1
|
Novruzov E, Peters HA, Jannusch K, Kobbe G, Dietrich S, Fischer JC, Rox J, Antoch G, Giesel FL, Antke C, Baermann BN, Mamlins E. The predictive power of baseline metabolic and volumetric [ 18F]FDG PET parameters with different thresholds for early therapy failure and mortality risk in DLBCL patients undergoing CAR-T-cell therapy. Eur J Radiol Open 2025; 14:100619. [PMID: 39803388 PMCID: PMC11719856 DOI: 10.1016/j.ejro.2024.100619] [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: 10/11/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025] Open
Abstract
Objective [18F]FDG imaging is an integral part of patient management in CAR-T-cell therapy for recurrent or therapy-refractory DLBCL. The calculation methods of predictive power of specific imaging parameters still remains elusive. With this retrospective study, we sought to evaluate the predictive power of the baseline metabolic parameters and tumor burden calculated with automated segmentation via different thresholding methods for early therapy failure and mortality risk in DLBCL patients. Materials and methods Eighteen adult patients were enrolled, who underwent CAR-T-cell therapy accompanied by at least one pretherapeutic and two posttherapeutic [18F]FDG PET scans within 30 and 90 days between December 2018 and October 2023. We performed single-click automatic segmentation within VOIs in addition to extracting the SUV parameters to calculate the MTVs and TLGs by applying thresholds based on the concepts of a fixed absolute threshold with an SUVmax > 4.0, a relative absolute threshold with an isocontour of > 40 % of the SUVmax, a background threshold involving the addition of the liver SUV value and its 2 SD values, and only the liver SUV value. Results For early therapy failure, baseline metabolic parameters such as the SUVmax, SUVpeak and SUVmean tended to have greater predictive power than did the baseline metabolic burden. However, the baseline metabolic burden was superior in the prediction of mortality risk regardless of the thresholding method used. Conclusion This study revealed that automated delineation methods of metabolic tumor burden using different thresholds do not differ in outcome substantially. Therefore, the current clinical standard with a fixed absolute threshold value of SUV > 4.0 seems to be a feasible option.
Collapse
Affiliation(s)
- Emil Novruzov
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Helena A. Peters
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Kai Jannusch
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Guido Kobbe
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Sascha Dietrich
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Johannes C. Fischer
- Institute for Transplantation Diagnostics and Cellular Therapy, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Jutta Rox
- Institute for Transplantation Diagnostics and Cellular Therapy, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Frederik L. Giesel
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Institute for Radiation Sciences, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Ben-Niklas Baermann
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Eduards Mamlins
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| |
Collapse
|
2
|
Boellaard R, Zwezerijnen GJC, Buvat I, Champion L, Hovhannisyan-Baghdasarian N, Orlhac F, Arens AIJ, Lobeek D, Celik F, Mitea C, Huijbregts JE, Tolboom N, de Keizer B, Valkema R, van Velden FHP, Dibbets-Schneider P, Wiegers SE, Lugtenburg PJ, Barrington SF, Zijlstra JM. Measuring Total Metabolic Tumor Volume from 18F-FDG PET: A Reality Check. J Nucl Med 2025; 66:802-805. [PMID: 40081961 DOI: 10.2967/jnumed.124.269271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
Measuring total metabolic tumor volume (TMTV) on 18F-FDG PET/CT images in clinical practice requires a fast, reliable, and easy-to-perform multilesional segmentation workflow. We conducted a field test to derive total metabolic volumes using 5 representative baseline 18F-FDG PET/CT scans from patients with diffuse large B-cell lymphoma. The scans were transferred to 10 different sites or readers who used different commercially available software platforms to derive TMTV after a recently proposed benchmark workflow. Observed TMTVs were compared with reference values, and overall analysis times were reported. Our results show that TMTVs can be obtained with reasonable accuracy across readers and platforms (within 10% compared with reference benchmark values for most TMTVs) but that processing times can vary considerably depending on reader experience and the software platform. Our study showed that there is an urgent need to improve TMTV segmentation workflows in clinical practice, requiring closer collaboration between users and software vendors.
