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
|
Jiang C, Qian C, Jiang Q, Zhou H, Jiang Z, Teng Y, Xu B, Li X, Ding C, Tian R. Virtual biopsy for non-invasive identification of follicular lymphoma histologic transformation using radiomics-based imaging biomarker from PET/CT. BMC Med 2025; 23:49. [PMID: 39875864 PMCID: PMC11776338 DOI: 10.1186/s12916-025-03893-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND This study aimed to construct a radiomics-based imaging biomarker for the non-invasive identification of transformed follicular lymphoma (t-FL) using PET/CT images. METHODS A total of 784 follicular lymphoma (FL), diffuse large B-cell lymphoma, and t-FL patients from 5 independent medical centers were included. The unsupervised EMFusion method was applied to fuse PET and CT images. Deep-based radiomic features were extracted from the fusion images using a deep learning model (ResNet18). These features, along with handcrafted radiomics, were utilized to construct a radiomic signature (R-signature) using automatic machine learning in the training and internal validation cohort. The R-signature was then tested for its predictive ability in the t-FL test cohort. Subsequently, this R-signature was combined with clinical parameters and SUVmax to develop a t-FL scoring system. RESULTS The R-signature demonstrated high accuracy, with mean AUC values as 0.994 in the training cohort and 0.976 in the internal validation cohort. In the t-FL test cohort, the R-signature achieved an AUC of 0.749, with an accuracy of 75.2%, sensitivity of 68.0%, and specificity of 77.5%. Furthermore, the t-FL scoring system, incorporating the R-signature along with clinical parameters (age, LDH, and ECOG PS) and SUVmax, achieved an AUC of 0.820, facilitating the stratification of patients into low, medium, and high transformation risk groups. CONCLUSIONS This study offers a promising approach for identifying t-FL non-invasively by radiomics analysis on PET/CT images. The developed t-FL scoring system provides a valuable tool for clinical decision-making, potentially improving patient management and outcomes.
Collapse
Affiliation(s)
- Chong Jiang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Guoxue Alley, Address: No.37, Chengdu City, Sichuan, 610041, China
| | - Chunjun Qian
- School of Electrical and Information Engineering, Changzhou Institute of Technology, Changzhou, Jiangsu, China
- The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Center of Medical Physics, Nanjing Medical University, Changzhou, China
| | - Qiuhui Jiang
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, Xiamen, China
| | - Hang Zhou
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, No.107, Wenhua Xilu, Lixia District, Jinan City, Shandong, 250012, China
| | - Zekun Jiang
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Teng
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bing Xu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, Xiamen, China
| | - Xin Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, No.107, Wenhua Xilu, Lixia District, Jinan City, Shandong, 250012, China.
| | - Chongyang Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No.321, Zhongshan Road, Nanjing City, Jiangsu Province, 210008, China.
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Guoxue Alley, Address: No.37, Chengdu City, Sichuan, 610041, China.
| |
Collapse
|
3
|
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 PMCID: PMC12099142 DOI: 10.1177/15330338251342860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/15/2025] [Accepted: 04/28/2025] [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
|
4
|
Hasanabadi S, Aghamiri SMR, Abin AA, Abdollahi H, Arabi H, Zaidi H. Enhancing Lymphoma Diagnosis, Treatment, and Follow-Up Using 18F-FDG PET/CT Imaging: Contribution of Artificial Intelligence and Radiomics Analysis. Cancers (Basel) 2024; 16:3511. [PMID: 39456604 PMCID: PMC11505665 DOI: 10.3390/cancers16203511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Lymphoma, encompassing a wide spectrum of immune system malignancies, presents significant complexities in its early detection, management, and prognosis assessment since it can mimic post-infectious/inflammatory diseases. The heterogeneous nature of lymphoma makes it challenging to definitively pinpoint valuable biomarkers for predicting tumor biology and selecting the most effective treatment strategies. Although molecular imaging modalities, such as positron emission tomography/computed tomography (PET/CT), specifically 18F-FDG PET/CT, hold significant importance in the diagnosis of lymphoma, prognostication, and assessment of treatment response, they still face significant challenges. Over the past few years, radiomics and artificial intelligence (AI) have surfaced as valuable tools for detecting subtle features within medical images that may not be easily discerned by visual assessment. The rapid expansion of AI and its application in medicine/radiomics is opening up new opportunities in the nuclear medicine field. Radiomics and AI capabilities seem to hold promise across various clinical scenarios related to lymphoma. Nevertheless, the need for more extensive prospective trials is evident to substantiate their reliability and standardize their applications. This review aims to provide a comprehensive perspective on the current literature regarding the application of AI and radiomics applied/extracted on/from 18F-FDG PET/CT in the management of lymphoma patients.
Collapse
Affiliation(s)
- Setareh Hasanabadi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Seyed Mahmud Reza Aghamiri
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Ahmad Ali Abin
- Faculty of Computer Science and Engineering, Shahid Beheshti University, Tehran 1983969411, Iran;
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, 500 Odense, Denmark
- University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| |
Collapse
|
5
|
Chauvie S, Castellino A, Bergesio F, De Maggi A, Durmo R. Lymphoma: The Added Value of Radiomics, Volumes and Global Disease Assessment. PET Clin 2024; 19:561-568. [PMID: 38910057 DOI: 10.1016/j.cpet.2024.05.009] [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: 06/25/2024]
Abstract
Lymphoma represents a condition that holds promise for cure with existing treatment modalities; nonetheless, the primary clinical obstacle lies in advancing therapeutic outcomes by pinpointing high-risk individuals who are unlikely to respond favorably to standard therapy. In this article, the authors will delineate the significant strides achieved in the lymphoma field, with a particular emphasis on the 3 prevalent subtypes: Hodgkin lymphoma, diffuse large B-cell lymphomas, and follicular lymphoma.
Collapse
Affiliation(s)
- Stéphane Chauvie
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy.
| | | | - Fabrizio Bergesio
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy
| | - Adriano De Maggi
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy
| | - Rexhep Durmo
- Nuclear Medicine Division, Department of Radiology, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
6
|
An S, Huang G, Yu X, Liu J, Chen Y. The added diagnostic value of 18 F-FDG PET/CT radiomic analysis in multiple myeloma patients with negative visual analysis. Nucl Med Commun 2024; 45:244-252. [PMID: 38165165 DOI: 10.1097/mnm.0000000000001809] [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: 01/03/2024]
Abstract
PURPOSE A small number of patients diagnosed with multiple myeloma (MM) by bone marrow aspiration reported as being disease-free on 18 F-FDG PET/CT. We aim to evaluate the diagnostic value of radiomics approach in patients with MM who were negative by visual analysis. MATERIALS AND METHODS Thirty-three patients judged negative by visual analysis were assigned to the MM group. Contemporaneous 31 disease-free patients served as the control group. 70% of the whole data set was used as training set (23 from MM group and 22 from control group) and 30% as testing set (10 from MM group and 9 from control group). Axial skeleton volumes were automatically segmented and high-dimensional imaging features were extracted from PET and CT. The unsupervised machine learning method was used to filter and reduce the dimensions of the extracted features. Random forest was used to construct the prediction model and then validated with 10-fold cross-validation and evaluated on the independent testing set. RESULTS One thousand seven hundred two quantitative features were extracted from PET and CT. Of those, three first-order and one high-order imaging features were uncorrelated. With the cross-validation on the training group, the sensitivity, specificity, accuracy and area under the curve of random forest were 0.850, 0.792, 0.818 and 0.894, respectively. On the independent testing set, the accuracy of the model was 0.850 and the area under the curve was 0.909. CONCLUSION Radiomic analysis based on 18 F-FDG PET/CT using machine learning model provides a quantitative, objective and efficient mechanism for diagnosing patients with MM who were negative by visual analysis.
