, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), was reported. RESULTS Fifty-six patients were included. The median follow-up was 9.7 months. Multivariate analysis showed that TMTV (cut-off of 36 mL) was an independent prognostic factor for PFS (p < 0.001) and that sDmax (cut-off of 0.15 m-1) was an independent prognostic factor for OS (p = 0.008). Concerning the occurrence of adverse events, a C-reactive protein level > 35 mg/L (p = 0.006) and a liver SUVmean > 2.5 (p = 0.027) before CAR T cells were associated with grade 2 to 4 CRS and a spleen SUVmean > 1.9 with grade 2 to 4 ICANS. CONCLUSION TMTV and sDmax had independent prognostic values, respectively, on PFS and OS. Regarding adverse events, the mean liver and spleen uptakes were associated with the occurrence of grade 2 to 4 CRS and ICANS, respectively. Integrating these biomarkers into the clinical workflow could be useful for early adaptation of patients management.
Collapse
Affiliation(s)
- E Marchal
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France.
| | - X Palard-Novello
- Department of Nuclear Medicine, University Rennes, CLCC Eugène Marquis, INSERM, LTSI-UMR 1099, Rennes, France
| | - F Lhomme
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - M E Meyer
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France
| | - G Manson
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - A Devillers
- Department of Nuclear Medicine, CLCC Eugène Marquis, Rennes, France
| | - J P Marolleau
- Department of Hematology, Amiens-Picardie University Hospital, Amiens, France
| | - R Houot
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - A Girard
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France
| |
Collapse
|
29
|
Rogasch JMM, Shi K, Kersting D, Seifert R. Methodological evaluation of original articles on radiomics and machine learning for outcome prediction based on positron emission tomography (PET). Nuklearmedizin 2023; 62:361-369. [PMID: 37995708 PMCID: PMC10667066 DOI: 10.1055/a-2198-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023]
Abstract
AIM Despite a vast number of articles on radiomics and machine learning in positron emission tomography (PET) imaging, clinical applicability remains limited, partly owing to poor methodological quality. We therefore systematically investigated the methodology described in publications on radiomics and machine learning for PET-based outcome prediction. METHODS A systematic search for original articles was run on PubMed. All articles were rated according to 17 criteria proposed by the authors. Criteria with >2 rating categories were binarized into "adequate" or "inadequate". The association between the number of "adequate" criteria per article and the date of publication was examined. RESULTS One hundred articles were identified (published between 07/2017 and 09/2023). The median proportion of articles per criterion that were rated "adequate" was 65% (range: 23-98%). Nineteen articles (19%) mentioned neither a test cohort nor cross-validation to separate training from testing. The median number of criteria with an "adequate" rating per article was 12.5 out of 17 (range, 4-17), and this did not increase with later dates of publication (Spearman's rho, 0.094; p = 0.35). In 22 articles (22%), less than half of the items were rated "adequate". Only 8% of articles published the source code, and 10% made the dataset openly available. CONCLUSION Among the articles investigated, methodological weaknesses have been identified, and the degree of compliance with recommendations on methodological quality and reporting shows potential for improvement. Better adherence to established guidelines could increase the clinical significance of radiomics and machine learning for PET-based outcome prediction and finally lead to the widespread use in routine clinical practice.
Collapse
Affiliation(s)
- Julian Manuel Michael Rogasch
- Department of Nuclear Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital University Hospital Bern, Bern, Switzerland
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
30
|
Lai Q, Zhao Y, Yan H, Peng H. Advances in diagnosis, treatment and prognostic factors of gastrointestinal DLBCL. Leuk Res 2023; 135:107406. [PMID: 37944240 DOI: 10.1016/j.leukres.2023.107406] [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: 08/12/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Gastrointestinal diffuse large B-cell lymphoma (GI-DLBCL) is an extremely aggressive form of B-cell non-Hodgkin lymphoma (BNHL) which has complex histological characteristics and manifests a high degree of heterogeneity in terms of clinical, morphological, immunological, and genetic features. GI-DLBCL mainly spreads by infiltrating neighboring lymph nodes, and common gastrointestinal complications (GICS) such as obstruction, perforation, or bleeding, frequently arise during the progression of the disease, posing significant challenges in both diagnosing and treating the condition. Meanwhile, the incidence of GI-DLBCL has been gradually increasing in recent years, and its strong invasiveness makes it prone to being misdiagnosed or completely missed. In clinical practice, over half of the patients diagnosed with the disease are in stage III or stage IV. What makes it worse is that certain patients may not exhibit a favorable response to chemotherapy. All these lead to intricacies in management of this disease. Unfortunately, there is currently no large prospective study or evidence-based medical evidence to provide clear guidance on treatment decisions for this specific type of lymphoma. Neither do physicians have a consensus regarding the optimal approach to address this condition. Recent studies have identified the presence of various prognostic factors that significantly impact survival in GI-DLBCL, which demonstrates the unique particularity of GI-DLBCL, and could help optimize the clinical decision.