Collapse
Affiliation(s)
- Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands;
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Laurence Champion
- LITO, Inserm, Institut Curie, Orsay, France
- Department of Nuclear Medicine, Institut Curie, Saint-Cloud, France
| | | | | | - Anne I J Arens
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daphne Lobeek
- Department of Radiology and Nuclear Medicine, Catharina Hospital, Eindhoven, The Netherlands
| | - Filiz Celik
- Center for Radiology and Nuclear Medicine, Department of Nuclear Medicine, Deventer Hospital, Deventer, The Netherlands
| | - Cristina Mitea
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre, and GROW - Research Institute for Oncology and Reproduction, Maastricht, The Netherlands
| | - Julia E Huijbregts
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Nelleke Tolboom
- Department of Nuclear Medicine and Radiology, UMC Utrecht, Utrecht, The Netherlands
| | - Bart de Keizer
- Department of Nuclear Medicine and Radiology, UMC Utrecht, Utrecht, The Netherlands
| | - Roelf Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Petra Dibbets-Schneider
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sanne E Wiegers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, and HOVON Imaging working group, Amsterdam, The Netherlands
| | | | - Sally F Barrington
- King's College London and Guy's and St. Thomas's PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Josée M Zijlstra
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, and HOVON Imaging working group, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Zhang G, Shen J, Hu T, Zheng W, Jia Q, Tan J, Meng Z. Utility of 18F-FDG PET/CT metabolic parameters on post-transplant lymphoproliferative disorder diagnosis. Ann Nucl Med 2025; 39:441-449. [PMID: 39826002 DOI: 10.1007/s12149-025-02016-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVE Using 18F-FDG PET/CT metabolic parameters to differentiate post-transplant lymphoproliferative disorder (PTLD) and reactive lymphoid hyperplasia (RLH), and PTLD subtypes. METHODS 18F-FDG PET/CT and clinical data from 63 PTLD cases and 19 RLH cases were retrospectively collected. According to the 2017 WHO classification, PTLD was categorized into four subtypes: nondestructive (ND-PTLD), polymorphic (P-PTLD), monomorphic (M-PTLD), and classic Hodgkin. Metabolic parameters included maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume (MTV), total lesion glycolysis (TLG) and at different thresholds of SUVmax (2.5 and 41%), as well as gross tumor volume (GTV) was also collected. Nonparametric test and receiver operating characteristic (ROC) curves were used for statistics. RESULTS There were 42 ND-PTLD patients, 7 P-PTLD patients, and 14 M-PTLD patients. Ki-67 was significantly correlated with all metabolic parameters (P all < 0.01). SUVmean, SUVmax, MTV, TLG and GTV were all highest in M-PTLD, followed by P-PTLD, ND-PTLD, and RLH. ROC curves showed 18F-FDG PET/CT metabolic parameters all had moderate diagnostic efficacy in differentiating between PTLD and RLH, the area under the curves (AUC) range from 0.682 to 0.747. Diagnostic efficacy for P-PTLD + M-PTLD showed excellent performance (AUC for RLH + ND-PTLD vs P-PTLD + M-PTLD was 0.848 for SUVmax, 0.846 for SUVmean41%, 0.834 for SUVmean2.5, and 0.819 for GTV). For MTV41%, TLG 41%, MTV2.5, TLG2.5, the AUC was 0.676, 0.761, 0.761, 0.787, respectively. CONCLUSION 18F-FDG PET/CT metabolic parameters at different thresholds of SUVmax (2.5 and 41%) exhibited comparable diagnostic efficacy for PTLD and its subtypes. All metabolic parameters demonstrated moderate diagnostic efficacy in distinguishing PTLD and RLH. SUVmax, SUVmean41%, SUVmean2.5 and GTV showed excellent performance in diagnosing P-PTLD + M-PTLD.