Collapse
Affiliation(s)
- Shuxian An
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
7
|
Feng L, Zhang S, Wang C, Li S, Kan Y, Wang C, Zhang H, Wang W, Yang J. Axial Skeleton Radiomics of 18F-FDG PET/CT: Impact on Event-Free Survival Prediction in High-Risk Pediatric Neuroblastoma. Acad Radiol 2023; 30:2487-2496. [PMID: 36828720 DOI: 10.1016/j.acra.2023.01.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/25/2023]
Abstract
OBJECTIVES To construct and validate a combined model based on axial skeleton radiomics of 18F-FDG PET/CT for predicting event-free survival in high-risk pediatric neuroblastoma patients. MATERIALS AND METHODS Eighty-seven high-risk neuroblastoma patients were retrospectively enrolled in this study and randomized in a 7:3 ratio to the training and validation cohorts. The radiomics model was constructed using radiomics features that were extracted from the axial skeleton. A univariate Cox regression analysis was then performed to screen clinical risk factors associated with event-free survival for building clinical model. Radiomics features and clinical risk factors were incorporated to construct the combined model for predicting the event-free survival in high-risk neuroblastoma patients. The performance of the models was evaluated by the C-index. RESULTS Eighteen radiomics features were selected to build the radiomics model. The radiomics model achieved better event-free survival prediction than the clinical model in the training cohort (C-index: 0.846 vs. 0.612) and validation cohort (C-index: 0.754 vs. 0.579). The combined model achieved the best prognostic prediction performance with a C-index of 0.863 and 0.799 in the training and validation cohorts, respectively. CONCLUSION The combined model integrating radiomics features and clinical risk factors showed more accurate predictive performance for event-free survival in high-risk pediatric neuroblastoma patients, which helps to design individualized treatment strategies and regular follow-ups.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Shuxin Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Chaoran Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Siqi Li
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China.
| |
Collapse
|
8
|
Development and validation of a [18F]FDG PET/CT-based radiomics nomogram to predict the prognostic risk of pretreatment diffuse large B cell lymphoma patients. Eur Radiol 2022; 33:3354-3365. [PMID: 36547676 PMCID: PMC10121518 DOI: 10.1007/s00330-022-09301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/03/2022] [Accepted: 11/13/2022] [Indexed: 12/24/2022]
Abstract
Abstract
Objective
In this study, based on PET/CT radiomics features, we developed and validated a nomogram to predict progression-free survival (PFS) for cases with diffuse large B cell lymphoma (DLBCL) treated with immunochemotherapy.
Methods
This study retrospectively recruited 129 cases with DLBCL. Among them, PET/CT scans were conducted and baseline images were collected for radiomics features along with their clinicopathological features. Radiomics features related to recurrence were screened for survival analysis using univariate Cox regression analysis with p < 0.05. Next, a weighted Radiomics-score (Rad-score) was generated and independent risk factors were obtained from univariate and multivariate Cox regressions to build the nomogram. Furthermore, the nomogram was tested for their ability to predict PFS using time-dependent receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA).
Results
Blood platelet, Rad-score, and gender were included in the nomogram as independent DLBCL risk factors for PFS. We found that the training cohort areas under the curve (AUCs) were 0.79, 0.84, and 0.88, and validation cohort AUCs were 0.67, 0.83, and 0.72, respectively. Further, the DCA and calibration curves confirmed the predictive nomogram’s clinical relevance.
Conclusion
Using Rad-score, blood platelet, and gender of the DLBCL patients, a PET/CT radiomics-based nomogram was developed to guide cases’ recurrence risk assessment prior to treatment. The developed nomogram can help provide more appropriate treatment plans to the cases.
Key Points
• DLBCL cases can be classified into low- and high-risk groups using PET/CT radiomics based Rad-score.
• When combined with other clinical characteristics (gender and blood platelet count), Rad-score can be used to predict the outcome of the pretreatment of DLBCL cases with a certain degree of accuracy.
• A prognostic nomogram was established in this study in order to aid in assessing prognostic risk and providing more accurate treatment plans for DLBCL cases.
Collapse
|
9
|
Deng H, Zhou Y, Lu W, Chen W, Yuan Y, Li L, Shu H, Zhang P, Ye X. Development and validation of nomograms by radiomic features on ultrasound imaging for predicting overall survival in patients with primary nodal diffuse large B-cell lymphoma. Front Oncol 2022; 12:991948. [PMID: 36568168 PMCID: PMC9768489 DOI: 10.3389/fonc.2022.991948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives To develop and validate a nomogram to predict the overall survival (OS) of patients with primary nodal diffuse large B-cell lymphoma(N-DLBCL) based on radiomic features and clinical features. Materials and methods A retrospective analysis was performed on 145 patients confirmed with N-DLBCL and they were randomly assigned to training set(n=78), internal validation set(n=33), external validation set(n=34). First, a clinical model (model 1) was established according to clinical features and ultrasound (US) results. Then, based on the radiomics features extracted from conventional ultrasound images, a radiomic signature was constructed (model 2), and the radiomics score (Rad-Score) was calculated. Finally, a comprehensive model was established (model 3) combined with Rad-score and clinical features. Receiver operating characteristic (ROC) curves were employed to evaluate the performance of model 1, model 2 and model 3. Based on model 3, we plotted a nomogram. Calibration curves were used to test the effectiveness of the nomogram, and decision curve analysis (DCA) was used to asset the nomogram in clinical use. Results According to multivariate analysis, 3 clinical features and Rad-score were finally selected to construct the model 3, which showed better predictive value for OS in patients with N-DLBCL than mode 1 and model 2 in training (AUC,0. 891 vs. 0.779 vs.0.756), internal validation (AUC, 0.868 vs. 0.713, vs.0.756) and external validation (AUC, 914 vs. 0.866, vs.0.789) sets. Decision curve analysis demonstrated that the nomogram based on model 3 was more clinically useful than the other two models. Conclusion The developed nomogram is a useful tool for precisely analyzing the prognosis of N-DLBCL patients, which could help clinicians in making personalized survival predictions and assessing individualized clinical options.
Collapse
Affiliation(s)
- Hongyan Deng
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yasu Zhou
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjuan Lu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenqin Chen
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ya Yuan
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hua Shu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pingyang Zhang
- Department of Cardiovascular Ultrasound, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xinhua Ye, ; Pingyang Zhang,
| | - Xinhua Ye
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xinhua Ye, ; Pingyang Zhang,
| |
Collapse
|
10
|
Feng L, Yang X, Lu X, Kan Y, Wang C, Zhang H, Wang W, Yang J. Diagnostic Value of 18F-FDG PET/CT-Based Radiomics Nomogram in Bone Marrow Involvement of Pediatric Neuroblastoma. Acad Radiol 2022; 30:940-951. [PMID: 36117128 DOI: 10.1016/j.acra.2022.08.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/06/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To develop and validate an 18F-FDG PET/CT-based radiomics nomogram and evaluate the value of the 18F-FDG PET/CT-based radiomics nomogram for the diagnosis of bone marrow involvement (BMI) in pediatric neuroblastoma. MATERIALS AND METHODS A total of 144 patients with neuroblastoma (100 in the training cohort and 44 in the validation cohort) were retrospectively included. The PET/CT images of patients were visually assessed. The results of bone marrow aspirates or biopsies were used as the gold standard for BMI. Radiomics features and conventional PET parameters were extracted using the 3D slicer. Features were selected by the least absolute shrinkage and selection operator regression, and radiomics signature was constructed. Univariate and multivariate logistic regression analyses were applied to identify the independent clinical risk factors and construct the clinical model. Other different models, including the conventional PET model, combined PET-clinical model and combined radiomics model, were built using logistic regression. The combined radiomics model was based on clinical factors, conventional PET parameters and radiomics signature, which was presented as a radiomics nomogram. The diagnostic performance of the different models was evaluated by receiver operating characteristic (ROC) curves and decision curve analysis (DCA). RESULTS By visual assessment, BMI was observed in 80 patients. Four conventional PET parameters (SUVmax, SUVmean, metabolic tumor volume, and total lesion glycolysis) were extracted. And 15 radiomics features were selected to build the radiomics signature. The 11q aberration, neuron-specific enolase and vanillylmandelic acid were identified as the independent clinical risk factors to establish the clinical model. The radiomics nomogram incorporating the radiomics signature, the independent clinical risk factors and SUVmean demonstrated the best diagnostic value for identifying BMI, with an area under the curve (AUC) of 0.963 and 0.931 in the training and validation cohorts, respectively. And the DCA demonstrated that the radiomics nomogram was clinically useful. CONCLUSION The 18F-FDG PET/CT-based radiomics nomogram which incorporates radiomics signature, independent clinical risk factors and conventional PET parameters could improve the diagnostic performance for BMI of pediatric neuroblastoma without additional medical costs and radiation exposure.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Xu Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Chao Wang
- Sinounion Medical Technology (Beijing) Co., Ltd. Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China.