Collapse
Affiliation(s)
- Qinqiao Lai
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiqing Yan
- Department of gastric and abdominal cancer ward, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China.
| |
Collapse
|
31
|
Huo Z, Chen F, Zhao J, Liu P, Chao Z, Liu K, Zhou J, Zhou D, Zhang L, Zhen H, Yang W, Tan Z, Zhu K, Luo Z. Prognostic impact of absolute peripheral blood NK cell count after four cycles of R-CHOP-like regimen treatment in patients with diffuse large B cell lymphoma. Clin Exp Med 2023; 23:4665-4672. [PMID: 37938466 PMCID: PMC10725372 DOI: 10.1007/s10238-023-01249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
As a subtype of lymphocyte, natural killer (NK) cell is the first line of defense that shows a strong function in tumor immunotherapy response and clinical outcomes. The current study aims to investigate the prognostic influence of peripheral blood absolute NK cell count after four cycles of rituximab combined with cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) treatment (NKCC4) in diffuse large B cell lymphoma (DLBCL) patients. A total of 261 DLBCL patients treated with R-CHOP from January 2018 to September 2022 were enrolled. The low NKCC4 was observed in patients who died during the study period compared with survival individuals. A NKCC4 < 135 cells/μl had a remarkable negative influence in overall survival and progression-free survival (PFS) compared to a NKCC4 ≥ 135 cells/μl (p < 0.0001 and p < 0.0004, respectively). In addition, the OS and PFS were synergistically lower in a NKCC4 < 135 cells/μl group among DLBCL patients with GCB type or high IPI. In conclusion, this study indicates NCKK4 as a valuable marker in clinical practice and provides an insight for combination treatment of R-CHOP to improve outcomes of DLBCL patients.
Collapse
Affiliation(s)
- Zhongjun Huo
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Fang Chen
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Jiajia Zhao
- Department of Reproductive and Genetic Center, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Ping Liu
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Zhi Chao
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Kang Liu
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Ji Zhou
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Dan Zhou
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Lu Zhang
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Haifeng Zhen
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Wenqun Yang
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Zhenqing Tan
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Kaibo Zhu
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Zimian Luo
- Department of Hematology, Central Hospital of Xiangtan, Xiangtan, 411100, China.
| |
Collapse
|
32
|
Wang F, Cui S, Lu L, Shao X, Yan F, Liu Y, He B, Wang J, Cao Y, Yue Y, Wang Y, Gu W. Dissemination feature based on PET/CT is a risk factor for diffuse large B cell lymphoma patients outcome. BMC Cancer 2023; 23:1165. [PMID: 38030989 PMCID: PMC10687880 DOI: 10.1186/s12885-023-11333-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/24/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND 18F-FDG PET/CT provides precise information about dissemination of lymphoma lesions. Dmax, defined as distance between the two lesions that were farthest apart by PET/CT, was found to be a promising predictor of Diffuse large B-cell lymphoma (DLBCL) outcome in a small size of clinical trial data. We analyzed the impact of Dmax on the outcome of a large real-world DLBCL cohort. METHODS Data of newly diagnosed DLBCL at the Third Affiliated Hospital of Soochow University were retrospectively collected. Baseline Dmax, clinical data and survival information were recorded. A metabolic parameter, metabolic bulk volume (MBV), was also measured to verify the independent impact of Dmax. RESULTS Optimal cut-off values for Dmax and MBV were 45.34 cm and 21.65 cm3. With a median follow-up of 32 months, Dmax significantly impacted progression-free survival (PFS) and overall survival (OS) in 253 DLBCL patients. For Dmaxlow and Dmaxhigh groups, estimated 3-year OS were 87.0% and 53.8% (p < 0.001), while 3-year PFS were 77.3% and 37.3% (p < 0.001). And for MBVlow and MBVhighgroups, 3-year OS were 84.5% and 58.8% (p < 0.001), and 3-year PFS were 68.7% and 50.4% (p = 0.003). Multivariate analysis identified Dmax and Eastern Cooperative Oncology Group performance status (ECOG PS) independently associated with PFS and OS, while MBV only independently associated with OS. A Dmax revised prognostic index (DRPI) combining Dmax and ECOG PS identified an ultra-risk DLBCL population with 3-year PFS of 31.7% and 3-year OS of 38.5%. The area under the curve (AUC) showed that this model performed better than International prognostic Index (IPI). CONCLUSION Dmax is a new and promising indicator to investigate dissemination of lymphoma lesions associated with the outcome of DLBCL. It significantly contributes to stratification of patients with disparate outcomes. TRIAL REGISTRATION This research has been retrospectively registered in the Ethics Committee institutional of the Third Affiliated Hospital of Soochow University, and the registration number was approval No. 155 (approved date: 31 May 2022).
Collapse
Affiliation(s)
- Fei Wang
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Silu Cui
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Luo Lu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Feng Yan
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yaqi Liu
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Bai He
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jianfeng Wang
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yanhua Yue
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
33
|
Lewis KL, Trotman J. Integration of PET in DLBCL. Semin Hematol 2023; 60:291-304. [PMID: 38326144 DOI: 10.1053/j.seminhematol.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 02/09/2024]
Abstract
F-fluorodeoxyglucose positron emission tomography-computerized tomography (18FDG-PET/CT) is the gold-standard imaging modality for staging and response assessment for most lymphomas. This review focuses on the utility of 18FDG-PET/CT, and its role in staging, prognostication and response assessment in diffuse large B-cell lymphoma (DLBCL), including emerging possibilities for future use.