Collapse
Affiliation(s)
- Guoying Zhang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Ultrasound, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Jie Shen
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Tianpeng Hu
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Wei Zheng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiang Jia
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Key Lab of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
4
|
Ceriani L, Milan L, Pirosa MC, Martelli M, Ruberto T, Cascione L, Johnson PWM, Davies AJ, Ciccone G, Zucca E. PET-Based Risk Stratification in Primary Mediastinal B-Cell Lymphoma: A Comparative Analysis of Different Segmentation Methods in the IELSG37 Trial Patient Cohort. J Nucl Med 2025; 66:209-214. [PMID: 39819690 DOI: 10.2967/jnumed.124.268874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
Standardizing tumor measurement on 18F-FDG PET is crucial for the routine clinical use of powerful PET-derived lymphoma prognostic factors such as metabolic tumor volume (MTV) and total lesion glycolysis (TLG). The recent proposal of an SUV of 4 as a new reference segmentation threshold for most aggressive lymphomas may homogenize volume-based metrics and facilitate their clinical application. Methods: This study compared MTV and TLG in primary mediastinal B-cell lymphoma (PMBCL) patients estimated using an SUV of 4 and the current threshold at 25% of SUVmax Baseline PET metrics were evaluated in 501 PMBCL patients from the IELSG37 trial. Results: Median MTV and TLG estimated with the 25% of SUVmax threshold were significantly lower than those obtained with the new reference threshold; however, an extremely high correlation was observed between the methods for both MTV (r = 0.95) and TLG (r = 0.99), resulting in superimposable prognostic power. Conclusion: These findings support the routine use of an SUV of 4 for volumetric measurements in PMBCL.
Collapse
Affiliation(s)
- Luca Ceriani
- 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
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Maria Cristina Pirosa
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Maurizio Martelli
- Hematology Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Teresa Ruberto
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Peter W M Johnson
- Cancer Research United Kingdom Centre, University of Southampton, Southampton, United Kingdom; and
| | - Andrew J Davies
- Cancer Research United Kingdom Centre, University of Southampton, Southampton, United Kingdom; and
| | - Giovannino Ciccone
- Clinical Epidemiology Unit, AOU Città della Salute e della Scienza di Torino and CPO Piemonte, Turin, Italy
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| |
Collapse
|
5
|
Zhou Y, Zhou XY, Xu YC, Ma XL, Tian R. Radiomics based on 18F-FDG PET for predicting treatment response and prognosis in newly diagnosed diffuse large B-cell lymphoma patients: do lesion selection and segmentation methods matter? Quant Imaging Med Surg 2025; 15:103-120. [PMID: 39839002 PMCID: PMC11744140 DOI: 10.21037/qims-24-585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/05/2024] [Indexed: 01/23/2025]
Abstract
Background Radiomics features extracted from baseline 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) scans have shown promising results in predicting the treatment response and outcome of diffuse large B-cell lymphoma (DLBCL) patients. This study aimed to assess the influence of lesion selection approaches and segmentation methods on the radiomics of DLBCL in terms of treatment response and prognosis prediction. Methods A total of 522 and 382 patients pathologically diagnosed with DLBCL were enrolled for complete regression and 2-year event-free survival prediction, respectively. Three lesion selection methods (largest or hottest lesion, patient level) and five segmentation methods (manual and four semiautomatic segmentations) were applied. A total of 112 radiomics features were extracted from the lesions and at the patient level. The feature selection was performed via random forest, and models were constructed via eXtreme Gradient Boosting. The performance of all the models was evaluated via the area under the curve (AUC), which was compared via the Delong test. Results The AUC values varied from 0.583 to 0.768 for the treatment response and prognosis prediction models on the basis of different lesion selection and segmentation methods. However, the prediction performance gap was not significant for each model (all P>0.05). The combined models (AUC =0.908 and 0.837 for treatment response and prognosis prediction, respectively) that incorporated radiomics and clinical features exhibited significant predictive superiority over the clinical models (AUC =0.622 and 0.636, respectively) and the international prognostic index model (AUC =0.623 for prognosis prediction) (all P<0.05). Conclusions Although there are differences in the selected radiomics features among lesion selection and segmentation methods, there is no substantial difference in the predictive power of each radiomics model. In addition, radiomics features have potential added value to clinical features.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xue-Yan Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yu-Chao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang, China