| |
Collapse
|
11
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 2, Infradiaphragmatic Cancers, Blood Malignancies, Melanoma and Musculoskeletal Cancers. Diagnostics (Basel) 2022; 12:diagnostics12061330. [PMID: 35741139 PMCID: PMC9222024 DOI: 10.3390/diagnostics12061330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
The objective of this review was to summarize published radiomics studies dealing with infradiaphragmatic cancers, blood malignancies, melanoma, and musculoskeletal cancers, and assess their quality. PubMed database was searched from January 1990 to February 2022 for articles performing radiomics on PET imaging of at least 1 specified tumor type. Exclusion criteria includd: non-oncological studies; supradiaphragmatic tumors; reviews, comments, cases reports; phantom or animal studies; technical articles without a clinically oriented question; studies including <30 patients in the training cohort. The review database contained PMID, first author, year of publication, cancer type, number of patients, study design, independent validation cohort and objective. This database was completed twice by the same person; discrepant results were resolved by a third reading of the articles. A total of 162 studies met inclusion criteria; 61 (37.7%) studies included >100 patients, 13 (8.0%) were prospective and 61 (37.7%) used an independent validation set. The most represented cancers were esophagus, lymphoma, and cervical cancer (n = 24, n = 24 and n = 19 articles, respectively). Most studies focused on 18F-FDG, and prognostic and response to treatment objectives. Although radiomics and artificial intelligence are technically challenging, new contributions and guidelines help improving research quality over the years and pave the way toward personalized medicine.
Collapse
Affiliation(s)
- David Morland
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
- Correspondence:
| | - Elizabeth Katherine Anna Triumbari
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Unità di Radioterapia Oncologica, Radiomics, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
12
|
Kostakoglu L, Dalmasso F, Berchialla P, Pierce LA, Vitolo U, Martelli M, Sehn LH, Trněný M, Nielsen TG, Bolen CR, Sahin D, Lee C, El‐Galaly TC, Mattiello F, Kinahan PE, Chauvie S. A prognostic model integrating PET-derived metrics and image texture analyses with clinical risk factors from GOYA. EJHAEM 2022; 3:406-414. [PMID: 35846039 PMCID: PMC9175666 DOI: 10.1002/jha2.421] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/05/2022]
Abstract
Image texture analysis (radiomics) uses radiographic images to quantify characteristics that may identify tumour heterogeneity and associated patient outcomes. Using fluoro-deoxy-glucose positron emission tomography/computed tomography (FDG-PET/CT)-derived data, including quantitative metrics, image texture analysis and other clinical risk factors, we aimed to develop a prognostic model that predicts survival in patients with previously untreated diffuse large B-cell lymphoma (DLBCL) from GOYA (NCT01287741). Image texture features and clinical risk factors were combined into a random forest model and compared with the international prognostic index (IPI) for DLBCL based on progression-free survival (PFS) and overall survival (OS) predictions. Baseline FDG-PET scans were available for 1263 patients, 832 patients of these were cell-of-origin (COO)-evaluable. Patients were stratified by IPI or radiomics features plus clinical risk factors into low-, intermediate- and high-risk groups. The random forest model with COO subgroups identified a clearer high-risk population (45% 2-year PFS [95% confidence interval (CI) 40%-52%]; 65% 2-year OS [95% CI 59%-71%]) than the IPI (58% 2-year PFS [95% CI 50%-67%]; 69% 2-year OS [95% CI 62%-77%]). This study confirms that standard clinical risk factors can be combined with PET-derived image texture features to provide an improved prognostic model predicting survival in untreated DLBCL.
Collapse
Affiliation(s)
- Lale Kostakoglu
- Department of Radiology and Medical ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | | | - Paola Berchialla
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| | - Larry A. Pierce
- Department of RadiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient ClinicCandiolo Cancer InstituteCandioloItaly
| | - Maurizio Martelli
- HematologyDepartment of Translational and Precision MedicineSapienza UniversityRomeItaly
| | - Laurie H. Sehn
- BC Cancer Center for Lymphoid Cancer and the University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Marek Trněný
- 1st Faculty of MedicineCharles University General HospitalPragueCzech Republic
| | | | | | | | - Calvin Lee
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | | | - Paul E. Kinahan
- Department of RadiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Stephane Chauvie
- Department of Clinical and Biological SciencesUniversity of TurinTurinItaly
| |
Collapse
|
13
|
Reinert CP, Perl RM, Faul C, Lengerke C, Nikolaou K, Dittmann H, Bethge WA, Horger M. Value of CT-Textural Features and Volume-Based PET Parameters in Comparison to Serologic Markers for Response Prediction in Patients with Diffuse Large B-Cell Lymphoma Undergoing CD19-CAR-T Cell Therapy. J Clin Med 2022; 11:jcm11061522. [PMID: 35329846 PMCID: PMC8951429 DOI: 10.3390/jcm11061522] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
The goal of this study was to investigate the value of CT-textural features and volume-based PET parameters in comparison to serologic markers for response prediction in patients with diffuse large B-cell lymphoma (DLBCL) undergoing cluster of differentiation (CD19)-chimeric antigen receptor (CAR)-T cell therapy. We retrospectively analyzed the whole-body (WB)-metabolic tumor volume (MTV), the WB-total lesion glycolysis (TLG) and first order textural features derived from 18F-FDG-PET/CT, as well as serologic parameters (C-reactive protein [CRP] and lactate dehydrogenase [LDH], leucocytes) prior and after CAR-T cell therapy in 21 patients with DLBCL (57.7 ± 14.7 year; 7 female). Interleukin 6 (IL-6) and IL-2 receptor peaks were monitored after treatment onset and compared with patient outcome judged by follow-up 18F-FDG-PET/CT. In 12/21 patients (57%), complete remission (CR) was observed, whereas 9/21 patients (43%) showed partial remission (PR). At baseline, WB-MTV and WB-TLG were lower in patients achieving CR (35 ± 38 mL and 319 ± 362) compared to patients achieving PR (88 ± 110 mL and 1487 ± 2254; p < 0.05). The “entropy” proved lower (1.81 ± 0.09) and “uniformity” higher (0.33 ± 0.02) in patients with CR compared to PR (2.08 ± 0.22 and 0.28 ± 0.47; p < 0.05). Patients achieving CR had lower levels of CRP, LDH and leucocytes at baseline compared to patients achieving PR (p < 0.05). In the entire cohort, WB-MTV and WB-TLG decreased after therapy onset (p < 0.01) becoming not measurable in the CR-group. Leucocytes and CRP significantly dropped after therapy (p < 0.01). The IL-6 and IL-2R peaks after therapy were lower in patients with CR compared to PR (p > 0.05). In conclusion, volume-based PET parameters derived from PET/CT and CT-textural features have the potential to predict therapy response in patients with DLBCL undergoing CAR-T cell therapy.
Collapse
Affiliation(s)
- Christian Philipp Reinert
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (R.M.P.); (K.N.); (M.H.)
- Correspondence: ; Tel.: +49-7071-298-7212; Fax: +49-7071-295-845
| | - Regine Mariette Perl
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (R.M.P.); (K.N.); (M.H.)
| | - Christoph Faul
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (C.F.); (C.L.); (W.A.B.)
| | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (C.F.); (C.L.); (W.A.B.)
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (R.M.P.); (K.N.); (M.H.)