Collapse
Affiliation(s)
| | - Judith Trotman
- Concord Repatriation General Hospital, Concord, NSW, Australia
| |
Collapse
|
34
|
Ligero M, Simó M, Carpio C, Iacoboni G, Balaguer‐Montero M, Navarro V, Sánchez‐Salinas MA, Bobillo S, Marín‐Niebla A, Iraola‐Truchuelo J, Abrisqueta P, Sala‐Llonch R, Bosch F, Perez‐Lopez R, Barba P. PET-based radiomics signature can predict durable responses to CAR T-cell therapy in patients with large B-cell lymphoma. EJHAEM 2023; 4:1081-1088. [PMID: 38024636 PMCID: PMC10660117 DOI: 10.1002/jha2.757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 12/01/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment option for relapsed or refractory (R/R) large B-cell lymphoma (LBCL). However, only a subset of patients will present long-term benefit. In this study, we explored the potential of PET-based radiomics to predict treatment outcomes with the aim of improving patient selection for CAR T-cell therapy. We conducted a single-center study including 93 consecutive R/R LBCL patients who received a CAR T-cell infusion from 2018 to 2021, split in training set (73 patients) and test set (20 patients). Radiomics features were extracted from baseline PET scans and clinical benefit was defined based on median progression-free survival (PFS). Cox regression models including the radiomics signature, conventional PET biomarkers and clinical variables were performed for most relevant outcomes. A radiomics signature including 4 PET-based parameters achieved an AUC = 0.73 for predicting clinical benefit in the test set, outperforming the predictive value of conventional PET biomarkers (total metabolic tumor volume [TMTV]: AUC = 0.66 and maximum standardized uptake value [SUVmax]: AUC = 0.59). A high radiomics score was also associated with longer PFS and OS in the multivariable analysis. In conclusion, the PET-based radiomics signature predicted efficacy of CAR T-cell therapy and outperformed conventional PET biomarkers in our cohort of LBCL patients.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics GroupVall d'Hebron Institute of Oncology (VHIO)Vall d'Hebron Barcelona Hospital Campus (VHUH)BarcelonaSpain
| | - Marc Simó
- Nuclear Medicine DepartmentVall d'Hebron University Hospital, Autonomous University of BarcelonaBarcelonaSpain
| | - Cecilia Carpio
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Gloria Iacoboni
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Maria Balaguer‐Montero
- Radiomics GroupVall d'Hebron Institute of Oncology (VHIO)Vall d'Hebron Barcelona Hospital Campus (VHUH)BarcelonaSpain
| | - Victor Navarro
- Oncology Data Science (ODysSey) GroupVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | - Mario Andres Sánchez‐Salinas
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Sabela Bobillo
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Ana Marín‐Niebla
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Josu Iraola‐Truchuelo
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Pau Abrisqueta
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Roser Sala‐Llonch
- Faculty of MedicineDepartment of BiomedicineInstitute of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)University of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)BarcelonaSpain
| | - Francesc Bosch
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| | - Raquel Perez‐Lopez
- Radiomics GroupVall d'Hebron Institute of Oncology (VHIO)Vall d'Hebron Barcelona Hospital Campus (VHUH)BarcelonaSpain
| | - Pere Barba
- Department of HematologyExperimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University HospitalBarcelonaBarcelonaSpain
| |
Collapse
|
35
|
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
|
36
|
Zhao S, Wang J, Jin C, Zhang X, Xue C, Zhou R, Zhong Y, Liu Y, He X, Zhou Y, Xu C, Zhang L, Qian W, Zhang H, Zhang X, Tian M. Stacking Ensemble Learning-Based [ 18F]FDG PET Radiomics for Outcome Prediction in Diffuse Large B-Cell Lymphoma. J Nucl Med 2023; 64:1603-1609. [PMID: 37500261 DOI: 10.2967/jnumed.122.265244] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/31/2023] [Indexed: 07/29/2023] Open
Abstract
This study aimed to develop an analytic approach based on [18F]FDG PET radiomics using stacking ensemble learning to improve the outcome prediction in diffuse large B-cell lymphoma (DLBCL). Methods: In total, 240 DLBCL patients from 2 medical centers were divided into the training set (n = 141), internal testing set (n = 61), and external testing set (n = 38). Radiomics features were extracted from pretreatment [18F]FDG PET scans at the patient level using 4 semiautomatic segmentation methods (SUV threshold of 2.5, SUV threshold of 4.0 [SUV4.0], 41% of SUVmax, and SUV threshold of mean liver uptake [PERCIST]). All extracted features were harmonized with the ComBat method. The intraclass correlation coefficient was used to evaluate the reliability of radiomics features extracted by different segmentation methods. Features from the most reliable segmentation method were selected by Pearson correlation coefficient analysis and the LASSO (least absolute shrinkage and selection operator) algorithm. A stacking ensemble learning approach was applied to build radiomics-only and combined clinical-radiomics models for prediction of 2-y progression-free survival and overall survival based on 4 machine learning classifiers (support vector machine, random forests, gradient boosting decision tree, and adaptive boosting). Confusion matrix, receiver-operating-characteristic curve analysis, and survival analysis were used to evaluate the model performance. Results: Among 4 semiautomatic segmentation methods, SUV4.0 segmentation yielded the highest interobserver reliability, with 830 (66.7%) selected radiomics features. The combined model constructed by the stacking method achieved the best discrimination performance. For progression-free survival prediction in the external testing set, the areas under the receiver-operating-characteristic curve and accuracy of the stacking-based combined model were 0.771 and 0.789, respectively. For overall survival prediction, the stacking-based combined model achieved an area under the curve of 0.725 and an accuracy of 0.763 in the external testing set. The combined model also demonstrated a more distinct risk stratification than the International Prognostic Index in all sets (log-rank test, all P < 0.05). Conclusion: The combined model that incorporates [18F]FDG PET radiomics and clinical characteristics based on stacking ensemble learning could enable improved risk stratification in DLBCL.