| | - Xue-Lei Ma
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Doma A, Studen A, Jezeršek Novaković B. The Impact of Bone Marrow Involvement on Prognosis in Diffuse Large B-Cell Lymphoma: An 18F-FDG PET/CT Volumetric Segmentation Study. Cancers (Basel) 2024; 16:3762. [PMID: 39594717 PMCID: PMC11592337 DOI: 10.3390/cancers16223762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND This study assessed the prognostic value of tumor burden in bone marrow (BM) and total disease (TD), as depicted on 18F-FDG PET/CT in 140 DLBCL patients, for complete remission after first-line systemic treatment (iCR) and 3- and 5-year overall survival (OS3 and OS5). METHODS Baseline 18F-FDG PET/CT scans of 140 DLBCL patients were segmented to quantify metabolic tumor volume (MTV), total lesion glycolysis (TLG), and SUVmax in BMI, findings elsewhere (XL), and TD. RESULTS Bone marrow involvement (BMI) presented in 35 (25%) patients. Median follow-up time was 47 months; 79 patients (56%) achieved iCR. iCR was significantly associated with TD MTV, XL MTV, BM PET positivity, and International Prognostic Index (IPI). OS3 was significantly worse with TD MTV, XL MTV, IPI, and age. OS5 was significantly associated with IPI, but not with MTVs and TLGs. Univariate factors predicting OS3 were XL MTV (hazard ratio [HR] = 1.29), BMI SUVmax (HR = 0.56), and IPI (HR = 1.92). By multivariate analysis, higher IPI (HR = 2.26) and BMI SUVmax (HR = 0.91) were significant independent predictors for OS3. BMI SUVmax resulted in a negative coefficient and hence indicated a protective effect. CONCLUSIONS Baseline 18F-FDG PET/CT MTV is significantly associated with survival. BMI identified on 18F-FDG PET/CT allows appropriate treatment that may improve survival.
Collapse
Affiliation(s)
- Andrej Doma
- Department of Nuclear Medicine, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Studen
- Experimental Particle Physics Department, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Mathematics and Physics, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Barbara Jezeršek Novaković
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Division of Medical Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Czibor S, Csatlós Z, Fábián K, Piroska M, Györke T. Volumetric and textural analysis of PET/CT in patients with diffuse large B-cell lymphoma highlights the importance of novel MTVrate feature. Nucl Med Commun 2024; 45:931-937. [PMID: 39102514 PMCID: PMC11460743 DOI: 10.1097/mnm.0000000000001884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVES To investigate the prognostic value of clinical, volumetric, and radiomics-based textural parameters in baseline [ 18 F]FDG-PET/CT scans of diffuse large B-cell lymphoma (DLBCL) patients. METHODS We retrospectively investigated baseline PET/CT scans and collected clinical data of fifty DLBCL patients. PET images were segmented semiautomatically to determine metabolic tumor volume (MTV), then the largest segmented lymphoma volume of interest (VOI) was used to extract first-, second-, and high-order textural features. A novel value, MTVrate was introduced as the quotient of the largest lesion's volume and total body MTV. Receiver operating characteristics (ROC) analyses were performed and 24-months progression-free survival (PFS) of low- and high-risk cohorts were compared by log-rank analyses. A machine learning algorithm was used to build a prognostic model from the available clinical, volumetric, and textural data based on logistic regression. RESULTS The area-under-the-curve (AUC) on ROC analysis was the highest of MTVrate at 0.74, followed by lactate-dehydrogenase, MTV, and skewness, with AUCs of 0.68, 0.63, and 0.55, respectively which parameters were also able to differentiate the PFS. A combined survival analysis including MTV and MTVrate identified a subgroup with particularly low PFS at 38%. In the machine learning-based model had an AUC of 0.83 and the highest relative importance was attributed to three textural features and both MTV and MTVrate as important predictors of PFS. CONCLUSION Individual evaluation of different biomarkers yielded only limited prognostic data, whereas a machine learning-based combined analysis had higher effectivity. MTVrate had the highest prognostic ability on individual analysis and, combined with MTV, it identified a patient group with particularly poor prognosis.