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University of Tuebingen, 72074 Tuebingen, Germany
| | - Helmut Dittmann
- Department of Radiology, Nuclear Medicine, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany;
| | - Wolfgang A. Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (C.F.); (C.L.); (W.A.B.)
| | - Marius Horger
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (R.M.P.); (K.N.); (M.H.)
| |
Collapse
|
14
|
Comparison of FDG PET/CT and Bone Marrow Biopsy Results in Patients with Diffuse Large B Cell Lymphoma with Subgroup Analysis of PET Radiomics. Diagnostics (Basel) 2022; 12:diagnostics12010222. [PMID: 35054389 PMCID: PMC8774933 DOI: 10.3390/diagnostics12010222] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 01/06/2023] Open
Abstract
Whether FDG PET/CT can replace bone marrow biopsy (BMBx) is undecided in patients with diffuse large B cell lymphoma (DLBCL). We compared the visual PET findings and PET radiomic features, with BMBx results. A total of 328 patients were included; 269 (82%) were PET-negative and 59 (18%) were PET-positive for bone lesions on visual assessment. A fair degree of agreement was present between PET and BMBx findings (ĸ = 0.362, p < 0.001). Bone involvement on PET/CT lead to stage IV in 12 patients, despite no other evidence of extranodal lesion. Of 35 discordant PET-positive and BMBx-negative cases, 22 (63%) had discrete bone uptake on PET/CT. A total of 144 patients were eligible for radiomic analysis, and two grey-level zone-length matrix derived parameters obtained from the iliac crests showed a trend for higher values in the BMBx-positive group compared to the BMBx-negative group (mean 436.6 ± 449.0 versus 227.2 ± 137.8, unadjusted p = 0.037 for high grey-level zone emphasis; mean 308.8 ± 394.4 versus 135.7 ± 97.2, unadjusted p = 0.048 for short-zone high grey-level emphasis), but statistical significance was not found after multiple comparison correction. Visual FDG PET/CT assessment and BMBx results were discordant in 17% of patients with newly diagnosed DLBCL, and the two tests are complementary in the evaluation of bone involvement.
Collapse
|
15
|
Jiang H, Li A, Ji Z, Tian M, Zhang H. Role of Radiomics-Based Baseline PET/CT Imaging in Lymphoma: Diagnosis, Prognosis, and Response Assessment. Mol Imaging Biol 2022; 24:537-549. [PMID: 35031945 DOI: 10.1007/s11307-022-01703-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
Radiomic analysis provides information on the underlying tumour heterogeneity in lymphoma, reflecting the real-time evolution of malignancy. 2-Deoxy-2-[18F] fluoro-D-glucose positron emission tomography ([18F] FDG PET/CT) imaging is recommended before, during, and at the end of treatment for almost all lymphoma patients. This methodology offers high specificity and sensitivity, which can aid in accurate staging and assist in prompt treatment. Pretreatment [18F] FDG PET/CT-based radiomics facilitates improved diagnostic ability, guides individual treatment regimens, and boosts outcome prognosis based on heterogeneity as well as the biological, pathological, and metabolic status of the lymphoma. This technique has attracted considerable attention given its numerous applications in medicine. In the current review, we will briefly describe the basic radiomics workflow and types of radiomic features. Details of current applications of baseline [18F] FDG PET/CT-based radiomics in lymphoma will be discussed, such as differential diagnosis from other primary malignancies, diagnosis of bone marrow involvement, and response and prognostic prediction. We will also describe how this technique provides a unique noninvasive platform to assess tumour heterogeneity. Newly emerging PET radiotracers and multimodality technology will improve diagnostic specificity and further clarify tumor biology and even genetic variations in lymphoma, potentially promoting the development of precision medicine.
Collapse
Affiliation(s)
- Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ang Li
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Zhongyou Ji
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, 8 Hangzhou, Hangzhou, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, 8 Hangzhou, Hangzhou, China. .,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
FDG PET/CT versus Bone Marrow Biopsy for Diagnosis of Bone Marrow Involvement in Non-Hodgkin Lymphoma: A Systematic Review. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12020540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The management of non-Hodgkin lymphoma (NHL) patients requires the identification of bone marrow involvement (BMI) using a bone marrow biopsy (BMB), as recommended by international guidelines. Multiple studies have shown that [18F]FDG positron emission tomography, combined with computed tomography (PET/CT), may provide important information and may detect BMI, but there is still an ongoing debate as to whether it is sensitive enough for NHL patients in order to replace or be used as a complimentary method to BMB. The objective of this article is to systematically review published studies on the performance of [18F]FDG PET/CT in detecting BMI compared to the BMB for NHL patients. A population, intervention, comparison, and outcome (PICO) search in PubMed and Scopus databases (until 1 November 2021) was performed. A total of 41 studies, comprising 6147 NHL patients, were found to be eligible and were included in the analysis conducted in this systematic review. The sensitivity and specificity for identifying BMI in NHL patients were 73% and 90% for [18F]FDG PET/CT and 56% and 100% for BMB. For aggressive NHL, the sensitivity and specificity to assess the BMI for the [18F]FDG PET/CT was 77% and 94%, while for the BMB it was 58% and 100%. However, sensitivity and specificity to assess the BMI for indolent NHL for the [18F]FDG PET/CT was 59% and 85%, while for the BMB it was superior, and equal to 94% and 100%. With regard to NHL, a [18F]FDG PET/CT scan can only replace BMB if it is found to be positive and if patients can be categorized as having advanced staged NHL with high certainty. [18F]FDG PET/CT might recover tumors missed by BMB, and is recommended for use as a complimentary method, even in indolent histologic subtypes of NHL.
Collapse
|
17
|
Kim M, Ahn SY, Ahn JS, Song GY, Jung SH, Lee JJ, Kim HJ, Lee JH, Shin MG, Song SY, Yang DH. Diagnostic Accuracy and Prognostic Relevance of Immunoglobulin Heavy Chain Rearrangement and 18F-FDG-PET/CT Compared With Unilateral Bone Marrow Trephination for Detecting Bone Marrow Involvement in Patients With Diffuse Large B-Cell Lymphoma. J Korean Med Sci 2022; 37:e2. [PMID: 34981678 PMCID: PMC8723897 DOI: 10.3346/jkms.2022.37.e2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND In diffuse large B-cell lymphoma (DLBCL), bone marrow involvement (BMI) has an important clinical implication as a component of staging and International Prognostic Index. This study aimed to determine whether molecular analysis of immunoglobulin heavy chain (IgH) genes and positron emission tomography-computed tomography (PET/CT) could overcome the limitation of defining morphologic BMI by trephination biopsy and could increase the diagnostic accuracy or prognostic prediction. METHODS A total of 94 de novo patients with DLBCL underwent PET/CT, polymerase chain reaction (PCR) test for detection of IgH gene rearrangement, and unilateral bone marrow (BM) trephination at diagnosis. RESULTS A total of 9 patients (9.6%) were confirmed to present morphologic BMI (mBMI) based on trephination biopsy. On the other hand, 21 patients (22.3%) were confirmed to have IgH clonality (IgH BMI), while 16 (17.0%) were classified with BMI based on the assessment of PET/CT (PET BMI). Each IgH rearrangement PCR and PET/CT showed the high negative predictive value of detecting the BMI. However, the combined assessment of IgH rearrangement and PET/CT could increase the diagnostic accuracy and specificity with 87.2% and 97.0%, respectively. The survival outcome of patients with double positive PET BMI and IgH BMI was significantly worse than that with either single positive PET BMI or IgH BMI, and even less than patients with neither PET BMI nor IgH BMI (3-year PFS: 50.0% vs. 75.4% vs. 97.9%, P = 0.007, 3-year OS: 50.0% vs. 75.6% vs. 80.1%, P = 0.035, respectively). CONCLUSION This study suggests that the combined evaluation of PET/CT and IgH rearrangement could give additional information for predicting therapeutic outcomes in patients with negative morphologic BMI as an important part of the prognosis.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biopsy/methods
- Bone Marrow Examination
- Bone Marrow Neoplasms/diagnostic imaging
- Bone Marrow Neoplasms/genetics
- Bone Marrow Neoplasms/immunology
- Bone Marrow Neoplasms/pathology
- Female
- Fluorodeoxyglucose F18
- Gene Rearrangement, B-Lymphocyte, Light Chain
- Humans
- Immunoglobulin Heavy Chains
- Lymphoma, Large B-Cell, Diffuse/diagnostic imaging
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Positron Emission Tomography Computed Tomography/methods
- Prognosis
- Prospective Studies
- Radiopharmaceuticals
- Young Adult
Collapse
Affiliation(s)
- Mihee Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Seo-Yeon Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ga-Young Song
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Jun Hyung Lee
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Myung-Geun Shin
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Sang Yun Song
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea.
| |
Collapse
|
18
|
Hasani N, Paravastu SS, Farhadi F, Yousefirizi F, Morris MA, Rahmim A, Roschewski M, Summers RM, Saboury B. Artificial Intelligence in Lymphoma PET Imaging:: A Scoping Review (Current Trends and Future Directions). PET Clin 2022; 17:145-174. [PMID: 34809864 PMCID: PMC8735853 DOI: 10.1016/j.cpet.2021.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Malignant lymphomas are a family of heterogenous disorders caused by clonal proliferation of lymphocytes. 18F-FDG-PET has proven to provide essential information for accurate quantification of disease burden, treatment response evaluation, and prognostication. However, manual delineation of hypermetabolic lesions is often a time-consuming and impractical task. Applications of artificial intelligence (AI) may provide solutions to overcome this challenge. Beyond segmentation and detection of lesions, AI could enhance tumor characterization and heterogeneity quantification, as well as treatment response prediction and recurrence risk stratification. In this scoping review, we have systematically mapped and discussed the current applications of AI (such as detection, classification, segmentation as well as the prediction and prognostication) in lymphoma PET.