Collapse
Affiliation(s)
- Shuilin Zhao
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xiang Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chenxi Xue
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yuwei Liu
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xuexin He
- Department of Medical Oncology, Huashan Hospital of Fudan University, Shanghai, China
| | - Youyou Zhou
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Caiyun Xu
- Department of Nuclear Medicine, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Lixia Zhang
- Department of Nuclear Medicine, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Wenbin Qian
- Department of Hematology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China;
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China; and
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China;
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Huang R, Geng H, Zhu L, Yan J, Li C, Li Y. CT radiomics can predict disease progression within 6 months after chimeric antigen receptor-modified T-cell therapy in relapsed/refractory B-cell non-Hodgkin's lymphoma patients. Clin Radiol 2023; 78:e707-e717. [PMID: 37407367 DOI: 10.1016/j.crad.2023.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/05/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023]
Abstract
AIM To predict progression within 6 months after chimeric antigen receptor-modified (CAR) T-cell therapy for relapsed/refractory (R/R) B-cell non-Hodgkin's lymphoma (B-NHL) patients by radiomic indexes derived from contrast-enhanced computed tomography (CECT) examinations. MATERIALS AND METHODS Seventy R/R B-NHL patients who underwent CECT before treatment with CAR T-cells were examined retrospectively. In total, 297 volumes of interest for lesions were segmented from CECT images. Patients without and with disease progression were assigned to groups 1 and 2, respectively. Radiomic and combined predictive models were constructed by three machine-learning algorithms using features from the training set, respectively. Furthermore, predictive models were constructed based on multi-lesion-based and largest-lesion-based radiomic features, respectively. RESULTS In the test set, no marked differences were observed between the areas under the curves (AUCs) of the combined and radiomic models for all three machine-learning algorithms (all p>0.05). Differences in machine-learning algorithms did not significantly affect the predictive performances of the models. Radiomics and combined models constructed with multi-lesion-based radiomic features showed better predictive performances than those applying largest-lesion-based radiomic features (all p<0.05 for comparisons between combined models). CONCLUSION CECT-based radiomic features may be applied to predict disease progression in R/R B-NHL patients within 6 months after CAR T-cell treatment, and radiomic features from multiple lesions may have better predictive efficacy. Different machine-learning algorithms may not show significant differences in prediction performance.
Collapse
Affiliation(s)
- R Huang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China
| | - H Geng
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China
| | - L Zhu
- Department of Ultrasound, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province, 215000, PR China
| | - J Yan
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China
| | - C Li
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China
| | - Y Li
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu province 215000, PR China; Institute of Medical Imaging, Soochow University, Suzhou City, Jiangsu province 215000, PR China.
| |
Collapse
|
38
|
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
|
39
|
Ferrández MC, Golla SSV, Eertink JJ, de Vries BM, Wiegers SE, Zwezerijnen GJC, Pieplenbosch S, Schilder L, Heymans MW, Zijlstra JM, Boellaard R. Sensitivity of an AI method for [ 18F]FDG PET/CT outcome prediction of diffuse large B-cell lymphoma patients to image reconstruction protocols. EJNMMI Res 2023; 13:88. [PMID: 37758869 PMCID: PMC10533444 DOI: 10.1186/s13550-023-01036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Convolutional neural networks (CNNs), applied to baseline [18F]-FDG PET/CT maximum intensity projections (MIPs), show potential for treatment outcome prediction in diffuse large B-cell lymphoma (DLBCL). The aim of this study is to investigate the robustness of CNN predictions to different image reconstruction protocols. Baseline [18F]FDG PET/CT scans were collected from 20 DLBCL patients. EARL1, EARL2 and high-resolution (HR) protocols were applied per scan, generating three images with different image qualities. Image-based transformation was applied by blurring EARL2 and HR images to generate EARL1 compliant images using a Gaussian filter of 5 and 7 mm, respectively. MIPs were generated for each of the reconstructions, before and after image transformation. An in-house developed CNN predicted the probability of tumor progression within 2 years for each MIP. The difference in probabilities per patient was then calculated between both EARL2 and HR with respect to EARL1 (delta probabilities or ΔP). We compared these to the probabilities obtained after aligning the data with ComBat using the difference in median and interquartile range (IQR). RESULTS CNN probabilities were found to be sensitive to different reconstruction protocols (EARL2 ΔP: median = 0.09, interquartile range (IQR) = [0.06, 0.10] and HR ΔP: median = 0.1, IQR = [0.08, 0.16]). Moreover, higher resolution images (EARL2 and HR) led to higher probability values. After image-based and ComBat transformation, an improved agreement of CNN probabilities among reconstructions was found for all patients. This agreement was slightly better after image-based transformation (transformed EARL2 ΔP: median = 0.022, IQR = [0.01, 0.02] and transformed HR ΔP: median = 0.029, IQR = [0.01, 0.03]). CONCLUSION Our CNN-based outcome predictions are affected by the applied reconstruction protocols, yet in a predictable manner. Image-based harmonization is a suitable approach to harmonize CNN predictions across image reconstruction protocols.