Collapse
Affiliation(s)
- Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| | | | - Krisztián Fábián
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
- Mediso Medical Imaging Systems, Budapest, Hungary
| | - Márton Piroska
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University
| |
Collapse
|
8
|
Ferrández MC, Golla SSV, Eertink JJ, Wiegers SE, Zwezerijnen GJC, Heymans MW, Lugtenburg PJ, Kurch L, Hüttmann A, Hanoun C, Dührsen U, Barrington SF, Mikhaeel NG, Ceriani L, Zucca E, Czibor S, Györke T, Chamuleau MED, Zijlstra JM, Boellaard R. Validation of an Artificial Intelligence-Based Prediction Model Using 5 External PET/CT Datasets of Diffuse Large B-Cell Lymphoma. J Nucl Med 2024; 65:1802-1807. [PMID: 39362767 DOI: 10.2967/jnumed.124.268191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
The aim of this study was to validate a previously developed deep learning model in 5 independent clinical trials. The predictive performance of this model was compared with the international prognostic index (IPI) and 2 models incorporating radiomic PET/CT features (clinical PET and PET models). Methods: In total, 1,132 diffuse large B-cell lymphoma patients were included: 296 for training and 836 for external validation. The primary outcome was 2-y time to progression. The deep learning model was trained on maximum-intensity projections from PET/CT scans. The clinical PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, SUVpeak, age, and performance status. The PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, and SUVpeak Model performance was assessed using the area under the curve (AUC) and Kaplan-Meier curves. Results: The IPI yielded an AUC of 0.60 on all external data. The deep learning model yielded a significantly higher AUC of 0.66 (P < 0.01). For each individual clinical trial, the model was consistently better than IPI. Radiomic model AUCs remained higher for all clinical trials. The deep learning and clinical PET models showed equivalent performance (AUC, 0.69; P > 0.05). The PET model yielded the highest AUC of all models (AUC, 0.71; P < 0.05). Conclusion: The deep learning model predicted outcome in all trials with a higher performance than IPI and better survival curve separation. This model can predict treatment outcome in diffuse large B-cell lymphoma without tumor delineation but at the cost of a lower prognostic performance than with radiomics.
Collapse
Affiliation(s)
- Maria C Ferrández
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sandeep S V Golla
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn W Heymans
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lars Kurch
- Clinic and Polyclinic for Nuclear Medicine, Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sally F Barrington
- School of Biomedical Engineering and Imaging Sciences, King's College London and Guy's and St Thomas' PET Centre, King's Health Partners, King's College London, London, United Kingdom
| | - N George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Luca Ceriani
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Emanuele Zucca
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, Oncology Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; and
| | - Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Martine E D Chamuleau
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
9
|
Ladbury C, Hao C, Watkins WT, Sampath S, Wong J, Amini A, Sokolov K, Yeh J, Al Feghali KA, de Jong D, Maniyedath A, Shirvani S, Nikolaenko L, Mei M, Herrera A, Popplewell L, Budde LE, Dandapani S. Prognostic significance of fludeoxyglucose positron emission tomography delta radiomics following bridging therapy in patients with large B-cell lymphoma undergoing CAR T-cell therapy. Front Immunol 2024; 15:1419788. [PMID: 39411711 PMCID: PMC11473334 DOI: 10.3389/fimmu.2024.1419788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose/objectives Bridging radiation therapy (bRT) is increasingly being utilized prior to chimeric antigen receptor (CAR) T-cell therapy for large B-cell lymphoma (LBCL). It is unknown how the extent of cytoreduction during bRT impacts outcomes. Materials/methods We retrospectively reviewed patients with LBCL treated with bRT followed by CAR T-cell therapy. Metabolic tumor volume (MTV), maximum standardized uptake value (SUVmax), SUVmean, and total lesion glycolysis (TLG) were extracted from F18-fluorodeoxyglucose positron emission tomography (PET) scans acquired prior to bRT and between completion of bRT and CAR T-cell infusion. Delta radiomics based on changes of these values were then calculated. The association between delta radiomics and oncologic outcomes [progression-free survival (PFS), freedom from distant progression (FFDP), and local control (LC)] were then examined. Results Thirty-three sites across 23 patients with LBCL were irradiated. All metabolically active disease was treated in 10 patients. Following bRT, median overall