Collapse
Affiliation(s)
- Navid Hasani
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA; University of Queensland Faculty of Medicine, Ochsner Clinical School, New Orleans, LA 70121, USA
| | - Sriram S Paravastu
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA
| | - Faraz Farhadi
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA
| | - Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Michael A Morris
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA; Department of Computer Science and Electrical Engineering, University of Maryland-Baltimore Country, Baltimore, MD, USA
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada; Department of Radiology, BC Cancer Research Institute, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| | - Ronald M Summers
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA.
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 1C455, Bethesda, MD 20892, USA; Department of Computer Science and Electrical Engineering, University of Maryland-Baltimore Country, Baltimore, MD, USA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Diagnostic value of baseline 18FDG PET/CT skeletal textural features in follicular lymphoma. Sci Rep 2021; 11:23812. [PMID: 34893676 PMCID: PMC8664828 DOI: 10.1038/s41598-021-03278-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/17/2021] [Indexed: 01/06/2023] Open
Abstract
At present, 18F-fluorodesoxyglucose (18FDG) positron emission tomography (PET)/computed tomography (CT) cannot be used to omit a bone marrow biopsy (BMB) among initial staging procedures in follicular lymphoma (FL). The additional diagnostic value of skeletal textural features on baseline 18FDG-PET/CT in diffuse large B-cell lymphoma (DLBCL) patients has given promising results. The aim of this study is to evaluate the value of 18FDG-PET/CT radiomics for the diagnosis of bone marrow involvement (BMI) in FL patients. This retrospective bicentric study enrolled newly diagnosed FL patients addressed for baseline 18FDG PET/CT. For visual assessment, examinations were considered positive in cases of obvious bone focal uptakes. For textural analysis, the skeleton volumes of interest (VOIs) were automatically extracted from segmented CT images and analysed using LifeX software. BMB and visual assessment were taken as the gold standard: BMB −/PET − patients were considered as bone-NEGATIVE patients, whereas BMB +/PET −, BMB −/PET + and BMB +/PET + patients were considered bone-POSITIVE patients. A LASSO regression algorithm was used to select features of interest and to build a prediction model. Sixty-six consecutive patients were included: 36 bone-NEGATIVE (54.5%) and 30 bone-POSITIVE (45.5%). The LASSO regression found variance_GLCM, correlation_GLCM, joint entropy_GLCM and busyness_NGLDM to have nonzero regression coefficients. Based on ROC analysis, a cut-off equal to − 0.190 was found to be optimal for the diagnosis of BMI using PET pred.score. The corresponding sensitivity, specificity, PPV and NPV values were equal to 70.0%, 83.3%, 77.8% and 76.9%, respectively. When comparing the ROC AUCs with using BMB alone, visual PET assessment or PET pred.score, a significant difference was found between BMB versus visual PET assessments (p = 0.010) but not between BMB and PET pred.score assessments (p = 0.097). Skeleton texture analysis is worth exploring to improve the performance of 18FDG-PET/CT for the diagnosis of BMI at baseline in FL patients.
Collapse
|
20
|
Zhou J, Zou S, Kuang D, Yan J, Zhao J, Zhu X. A Novel Approach Using FDG-PET/CT-Based Radiomics to Assess Tumor Immune Phenotypes in Patients With Non-Small Cell Lung Cancer. Front Oncol 2021; 11:769272. [PMID: 34868999 PMCID: PMC8635743 DOI: 10.3389/fonc.2021.769272] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Tumor microenvironment immune types (TMITs) are closely related to the efficacy of immunotherapy. We aimed to assess the predictive ability of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT)-based radiomics of TMITs in treatment-naive patients with non-small cell lung cancer (NSCLC). METHODS A retrospective analysis was performed in 103 patients with NSCLC who underwent 18F-FDG PET/CT scans. The patients were randomly assigned into a training set (n = 71) and a validation set (n = 32). Tumor specimens were analyzed by immunohistochemistry for the expression of programmed death-ligand 1 (PD-L1), programmed death-1 (PD-1), and CD8+ tumor-infiltrating lymphocytes (TILs) and categorized into four TMITs according to their expression of PD-L1 and CD8+ TILs. LIFEx package was used to extract radiomic features. The optimal features were selected using the least absolute shrinkage and selection operator (LASSO) algorithm, and a radiomics signature score (rad-score) was developed. We constructed a combined model based on the clinical variables and radiomics signature and compared the predictive performance of models using receiver operating characteristic (ROC) curves. RESULTS Four radiomic features (GLRLM_LRHGE, GLZLM_SZE, SUVmax, NGLDM_Contrast) were selected to build the rad-score. The rad-score showed a significant ability to discriminate between TMITs in both sets (p < 0.001, p < 0.019), with an area under the ROC curve (AUC) of 0.800 [95% CI (0.688-0.885)] in the training set and that of 0.794 [95% CI (0.615-0.916)] in the validation set, while the AUC values of clinical variables were 0.738 and 0.699, respectively. When clinical variables and radiomics signature were combined, the complex model showed better performance in predicting TMIT-I tumors, with the AUC values increased to 0.838 [95% CI (0.731-0.914)] in the training set and 0.811 [95% CI (0.634-0.927)] in the validation set. CONCLUSION The FDG-PET/CT-based radiomic features showed good performance in predicting TMIT-I tumors in NSCLC, providing a promising approach for the choice of immunotherapy in a clinical setting.