Collapse
Affiliation(s)
- Maria C Ferrández
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Sandeep S V Golla
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bart M de Vries
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Louise Schilder
- Department of Internal Medicine, Amstelland Hospital, Amstelveen, The Netherlands
| | - Martijn W Heymans
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Yin J, Wang H, Zhu G, Chen N, Khan MI, Zhao Y. Prognostic value of whole-body dynamic 18F-FDG PET/CT Patlak in diffuse large B-cell lymphoma. Heliyon 2023; 9:e19749. [PMID: 37809527 PMCID: PMC10559051 DOI: 10.1016/j.heliyon.2023.e19749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/23/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Objective This study aims to investigate the significance of interim whole-body dynamic 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) Patlak parameters for predicting the prognosis of patients with diffuse large B-cell lymphoma. To estimate the predictive value of the whole-body dynamic 18F-FDG PET/CT Patlak parameter for 2-year progression-free survival (PFS) and 2-year overall survival (OS). Methods This study reports the findings of 67 patients with diffuse large B-cell lymphoma (DLBCL). These patients underwent interim whole-body dynamic 18F-FDG PET/CT scans from June 2021 to January 2023 at the Department of Nuclear Medicine, First Affiliated Hospital of Anhui Medical University. The predictive values of maximum standard uptake value (SUVmax), maximum of net glucose uptake rate (Kimax) and the predictive model combining Kimax and interim treatment response on the prognosis of patients was analyzed using receiver operating characteristic (ROC) curves. Kaplan-Meier survival curves and log-rank tests were used for survival analysis. Univariate and multivariate analyses were performed to screen for independent prognostic risk factors. Results After a median follow-up of 18 months, 21 patients (31.3%) experienced disease recurrence or death. The cut-off values for the SUVmax and the Kimax were 6.1 and 0.13 μmol min-1·ml-1, respectively. Ann Arbor stage, IPI, SUVmax, Kimax and interim treatment response were associated with PFS and OS in the univariate analysis. However, only Kimax and interim treatment response were independent influences on PFS and OS in multivariate analysis. Conclusion Interim whole-body dynamic 18F-FDG PET/CT Patlak imaging has significant prognostic value in patients with DLBCL. Among them, the interim dynamic parameter Kimax showed the best predictive value for prognosis compared with the interim SUVmax and interim treatment response. The predictive model established by Kimax and the interim treatment response allowed for the accurate stratification of the prognostic risk of DLBCL.
Collapse
Affiliation(s)
- Jiankang Yin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China
| | - Hui Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Gan Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Ni Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China
| | - Muhammad Imran Khan
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, Anhui, PR China
- Department of Pathology, District Headquarters Hospital, Jhang, 35200, Punjab Province, Pakistan
- Hefei National Lab for Physical Sciences at Microscale and the Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, PR China
| | - Ye Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China
| |
Collapse
|
41
|
Alderuccio JP, Kuker RA, Yang F, Moskowitz CH. Quantitative PET-based biomarkers in lymphoma: getting ready for primetime. Nat Rev Clin Oncol 2023; 20:640-657. [PMID: 37460635 DOI: 10.1038/s41571-023-00799-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 08/20/2023]
Abstract
The use of functional quantitative biomarkers extracted from routine PET-CT scans to characterize clinical responses in patients with lymphoma is gaining increased attention, and these biomarkers can outperform established clinical risk factors. Total metabolic tumour volume enables individualized estimation of survival outcomes in patients with lymphoma and has shown the potential to predict response to therapy suitable for risk-adapted treatment approaches in clinical trials. The deployment of machine learning tools in molecular imaging research can assist in recognizing complex patterns and, with image classification, in tumour identification and segmentation of data from PET-CT scans. Initial studies using fully automated approaches to calculate metabolic tumour volume and other PET-based biomarkers have demonstrated appropriate correlation with calculations from experts, warranting further testing in large-scale studies. The extraction of computer-based quantitative tumour characterization through radiomics can provide a comprehensive view of phenotypic heterogeneity that better captures the molecular and functional features of the disease. Additionally, radiomics can be integrated with genomic data to provide more accurate prognostic information. Further improvements in PET-based biomarkers are imminent, although their incorporation into clinical decision-making currently has methodological shortcomings that need to be addressed with confirmatory prospective validation in selected patient populations. In this Review, we discuss the current knowledge, challenges and opportunities in the integration of quantitative PET-based biomarkers in clinical trials and the routine management of patients with lymphoma.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Russ A Kuker
- Department of Radiology, Division of Nuclear Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fei Yang
- Department of Radiation Oncology, Division of Medical Physics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Craig H Moskowitz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
42
|
Ferrández MC, Golla SSV, Eertink JJ, de Vries BM, Lugtenburg PJ, Wiegers SE, Zwezerijnen GJC, Pieplenbosch S, Kurch L, Hüttmann A, Hanoun C, Dührsen U, de Vet HCW, Zijlstra JM, Boellaard R. An artificial intelligence method using FDG PET to predict treatment outcome in diffuse large B cell lymphoma patients. Sci Rep 2023; 13:13111. [PMID: 37573446 PMCID: PMC10423266 DOI: 10.1038/s41598-023-40218-1] [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: 03/31/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Convolutional neural networks (CNNs) may improve response prediction in diffuse large B-cell lymphoma (DLBCL). The aim of this study was to investigate the feasibility of a CNN using maximum intensity projection (MIP) images from 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) baseline scans to predict the probability of time-to-progression (TTP) within 2 years and compare it with the International Prognostic Index (IPI), i.e. a clinically used score. 296 DLBCL 18F-FDG PET/CT baseline scans collected from a prospective clinical trial (HOVON-84) were analysed. Cross-validation was performed using coronal and sagittal MIPs. An external dataset (340 DLBCL patients) was used to validate the model. Association between the probabilities, metabolic tumour volume and Dmaxbulk was assessed. Probabilities for PET scans with synthetically removed tumors were also assessed. The CNN provided a 2-year TTP prediction with an area under the curve (AUC) of 0.74, outperforming the IPI-based model (AUC = 0.68). Furthermore, high probabilities (> 0.6) of the original MIPs were considerably decreased after removing the tumours (< 0.4, generally). These findings suggest that MIP-based CNNs are able to predict treatment outcome in DLBCL.