decreases (including unirradiated sites) in MTV, SUVmax, SUVmean, and TLG were 22.2 cc (63.1%), 8.9 (36.8%), 3.4 (31.1%), and 297.9 cc (75.8%), respectively. Median decreases in MTV, SUVmax, SUVmean, and TLG in irradiated sites were 15.6 cc (91.1%), 17.0 (74.6%), 6.8 (55.3%), and 157.0 cc (94.6%), respectively. Median follow-up was 15.2 months. A decrease in SUVmax of at least 54% was associated with improved PFS (24-month PFS: 83.3% vs. 28.1%; p = 0.037) and FFDP (24-month FFDP: 100% vs. 62.4%; p < 0.001). A decrease in MTV of at least 90% was associated with improved FFDP (24-month FFDP: 100% vs. 62.4%; p < 0.001). LC was improved in sites with decreases in SUVmax of at least 71% (24-month LC: 100% vs. 72.7%; p < 0.001). Decreases of MTV by at least 90% (100% vs. 53.3%; p = 0.038) and TLG by at least 95% (100% vs. 56.3%; p = 0.067) were associated with an improved complete response rate. Conclusion bRT led to substantial reductions in MTV, SUVmax, SUVmean, and TLG. The relative extent of these decreases correlated with improved outcomes after CAR T-cell infusion. Prospective cohorts should validate the value of interim PET following bRT for quantifying changes in disease burden and associated prognosis.
Collapse
Affiliation(s)
- Colton Ladbury
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Claire Hao
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - William Tyler Watkins
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Sagus Sampath
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Jeffrey Wong
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Arya Amini
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Karen Sokolov
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Jekwon Yeh
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | | | | | | | | | - Liana Nikolaenko
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Matthew Mei
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Alex Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Leslie Popplewell
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Lihua Elizabeth Budde
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Savita Dandapani
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
10
|
Boellaard R, Buvat I, Nioche C, Ceriani L, Cottereau AS, Guerra L, Hicks RJ, Kanoun S, Kobe C, Loft A, Schöder H, Versari A, Voltin CA, Zwezerijnen GJC, Zijlstra JM, Mikhaeel NG, Gallamini A, El-Galaly TC, Hanoun C, Chauvie S, Ricci R, Zucca E, Meignan M, Barrington SF. International Benchmark for Total Metabolic Tumor Volume Measurement in Baseline 18F-FDG PET/CT of Lymphoma Patients: A Milestone Toward Clinical Implementation. J Nucl Med 2024; 65:1343-1348. [PMID: 39089812 PMCID: PMC11372260 DOI: 10.2967/jnumed.124.267789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/21/2024] [Indexed: 08/04/2024] Open
Abstract
Total metabolic tumor volume (TMTV) is prognostic in lymphoma. However, cutoff values for risk stratification vary markedly, according to the tumor delineation method used. We aimed to create a standardized TMTV benchmark dataset allowing TMTV to be tested and applied as a reproducible biomarker. Methods: Sixty baseline 18F-FDG PET/CT scans were identified with a range of disease distributions (20 follicular, 20 Hodgkin, and 20 diffuse large B-cell lymphoma). TMTV was measured by 12 nuclear medicine experts, each analyzing 20 cases split across subtypes, with each case processed by 3-4 readers. LIFEx or ACCURATE software was chosen according to reader preference. Analysis was performed stepwise: TMTV1 with automated preselection of lesions using an SUV of at least 4 and a volume of at least 3 cm3 with single-click removal of physiologic uptake; TMTV2 with additional removal of reactive bone marrow and spleen with single clicks; TMTV3 with manual editing to remove other physiologic uptake, if required; and TMTV4 with optional addition of lesions using mouse clicks with an SUV of at least 4 (no volume threshold). Results: The final TMTV (TMTV4) ranged from 8 to 2,288 cm3, showing excellent agreement among all readers in 87% of cases (52/60) with a difference of less than 10% or less than 10 cm3 In 70% of the cases, TMTV4 equaled TMTV1, requiring no additional reader interaction. Differences in the TMTV4 were exclusively related to reader interpretation of lesion inclusion or physiologic high-uptake region removal, not to the choice of software. For 5 cases, large TMTV differences (>25%) were due to disagreement about inclusion of diffuse splenic uptake. Conclusion: The proposed segmentation method enabled highly reproducible TMTV measurements, with minimal reader interaction in 70% of the patients. The inclusion or exclusion of diffuse splenic uptake requires definition of specific criteria according to lymphoma subtype. The publicly available proposed benchmark allows comparison of study results and could serve as a reference to test improvements using other segmentation approaches.