Collapse
Affiliation(s)
- Jianyuan Zhou
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijuan Zou
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Kuang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Yan
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Zhang X, Chen L, Jiang H, He X, Feng L, Ni M, Ma M, Wang J, Zhang T, Wu S, Zhou R, Jin C, Zhang K, Qian W, Chen Z, Zhuo C, Zhang H, Tian M. A novel analytic approach for outcome prediction in diffuse large B-cell lymphoma by [ 18F]FDG PET/CT. Eur J Nucl Med Mol Imaging 2021; 49:1298-1310. [PMID: 34651227 PMCID: PMC8921097 DOI: 10.1007/s00259-021-05572-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/22/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE This study aimed to develop a novel analytic approach based on 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography ([18F]FDG PET/CT) radiomic signature (RS) and International Prognostic Index (IPI) to predict the progression-free survival (PFS) and overall survival (OS) of patients with diffuse large B-cell lymphoma (DLBCL). METHODS We retrospectively enrolled 152 DLBCL patients and divided them into a training cohort (n = 100) and a validation cohort (n = 52). A total of 1245 radiomic features were extracted from the total metabolic tumor volume (TMTV) and the metabolic bulk volume (MBV) of pre-treatment PET/CT images. The least absolute shrinkage and selection operator (LASSO) algorithm was applied to develop the RS. Cox regression analysis was used to construct hybrid nomograms based on different RS and clinical variables. The performances of hybrid nomograms were evaluated using the time-dependent receiver operator characteristic (ROC) curve and the Hosmer-Lemeshow test. The clinical utilities of prediction nomograms were determined via decision curve analysis. The predictive efficiency of different RS, clinical variables, and hybrid nomograms was compared. RESULTS The RS and IPI were identified as independent predictors of PFS and OS, and were selected to construct hybrid nomograms. Both TMTV- and MBV-based hybrid nomograms had significantly higher values of area under the curve (AUC) than IPI in training and validation cohorts (all P < 0.05), while no significant difference was found between TMTV- and MBV-based hybrid nomograms (P > 0.05). The Hosmer-Lemeshow test showed that both TMTV- and MBV-based hybrid nomograms calibrated well in the training and validation cohorts (all P > 0.05). Decision curve analysis indicated that hybrid nomograms had higher net benefits than IPI. CONCLUSION The hybrid nomograms combining RS with IPI could significantly improve survival prediction in DLBCL. Radiomic analysis on MBV may serve as a potential approach for prognosis assessment in DLBCL. TRIAL REGISTRATION NCT04317313. Registered March 16, 2020. Public site: https://clinicaltrials.gov/ct2/show/NCT04317313.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Lin Chen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xuexin He
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Liu Feng
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Miaoqi Ni
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Mindi Ma
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Teng Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Kai Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zexin Chen
- Department of Clinical Epidemiology & Biostatistics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhuo
- College of Information Science & Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China. .,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
22
|
Mesguich C, Hindie E, de Senneville BD, Tlili G, Pinaquy JB, Marit G, Saut O. Improved 18-FDG PET/CT diagnosis of multiple myeloma diffuse disease by radiomics analysis. Nucl Med Commun 2021; 42:1135-1143. [PMID: 34001823 DOI: 10.1097/mnm.0000000000001437] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES In multiple myeloma, the diagnosis of diffuse bone marrow infiltration on 18-FDG PET/CT can be challenging. We aimed to develop a PET/CT radiomics-based model that could improve the diagnosis of multiple myeloma diffuse disease on 18-FDG PET/CT. METHODS We prospectively performed PET/CT and whole-body diffusion-weighted MRI in 30 newly diagnosed multiple myeloma. MRI was the reference standard for diffuse disease assessment. Twenty patients were randomly assigned to a training set and 10 to an independent test set. Visual analysis of PET/CT was performed by two nuclear medicine physicians. Spine volumes were automatically segmented, and a total of 174 Imaging Biomarker Standardisation Initiative-compliant radiomics features were extracted from PET and CT. Selection of best features was performed with random forest features importance and correlation analysis. Machine-learning algorithms were trained on the selected features with cross-validation and evaluated on the independent test set. RESULTS Out of the 30 patients, 18 had established diffuse disease on MRI. The sensitivity, specificity and accuracy of visual analysis were 67, 75 and 70%, respectively, with a moderate kappa coefficient of agreement of 0.6. Five radiomics features were selected. On the training set, random forest classifier reached a sensitivity, specificity and accuracy of 93, 86 and 91%, respectively, with an area under the curve of 0.90 (95% confidence interval, 0.89-0.91). On the independent test set, the model achieved an accuracy of 80%. CONCLUSIONS Radiomics analysis of 18-FDG PET/CT images with machine-learning overcame the limitations of visual analysis, providing a highly accurate and more reliable diagnosis of diffuse bone marrow infiltration in multiple myeloma patients.
Collapse
Affiliation(s)
- Charles Mesguich
- Nuclear Medicine Department, CHU Bordeaux
- INSERM U1035, University of Bordeaux, Bordeaux
- University of Bordeaux, IMB, UMR CNRS 5251, INRIA Project team Monc, Talence, France
| | | | | | | | | | | | - Olivier Saut
- University of Bordeaux, IMB, UMR CNRS 5251, INRIA Project team Monc, Talence, France
| |
Collapse
|
23
|
|
24
|
A Systematic Review of PET Textural Analysis and Radiomics in Cancer. Diagnostics (Basel) 2021; 11:diagnostics11020380. [PMID: 33672285 PMCID: PMC7926413 DOI: 10.3390/diagnostics11020380] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although many works have supported the utility of PET radiomics, several authors have raised concerns over the robustness and replicability of the results. This study aimed to perform a systematic review on the topic of PET radiomics and the used methodologies. Methods: PubMed was searched up to 15 October 2020. Original research articles based on human data specifying at least one tumor type and PET image were included, excluding those that apply only first-order statistics and those including fewer than 20 patients. Each publication, cancer type, objective and several methodological parameters (number of patients and features, validation approach, among other things) were extracted. Results: A total of 290 studies were included. Lung (28%) and head and neck (24%) were the most studied cancers. The most common objective was prognosis/treatment response (46%), followed by diagnosis/staging (21%), tumor characterization (18%) and technical evaluations (15%). The average number of patients included was 114 (median = 71; range 20–1419), and the average number of high-order features calculated per study was 31 (median = 26, range 1–286). Conclusions: PET radiomics is a promising field, but the number of patients in most publications is insufficient, and very few papers perform in-depth validations. The role of standardization initiatives will be crucial in the upcoming years.
Collapse
|
25
|
Frood R, Burton C, Tsoumpas C, Frangi AF, Gleeson F, Patel C, Scarsbrook A. Baseline PET/CT imaging parameters for prediction of treatment outcome in Hodgkin and diffuse large B cell lymphoma: a systematic review. Eur J Nucl Med Mol Imaging 2021; 48:3198-3220. [PMID: 33604689 PMCID: PMC8426243 DOI: 10.1007/s00259-021-05233-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Purpose To systematically review the literature evaluating clinical utility of imaging metrics derived from baseline fluorine-18 fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT) for prediction of progression-free (PFS) and overall survival (OS) in patients with classical Hodgkin lymphoma (HL) and diffuse large B cell lymphoma (DLBCL). Methods A search of MEDLINE/PubMed, Web of Science, Cochrane, Scopus and clinicaltrials.gov databases was undertaken for articles evaluating PET/CT imaging metrics as outcome predictors in HL and DLBCL. PRISMA guidelines were followed. Risk of bias was assessed using the Quality in Prognosis Studies (QUIPS) tool. Results Forty-one articles were included (31 DLBCL, 10 HL). Significant predictive ability was reported in 5/20 DLBCL studies assessing SUVmax (PFS: HR 0.13–7.35, OS: HR 0.83–11.23), 17/19 assessing metabolic tumour volume (MTV) (PFS: HR 2.09–11.20, OS: HR 2.40–10.32) and 10/13 assessing total lesion glycolysis (TLG) (PFS: HR 1.078–11.21, OS: HR 2.40–4.82). Significant predictive ability was reported in 1/4 HL studies assessing SUVmax (HR not reported), 6/8 assessing MTV (PFS: HR 1.2–10.71, OS: HR 1.00–13.20) and 2/3 assessing TLG (HR not reported). There are 7/41 studies assessing the use of radiomics (4 DLBCL, 2 HL); 5/41 studies had internal validation and 2/41 included external validation. All studies had overall moderate or high risk of bias. Conclusion Most studies are retrospective, underpowered, heterogenous in their methodology and lack external validation of described models. Further work in protocol harmonisation, automated segmentation techniques and optimum performance cut-off is required to develop robust methodologies amenable for clinical utility. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05233-2.
Collapse
Affiliation(s)
- R Frood
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK. .,Leeds Institute of Health Research, University of Leeds, Leeds, UK.