Collapse
Affiliation(s)
- Maria C Ferrández
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Sandeep S V Golla
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bart M de Vries
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sanne E Wiegers
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lars Kurch
- Department of Nuclear Medicine, Clinic and Polyclinic for Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Henrica C W de Vet
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Methodology, Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Constantino CS, Leocádio S, Oliveira FPM, Silva M, Oliveira C, Castanheira JC, Silva Â, Vaz S, Teixeira R, Neves M, Lúcio P, João C, Costa DC. Evaluation of Semiautomatic and Deep Learning-Based Fully Automatic Segmentation Methods on [ 18F]FDG PET/CT Images from Patients with Lymphoma: Influence on Tumor Characterization. J Digit Imaging 2023; 36:1864-1876. [PMID: 37059891 PMCID: PMC10407010 DOI: 10.1007/s10278-023-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
The objective is to assess the performance of seven semiautomatic and two fully automatic segmentation methods on [18F]FDG PET/CT lymphoma images and evaluate their influence on tumor quantification. All lymphoma lesions identified in 65 whole-body [18F]FDG PET/CT staging images were segmented by two experienced observers using manual and semiautomatic methods. Semiautomatic segmentation using absolute and relative thresholds, k-means and Bayesian clustering, and a self-adaptive configuration (SAC) of k-means and Bayesian was applied. Three state-of-the-art deep learning-based segmentations methods using a 3D U-Net architecture were also applied. One was semiautomatic and two were fully automatic, of which one is publicly available. Dice coefficient (DC) measured segmentation overlap, considering manual segmentation the ground truth. Lymphoma lesions were characterized by 31 features. Intraclass correlation coefficient (ICC) assessed features agreement between different segmentation methods. Nine hundred twenty [18F]FDG-avid lesions were identified. The SAC Bayesian method achieved the highest median intra-observer DC (0.87). Inter-observers' DC was higher for SAC Bayesian than manual segmentation (0.94 vs 0.84, p < 0.001). Semiautomatic deep learning-based median DC was promising (0.83 (Obs1), 0.79 (Obs2)). Threshold-based methods and publicly available 3D U-Net gave poorer results (0.56 ≤ DC ≤ 0.68). Maximum, mean, and peak standardized uptake values, metabolic tumor volume, and total lesion glycolysis showed excellent agreement (ICC ≥ 0.92) between manual and SAC Bayesian segmentation methods. The SAC Bayesian classifier is more reproducible and produces similar lesion features compared to manual segmentation, giving the best concordant results of all other methods. Deep learning-based segmentation can achieve overall good segmentation results but failed in few patients impacting patients' clinical evaluation.
Collapse
Affiliation(s)
- Cláudia S Constantino
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal.
| | - Sónia Leocádio
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Francisco P M Oliveira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Mariana Silva
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Carla Oliveira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Joana C Castanheira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Ângelo Silva
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Sofia Vaz
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Ricardo Teixeira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Manuel Neves
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Paulo Lúcio
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Cristina João
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Durval C Costa
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| |
Collapse
|
44
|
Eertink JJ, Zwezerijnen GJC, Heymans MW, Pieplenbosch S, Wiegers SE, Dührsen U, Hüttmann A, Kurch L, Hanoun C, Lugtenburg PJ, Barrington SF, Mikhaeel NG, Ceriani L, Zucca E, Czibor S, Györke T, Chamuleau MED, Hoekstra OS, de Vet HCW, Boellaard R, Zijlstra JM. Baseline PET radiomics outperforms the IPI risk score for prediction of outcome in diffuse large B-cell lymphoma. Blood 2023; 141:3055-3064. [PMID: 37001036 PMCID: PMC10646814 DOI: 10.1182/blood.2022018558] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/31/2023] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
The objective of this study is to externally validate the clinical positron emission tomography (PET) model developed in the HOVON-84 trial and to compare the model performance of our clinical PET model using the international prognostic index (IPI). In total, 1195 patients with diffuse large B-cell lymphoma (DLBCL) were included in the study. Data of 887 patients from 6 studies were used as external validation data sets. The primary outcomes were 2-year progression-free survival (PFS) and 2-year time to progression (TTP). The metabolic tumor volume (MTV), maximum distance between the largest lesion and another lesion (Dmaxbulk), and peak standardized uptake value (SUVpeak) were extracted. The predictive values of the IPI and clinical PET model (MTV, Dmaxbulk, SUVpeak, performance status, and age) were tested. Model performance was assessed using the area under the curve (AUC), and diagnostic performance, using the positive predictive value (PPV). The IPI yielded an AUC of 0.62. The clinical PET model yielded a significantly higher AUC of 0.71 (P < .001). Patients with high-risk IPI had a 2-year PFS of 61.4% vs 51.9% for those with high-risk clinical PET, with an increase in PPV from 35.5% to 49.1%, respectively. A total of 66.4% of patients with high-risk IPI were free from progression or relapse vs 55.5% of patients with high-risk clinical PET scores, with an increased PPV from 33.7% to 44.6%, respectively. The clinical PET model remained predictive of outcome in 6 independent first-line DLBCL studies, and had higher model performance than the currently used IPI in all studies.