Collapse
Affiliation(s)
- Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands;
| | | | | | - Luca Ceriani
- Clinic of Nuclear Medicine and PET-CT Centre, Imaging Institute of Southern Switzerland; and EOC, Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Anne-Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, APHP; and Faculté de Médecine, Université Paris Cité, Paris, France
| | - Luca Guerra
- Nuclear Medicine Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Rodney J Hicks
- Department of Medicine, St. Vincent's Hospital Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - Salim Kanoun
- Centre de Recherche Clinique de Toulouse, Team 9, Toulouse, France
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Annika Loft
- PET & Cyclotron Unit 3982, Copenhagen University Hospital, Copenhagen, Denmark
| | - Heiko Schöder
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Annibale Versari
- Nuclear Medicine Department, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| | - Conrad-Amadeus Voltin
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - N George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London University, London, United Kingdom
| | - Andrea Gallamini
- Research and Innovation Department, Antoine Lacassagne Cancer Center, Nice, France
| | - Tarec C El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Christine Hanoun
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stephane Chauvie
- Medical Physics Division, Santa Croce e Carle Hospital, Cuneo, Italy
| | - Romain Ricci
- LYSARC, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland; and EOC, Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland; and
| | - Michel Meignan
- Department of Nuclear Medicine, Cochin Hospital, APHP; and Faculté de Médecine, Université Paris Cité, Paris, France
| | - Sally F Barrington
- King's College London and Guy's and St. Thomas's PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
11
|
Al-Ibraheem A, Abdlkadir AS, Al-Adhami DA, Sathekge M, Bom HHS, Ma’koseh M, Mansour A, Abdel-Razeq H, Al-Rabi K, Estrada-Lobato E, Al-Hussaini M, Matalka I, Abdel Rahman Z, Fanti S. The prognostic utility of 18F-FDG PET parameters in lymphoma patients under CAR-T-cell therapy: a systematic review and meta-analysis. Front Immunol 2024; 15:1424269. [PMID: 39286245 PMCID: PMC11402741 DOI: 10.3389/fimmu.2024.1424269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy has attracted considerable attention since its recent endorsement by the Food and Drug Administration, as it has emerged as a promising immunotherapeutic modality within the landscape of oncology. This study explores the prognostic utility of [18F]Fluorodeoxyglucose positron emission tomography ([18F]FDG PET) in lymphoma patients undergoing CAR T-cell therapy. Through meta-analysis, pooled hazard ratio (HR) values were calculated for specific PET metrics in this context. METHODS PubMed, Scopus, and Ovid databases were explored to search for relevant topics. Dataset retrieval from inception until March 12, 2024, was carried out. The primary endpoints were impact of specific PET metrics on overall survival (OS) and progression-free survival (PFS) before and after treatment. Data from the studies were extracted for a meta-analysis using Stata 17.0. RESULTS Out of 27 studies identified for systematic review, 15 met the criteria for meta-analysis. Baseline OS analysis showed that total metabolic tumor volume (TMTV) had the highest HR of 2.66 (95% CI: 1.52-4.66), followed by Total-body total lesion glycolysis (TTLG) at 2.45 (95% CI: 0.98-6.08), and maximum standardized uptake values (SUVmax) at 1.30 (95% CI: 0.77-2.19). TMTV and TTLG were statistically significant (p < 0.0001), whereas SUVmax was not (p = 0.33). For PFS, TMTV again showed the highest HR at 2.65 (95% CI: 1.63-4.30), with TTLG at 2.35 (95% CI: 1.40-3.93), and SUVmax at 1.48 (95% CI: 1.08-2.04), all statistically significant (p ≤ 0.01). The ΔSUVmax was a significant predictor for PFS with an HR of 2.05 (95% CI: 1.13-3.69, p = 0.015). CONCLUSION [18F]FDG PET parameters are valuable prognostic tools for predicting outcome of lymphoma patients undergoing CAR T-cell therapy.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Ahmed Saad Abdlkadir