| | - C Burton
- Department of Haematology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - C Tsoumpas
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - A F Frangi
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.,Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), School of Computing and School of Medicine, University of Leeds, Leeds, UK.,Medical Imaging Research Center (MIRC), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - F Gleeson
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - C Patel
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - A Scarsbrook
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Leeds Institute of Health Research, University of Leeds, Leeds, UK
| |
Collapse
|
26
|
Prognostic Value of Baseline Radiomic Features of 18F-FDG PET in Patients with Diffuse Large B-Cell Lymphoma. Diagnostics (Basel) 2020; 11:diagnostics11010036. [PMID: 33379166 PMCID: PMC7824203 DOI: 10.3390/diagnostics11010036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
This study investigates whether baseline 18F-FDG PET radiomic features can predict survival outcomes in patients with diffuse large B-cell lymphoma (DLBCL). We retrospectively enrolled 83 patients diagnosed with DLBCL who underwent 18F-FDG PET scans before treatment. The patients were divided into the training cohort (n = 58) and the validation cohort (n = 25). Eighty radiomic features were extracted from the PET images for each patient. Least absolute shrinkage and selection operator regression were used to reduce the dimensionality within radiomic features. Cox proportional hazards model was used to determine the prognostic factors for progression-free survival (PFS) and overall survival (OS). A prognostic stratification model was built in the training cohort and validated in the validation cohort using Kaplan-Meier survival analysis. In the training cohort, run length non-uniformity (RLN), extracted from a gray level run length matrix (GLRLM), was independently associated with PFS (hazard ratio (HR) = 15.7, p = 0.007) and OS (HR = 8.64, p = 0.040). The International Prognostic Index was an independent prognostic factor for OS (HR = 2.63, p = 0.049). A prognostic stratification model was devised based on both risk factors, which allowed identification of three risk groups for PFS and OS in the training (p < 0.001 and p < 0.001) and validation (p < 0.001 and p = 0.020) cohorts. Our results indicate that the baseline 18F-FDG PET radiomic feature, RLNGLRLM, is an independent prognostic factor for survival outcomes. Furthermore, we propose a prognostic stratification model that may enable tailored therapeutic strategies for patients with DLBCL.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Imaging features of lymphoma vary regionally. Awareness of site-specific key imaging characteristics of lymphoma can aid in rapid staging and assist in prompt treatment. FDG PET/CT and conventional MRI are readily available diagnostic modalities with excellent sensitivity and good specificity. Diagnostic specificity can be enhanced using emerging PET radiotracers, e.g., FLT and FET. RECENT FINDINGS Emerging research has shown higher dimensional analysis (radiomics and radiogenomics) of imaging data can yield information of the underlying genetic aberrations in lymphoma, which can aid in assessing real-time evolution of tumor. CT, PET/CT, MRI, and ultrasound accentuate the intrinsic qualities of lymphoma (e.g., FDG PET/CT for increased metabolic activity, FLT PET/CT for increased proliferation index, and DWI for increased cellularity) and play an essential role in its diagnosis and examination. Advanced radiogenomic analyses use radiomic parameters to deduce genetic variations of lymphoma, providing noninvasive, repeatable, and real-time surveillance of its genetic progression.
Collapse
|
28
|
Mayerhoefer ME, Umutlu L, Schöder H. Functional imaging using radiomic features in assessment of lymphoma. Methods 2020; 188:105-111. [PMID: 32634555 DOI: 10.1016/j.ymeth.2020.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
Lymphomas are typically large, well-defined, and relatively homogeneous tumors, and therefore represent ideal targets for the use of radiomics. Of the available functional imaging tests, [18F]FDG-PET for body lymphoma and diffusion-weighted MRI (DWI) for central nervous system (CNS) lymphoma are of particular interest. The current literature suggests that two main applications for radiomics in lymphoma show promise: differentiation of lymphomas from other tumors, and lymphoma treatment response and outcome prognostication. In particular, encouraging results reported in the limited number of presently available studies that utilize functional imaging suggest that (1) MRI-based radiomics enables differentiation of CNS lymphoma from glioblastoma, and (2) baseline [18F]FDG-PET radiomics could be useful for survival prognostication, adding to or even replacing commonly used metrics such as standardized uptake values and metabolic tumor volume. However, due to differences in biological and clinical characteristics of different lymphoma subtypes and an increasing number of treatment options, more data are required to support these findings. Furthermore, a consensus on several critical steps in the radiomics workflow -most importantly, image reconstruction and post processing, lesion segmentation, and choice of classification algorithm- is desirable to ensure comparability of results between research institutions.
Collapse
Affiliation(s)
- Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, NY, USA; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria.
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Germany
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, NY, USA
| |
Collapse
|
29
|
Current status and quality of radiomics studies in lymphoma: a systematic review. Eur Radiol 2020; 30:6228-6240. [PMID: 32472274 DOI: 10.1007/s00330-020-06927-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/25/2020] [Accepted: 04/28/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To perform a systematic review regarding the developments in the field of radiomics in lymphoma. To evaluate the quality of included articles by the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2), the phases classification criteria for image mining studies, and the radiomics quality scoring (RQS) tool. METHODS We searched for eligible articles in the MEDLINE/PubMed and EMBASE databases using the terms "radiomics", "texture" and "lymphoma". The included studies were divided into two categories: diagnosis-, therapy response- and outcome-related studies. The diagnosis-related studies were evaluated using the QUADAS-2; all studies were evaluated using the phases classification criteria for image mining studies and the RQS tool by two reviewers. RESULTS Forty-five studies were included; thirteen papers (28.9%) focused on the differential diagnosis of primary central nervous system lymphoma (PCNSL) and glioblastoma (GBM). Thirty-two (71.1%) studies were classified as discovery science according to the phase classification criteria for image mining studies. The mean RQS score of all studies was 14.2% (ranging from 0.0 to 40.3%), and 23 studies (51.1%) were given a score of < 10%. CONCLUSION The radiomics features could serve as diagnostic and prognostic indicators in lymphoma. However, the current conclusions should be interpreted with caution due to the suboptimal quality of the studies. In order to introduce radiomics into lymphoma clinical settings, the lesion segmentation and selection, the influence of the pathological pattern and the extraction of multiple modalities and multiple time points features need to be further studied. KEY POINTS • The radiomics approach may provide useful information for diagnosis, prediction of the therapy response, and outcome of lymphoma. • The quality of published radiomics studies in lymphoma has been suboptimal to date. • More studies are needed to examine lesion selection and segmentation, the influence of pathological patterns, and the extraction of multiple modalities and multiple time point features.
Collapse
|
30
|
Development and validation of an 18F-FDG PET radiomic model for prognosis prediction in patients with nasal-type extranodal natural killer/T cell lymphoma. Eur Radiol 2020; 30:5578-5587. [PMID: 32435928 DOI: 10.1007/s00330-020-06943-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 05/07/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To identify an 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) radiomics-based model for predicting progression-free survival (PFS) and overall survival (OS) of nasal-type extranodal natural killer/T cell lymphoma (ENKTL). METHODS In this retrospective study, a total of 110 ENKTL patients were divided into a training cohort (n = 82) and a validation cohort (n = 28). Forty-one features were extracted from pretreatment PET images of the patients. Least absolute shrinkage and selection operator (LASSO) regression was used to develop the radiomic signatures (R-signatures). A radiomics-based model was built and validated in the two cohorts and compared with a metabolism-based model. RESULTS The R-signatures were constructed with moderate predictive ability in the training and validation cohorts (R-signaturePFS: AUC = 0.788 and 0.473; R-signatureOS: AUC = 0.637 and 0.730). For PFS, the radiomics-based model showed better discrimination than the metabolism-based model in the training cohort (C-index = 0.811 vs. 0.751) but poorer discrimination in the validation cohort (C-index = 0.588 vs. 0.693). The calibration of the radiomics-based model was poorer than that of the metabolism-based model (training cohort: p = 0.415 vs. 0.428, validation cohort: p = 0.228 vs. 0.652). For OS, the performance of the radiomics-based model was poorer (training cohort: C-index = 0.818 vs. 0.828, p = 0.853 vs. 0.885; validation cohort: C-index = 0.628 vs. 0.753, p < 0.05 vs. 0.913). CONCLUSIONS Radiomic features derived from PET images can predict the outcomes of patients with ENKTL, but the performance of the radiomics-based model was inferior to that of the metabolism-based model. KEY POINTS • The R-signatures calculated by using 18F-FDG PET radiomic features can predict the survival of patients with ENKTL. • The radiomics-based models integrating the R-signatures and clinical factors achieved good predictive values. • The performance of the radiomics-based model was inferior to that of the metabolism-based model in the two cohorts.