Collapse
Affiliation(s)
- J. J. Eertink
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - G. J. C. Zwezerijnen
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M. W. Heymans
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - S. Pieplenbosch
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S. E. Wiegers
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - U. Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - A. Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - L. Kurch
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Leipzig, Leipzig, Germany
| | - C. Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - P. J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - S. F. Barrington
- King’s College London and Guy’s and St Thomas’ PET Centre, School of Biomedical Engineering and Imaging Sciences, King’s Health Partners, King’s College London, London, United Kingdom
| | - N. G. Mikhaeel
- Department of Clinical Oncology, Guy’s Cancer Centre and School of Cancer and Pharmaceutical Sciences, King’s College London University, London, United Kingdom
| | - L. Ceriani
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - E. Zucca
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, IOSI - Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - S. Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - T. Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - M. E. D. Chamuleau
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - O. S. Hoekstra
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H. C. W. de Vet
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - R. Boellaard
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J. M. Zijlstra
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - PETRA Consortium
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Leipzig, Leipzig, Germany
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- King’s College London and Guy’s and St Thomas’ PET Centre, School of Biomedical Engineering and Imaging Sciences, King’s Health Partners, King’s College London, London, United Kingdom
- Department of Clinical Oncology, Guy’s Cancer Centre and School of Cancer and Pharmaceutical Sciences, King’s College London University, London, United Kingdom
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, IOSI - Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| |
Collapse
|
45
|
Barrington SF. Advances in positron emission tomography and radiomics. Hematol Oncol 2023; 41 Suppl 1:11-19. [PMID: 37294959 PMCID: PMC10775708 DOI: 10.1002/hon.3137] [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: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
Positron emission tomography is established for staging and response evaluation in lymphoma using visual evaluation and semi-quantitative analysis. Radiomic analysis involving quantitative imaging features at baseline, such as metabolic tumor volume and markers of disease dissemination and changes in the standardized uptake value during treatment are emerging as powerful biomarkers. The combination of radiomic features with clinical risk factors and genomic analysis offers the potential to improve clinical risk prediction. This review discusses the state of current knowledge, progress toward standardization of tumor delineation for radiomic analysis and argues that radiomic features, molecular markers and circulating tumor DNA should be included in clinical trial designs to enable the development of baseline and dynamic risk scores that could further advance the field to facilitate testing of novel treatments and personalized therapy in aggressive lymphomas.
Collapse
Affiliation(s)
- Sally F. Barrington
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Campus, Kings College LondonLondonUK
| |
Collapse
|
46
|
Chen K, Wang J, Li S, Zhou W, Xu W. Predictive value of 18F-FDG PET/CT-based radiomics model for neoadjuvant chemotherapy efficacy in breast cancer: a multi-scanner/center study with external validation. Eur J Nucl Med Mol Imaging 2023; 50:1869-1880. [PMID: 36808002 DOI: 10.1007/s00259-023-06150-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/12/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE To develop and validate the predictive value of an 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) model for breast cancer neoadjuvant chemotherapy (NAC) efficacy based on the tumor-to-liver ratio (TLR) radiomic features and multiple data pre-processing methods. METHODS One hundred and ninety-three breast cancer patients from multiple centers were retrospectively included in this study. According to the endpoint of NAC, we divided the patients into pathological complete remission (pCR) and non-pCR groups. All patients underwent 18F-FDG PET/CT imaging before NAC treatment, and CT and PET images volume of interest (VOI) segmentation by manual segmentation and semi-automated absolute threshold segmentation, respectively. Then, feature extraction of VOI was performed with the pyradiomics package. A total of 630 models were created based on the source of radiomic features, the elimination of the batch effect approach, and the discretization method. The differences in data pre-processing approaches were compared and analyzed to identify the best-performing model, which was further tested by the permutation test. RESULTS A variety of data pre-processing methods contributed in varying degrees to the improvement of model effects. Among them, TLR radiomic features and Combat and Limma methods that eliminate batch effects could enhance the model prediction overall, and data discretization could be used as a potential method that can further optimize the model. A total of seven excellent models were selected and then based on the AUC of each model in the four test sets and their standard deviations, we selected the optimal model. The optimal model predicted AUC between 0.7 and 0.77 for the four test groups, with p-values less than 0.05 for the permutation test. CONCLUSION It is necessary to enhance the predictive effect of the model by eliminating confounding factors through data pre-processing. The model developed in this way is effective in predicting the efficacy of NAC for breast cancer.
Collapse
Affiliation(s)
- Kun Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Jian Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Shuai Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Wen Zhou
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| | - Wengui Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
47
|
Jiang L, Guo S, Zhao Y, Cheng Z, Zhong X, Zhou P. Predicting Extrathyroidal Extension in Papillary Thyroid Carcinoma Using a Clinical-Radiomics Nomogram Based on B-Mode and Contrast-Enhanced Ultrasound. Diagnostics (Basel) 2023; 13:diagnostics13101734. [PMID: 37238217 DOI: 10.3390/diagnostics13101734] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common pathological type of thyroid cancer. PTC patients with extrathyroidal extension (ETE) are associated with poor prognoses. The preoperative accurate prediction of ETE is crucial for helping the surgeon decide on the surgical plan. This study aimed to establish a novel clinical-radiomics nomogram based on B-mode ultrasound (BMUS) and contrast-enhanced ultrasound (CEUS) for the prediction of ETE in PTC. A total of 216 patients with PTC between January 2018 and June 2020 were collected and divided into the training set (n = 152) and the validation set (n = 64). The least absolute shrinkage and selection operator (LASSO) algorithm was applied for radiomics feature selection. Univariate analysis was performed to find clinical risk factors for predicting ETE. The BMUS Radscore, CEUS Radscore, clinical model, and clinical-radiomics model were established using multivariate backward stepwise logistic regression (LR) based on BMUS radiomics features, CEUS radiomics features, clinical risk factors, and the combination of those features, respectively. The diagnostic efficacy of the models was assessed using receiver operating characteristic (ROC) curves and the DeLong test. The model with the best performance was then selected to develop a nomogram. The results show that the clinical-radiomics model, which is constructed by age, CEUS-reported ETE, BMUS Radscore, and CEUS Radscore, showed the best diagnostic efficiency in both the training set (AUC = 0.843) and validation set (AUC = 0.792). Moreover, a clinical-radiomics nomogram was established for easier clinical practices. The Hosmer-Lemeshow test and the calibration curves demonstrated satisfactory calibration. The decision curve analysis (DCA) showed that the clinical-radiomics nomogram had substantial clinical benefits. The clinical-radiomics nomogram constructed from the dual-modal ultrasound can be exploited as a promising tool for the pre-operative prediction of ETE in PTC.
Collapse
Affiliation(s)
- Liqing Jiang
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shiyan Guo
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yongfeng Zhao
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhe Cheng
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinyu Zhong
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ping Zhou
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
48
|
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
|
49
|
Veziroglu EM, Farhadi F, Hasani N, Nikpanah M, Roschewski M, Summers RM, Saboury B. Role of Artificial Intelligence in PET/CT Imaging for Management of Lymphoma. Semin Nucl Med 2023; 53:426-448. [PMID: 36870800 DOI: 10.1053/j.semnuclmed.2022.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 03/06/2023]
Abstract
Our review shows that AI-based analysis of lymphoma whole-body FDG-PET/CT can inform all phases of clinical management including staging, prognostication, treatment planning, and treatment response evaluation. We highlight advancements in the role of neural networks for performing automated image segmentation to calculate PET-based imaging biomarkers such as the total metabolic tumor volume (TMTV). AI-based image segmentation methods are at levels where they can be semi-automatically implemented with minimal human inputs and nearing the level of a second-opinion radiologist. Advances in automated segmentation methods are particularly apparent in the discrimination of lymphomatous vs non-lymphomatous FDG-avid regions, which carries through to automated staging. Automated TMTV calculators, in addition to automated calculation of measures such as Dmax are informing robust models of progression-free survival which can then feed into improved treatment planning.
Collapse
Affiliation(s)
| | - Faraz Farhadi
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD; Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Navid Hasani
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Moozhan Nikpanah
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD; Department of Radiology, University of Alabama at Birmingham, AL
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ronald M Summers
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD; Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD
| | - Babak Saboury
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
50
|
Albano D, Treglia G, Dondi F, Calabrò A, Rizzo A, Annunziata S, Guerra L, Morbelli S, Tucci A, Bertagna F. 18F-FDG PET/CT Maximum Tumor Dissemination (Dmax) in Lymphoma: A New Prognostic Factor? Cancers (Basel) 2023; 15:cancers15092494. [PMID: 37173962 PMCID: PMC10177347 DOI: 10.3390/cancers15092494] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Recently, several studies introduced the potential prognostic usefulness of maximum tumor dissemination (Dmax) measured by 2-deoxy-2-fluorine-18-fluoro-D-glucose positron-emission tomography/computed tomography (18F-FDG PET/CT). Dmax is a simple three-dimensional feature that represents the maximal distance between the two farthest hypermetabolic PET lesions. A comprehensive computer literature search of PubMed/MEDLINE, Embase, and Cochrane libraries was conducted, including articles indexed up to 28 February 2023. Ultimately, 19 studies analyzing the value of 18F-FDG PET/CT Dmax in patients with lymphomas were included. Despite their heterogeneity, most studies showed a significant prognostic role of Dmax in predicting progression-free survival (PFS) and overall survival (OS). Some articles showed that the combination of Dmax with other metabolic features, such as MTV and interim PET response, proved to better stratify the risk of relapse or death. However, some methodological open questions need to be clarified before introducing Dmax into clinical practice.
Collapse
Affiliation(s)
- Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Anna Calabrò
- Division of Nuclear Medicine, Università degli Studi di Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Luca Guerra
- Nuclear Medicine Division, Ospedale San Gerardo, 20900 Monza, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | | | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|