Collapse
|
31
|
Mayerhoefer ME, Riedl CC, Kumar A, Dogan A, Gibbs P, Weber M, Staber PB, Huicochea Castellanos S, Schöder H. [18F]FDG-PET/CT Radiomics for Prediction of Bone Marrow Involvement in Mantle Cell Lymphoma: A Retrospective Study in 97 Patients. Cancers (Basel) 2020; 12:cancers12051138. [PMID: 32370121 PMCID: PMC7281173 DOI: 10.3390/cancers12051138] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
Biopsy is the standard for assessment of bone marrow involvement in mantle cell lymphoma (MCL). We investigated whether [18F]FDG-PET radiomic texture features can improve prediction of bone marrow involvement in MCL, compared to standardized uptake values (SUV), and whether combination with laboratory data improves results. Ninety-seven MCL patients were retrospectively included. SUVmax, SUVmean, SUVpeak and 16 co-occurrence matrix texture features were extracted from pelvic bones on [18F]FDG-PET/CT. A multi-layer perceptron neural network was used to compare three combinations for prediction of bone marrow involvement—the SUVs, a radiomic signature based on SUVs and texture features, and the radiomic signature combined with laboratory parameters. This step was repeated using two cut-off values for relative bone marrow involvement: REL > 5% (>5% of red/cellular bone marrow); and REL > 10%. Biopsy demonstrated bone marrow involvement in 67/97 patients (69.1%). SUVs, the radiomic signature, and the radiomic signature with laboratory data showed AUCs of up to 0.66, 0.73, and 0.81 for involved vs. uninvolved bone marrow; 0.68, 0.84, and 0.84 for REL ≤ 5% vs. REL > 5%; and 0.69, 0.85, and 0.87 for REL ≤ 10% vs. REL > 10%. In conclusion, [18F]FDG-PET texture features improve SUV-based prediction of bone marrow involvement in MCL. The results may be further improved by combination with laboratory parameters.
Collapse
Affiliation(s)
- Marius E. Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.R.); (P.G.); (S.H.C.); (H.S.)
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +1-646-961-5030
| | - Christopher C. Riedl
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.R.); (P.G.); (S.H.C.); (H.S.)
| | - Anita Kumar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.R.); (P.G.); (S.H.C.); (H.S.)
| | - Michael Weber
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria;
| | - Philipp B. Staber
- Department of Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Sandra Huicochea Castellanos
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.R.); (P.G.); (S.H.C.); (H.S.)
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.R.); (P.G.); (S.H.C.); (H.S.)
| |
Collapse
|
32
|
Gao J, Huang X, Meng H, Zhang M, Zhang X, Lin X, Li B. Performance of Multiparametric Functional Imaging and Texture Analysis in Predicting Synchronous Metastatic Disease in Pancreatic Ductal Adenocarcinoma Patients by Hybrid PET/MR: Initial Experience. Front Oncol 2020; 10:198. [PMID: 32158690 PMCID: PMC7052324 DOI: 10.3389/fonc.2020.00198] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives: To assess the imaging biomarkers of glucose metabolic activity and diffusion-weighted imaging (DWI) derived from pretreatment integrated 18F-fluorodeoxyglucose positron emission tomography-magnetic resonance (18F-FDG PET/MR) imaging as potential predictive factors of metastasis in patients with pancreatic ductal adenocarcinoma (PDAC). Patients and Methods: We retrospectively included 17 consecutive patients with pathologically confirmed PDAC by pretreatment 18F-FDG PET/MR. The study subjects were divided into a non-metastatic group (M0, six cases) and a metastatic group (M1, 11 cases). The 18F-FDG PET/MR images were reviewed independently by two board certificated nuclear medicine physicians and one radiologist. Conventional characteristics and quantitative parameters from both PET and apparent diffusion coefficient (ADC) were assessed. The texture features were extracted from LIFEx packages (www.lifexsoft.org), and a 3D tumor volume of interest was manually drawn on fused PET/ADC images. Chi-square tests, independent-samples t-tests and Mann-Whitney U-tests were used to compare the differences in single parameters between the two groups. A logistic regression analysis was performed to determine independent predictors. A receiver operating characteristic (ROC) curve analysis was performed to assess the discriminatory power of the selected parameters. Correlations between metabolic parameters and ADC features were calculated with Spearman's rank correlation coefficient test. Results: For conventional parameters, univariable analysis demonstrated that the M1 group had a significantly larger size and a higher peak of standardized uptake value (SUVpeak), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) than those of the M0 group (p < 0.05 for all). TLG remained significant predictor in the multivariable analysis, but there were no significant differences for the area under the ROC curve (AUC) among the four conventional features in differential diagnoses (p > 0.05 for all). For the texture features, there were four features from the PET image and 13 from the ADC map that showed significant differences between the two groups. Multivariate analysis indicated that one feature from PET and three from the ADC were significant predictors. TLG was associated with ADC-GLRLM_GLNU (r = 0.659), ADC-GLRLM_LRHGE (r = 0.762), and PET-GLRLM_LRHGE (r = 0.806). Conclusions: Multiple parameters and texture features of primary tumors from 18F-FDG PET/MR images maybe reliable biomarkers to predict synchronous metastatic disease for the pretreatment PDAC.
Collapse
Affiliation(s)
- Jing Gao
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyun Huang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Meng
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhe Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhu Lin
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Biao Li
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Li H, Xu C, Xin B, Zheng C, Zhao Y, Hao K, Wang Q, Wahl RL, Wang X, Zhou Y. 18F-FDG PET/CT Radiomic Analysis with Machine Learning for Identifying Bone Marrow Involvement in the Patients with Suspected Relapsed Acute Leukemia. Am J Cancer Res 2019; 9:4730-4739. [PMID: 31367253 PMCID: PMC6643435 DOI: 10.7150/thno.33841] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
18F-FDG PET / CT is used clinically for the detection of extramedullary lesions in patients with relapsed acute leukemia (AL). However, the visual analysis of 18F-FDG diffuse bone marrow uptake in detecting bone marrow involvement (BMI) in routine clinical practice is still challenging. This study aims to improve the diagnostic performance of 18F-FDG PET/CT in detecting BMI for patients with suspected relapsed AL. Methods: Forty-one patients (35 in training group and 6 in independent validation group) with suspected relapsed AL were retrospectively included in this study. All patients underwent both bone marrow biopsy (BMB) and 18F-FDG PET/CT within one week. The BMB results were used as the gold standard or real “truth” for BMI. The bone marrow 18F-FDG uptake was visually diagnosed as positive and negative by three nuclear medicine physicians. The skeletal volumes of interest were manually drawn on PET/CT images. A total of 781 PET and 1045 CT radiomic features were automatically extracted to provide a more comprehensive understanding of the embedded pattern. To select the most important and predictive features, an unsupervised consensus clustering method was first performed to analyze the feature correlations and then used to guide a random forest supervised machine learning model for feature importance analysis. Cross-validation and independent validation were conducted to justify the performance of our model. Results: The training group involved 16 BMB positive and 19 BMB negative patients. Based on the visual analysis of 18F-FDG PET, 3 patients had focal uptake, 8 patients had normal uptake, and 24 patients had diffuse uptake. The sensitivity, specificity, and accuracy of visual analysis for BMI diagnosis were 62.5%, 73.7%, and 68.6%, respectively. With the cross-validation on the training group, the machine learning model correctly predicted 31 patients in BMI. The sensitivity, specificity, and accuracy of the machine learning model in BMI detection were 87.5%, 89.5%, and 88.6%, respectively, significantly higher than the ones in visual analysis (P < 0.05). The evaluation on the independent validation group showed that the machine learning model could achieve 83.3% accuracy. Conclusions:18F-FDG PET/CT radiomic analysis with machine learning model provided a quantitative, objective and efficient mechanism for identifying BMI in the patients with suspected relapsed AL. It is suggested in particular for the diagnosis of BMI in the patients with 18F-FDG diffuse uptake patterns.
Collapse
|
34
|
Heterogeneity analysis of 18F-FDG PET imaging in oncology: clinical indications and perspectives. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0299-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Aide N, Lasnon C, Damaj G. Combining baseline TMTV and gene profiling for a better risk stratification in diffuse large B cell lymphoma. Eur J Nucl Med Mol Imaging 2018; 45:677-679. [PMID: 29455312 DOI: 10.1007/s00259-018-3966-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Nicolas Aide
- Nuclear Medicine Department, Caen University Hospital, Caen, France. .,Normandie University, Caen, France. .,INSERM 1086 ANTICIPE, Normandie University, Caen, France.
| | - Charline Lasnon
- INSERM 1086 ANTICIPE, Normandie University, Caen, France.,Nuclear Medicine Department, François Baclesse Cancer Centre, Caen, France
| | - Gandhi Damaj
- Normandie University, Caen, France.,Haematology Institute, Caen University Hospital, Caen, France.,MICAH INSERM U1245, Rouen University, Rouen, France
| |
Collapse
|