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Dhuha Ali Al-Adhami
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Henry Hee-Seung Bom
- Department of Nuclear Medicine, Chonnam National University Medical School (CNUMS) and Hospital, Gwangju, Republic of Korea
| | - Mohammad Ma’koseh
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Asem Mansour
- Department of Diagnostic Radiology, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Kamal Al-Rabi
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Enrique Estrada-Lobato
- Nuclear Medicine and Diagnostic Section, Division of Human Health, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Maysaa Al-Hussaini
- Department of Pathology, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Ismail Matalka
- Department of Pathology and Microbiology, King Abdullah University Hospital- Jordan University of Science and Technology, Irbid, Jordan
- Department of Pathology, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Zaid Abdel Rahman
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Stephano Fanti
- Nuclear Medicine Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Zhang H, Xu Z, Zhou W, Chen J, Wei Y, Wu H, Wei X, Feng R. Metabolic tumor volume from baseline [18 F]FDG PET/CT at diagnosis improves the IPI stratification in patients with diffuse large B-cell lymphoma. Ann Hematol 2024:10.1007/s00277-024-05717-9. [PMID: 39222121 DOI: 10.1007/s00277-024-05717-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/18/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Although several different parameters of PET/CT were reported to be predictive of survival in DLBCL, the best parameter remains to be elucidated and whether it could improve the risk stratification of IPI in patients with DLBCL. PROCEDURES 262 DLBCL patients including in the training and validation cohort were retrospectively analyzed in this study. RESULTS Among different parameters, MTV was identified as the optimal prognostic parameter with a maximum area under the curve (AUC) of 0.652 ± 0.112 than TLG and SDmax (0.645 ± 0.113 and 0.600 ± 0.117, respectively). Patients with high MTV were associated with inferior PFS (p < 0.001 and p = 0.021, respectively) and OS (p < 0.001 and p < 0.001, respectively) in both the training and validation cohort. The multivariate analysis revealed that high MTV was an unfavorable factor for PFS (relative ratio [RR], 2.295; 95% confidence interval [CI], 1.457-3.615; p < 0.01) and OS (RR, 2.929; 95% CI 1.679-5.109; p < 0.01) independent of IPI. CONCLUSIONS Further analysis showed MTV could improve the risk stratification of IPI for both PFS and OS (p < 0.01 and p < 0.01, respectively). In conclusion, our study suggests that MTV was an optimal prognostic parameter of PET/CT for survival and it could improve the risk stratification of IPI in DLBCL, which may help to guide treatment in clinical trial.
Collapse
Affiliation(s)
- Hanzhen Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Zihan Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Wenlan Zhou
- PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Junjie Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Yongqiang Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Hubing Wu
- PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China.
| | - Ru Feng
- Department of Hematology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, China.
| |
Collapse
|
13
|
Cui S, Xin W, Wang F, Shao X, Shao X, Niu R, Zhang F, Shi Y, Liu B, Gu W, Wang Y. Metabolic tumour area: a novel prognostic indicator based on 18F-FDG PET/CT in patients with diffuse large B-cell lymphoma in the R-CHOP era. BMC Cancer 2024; 24:895. [PMID: 39054508 PMCID: PMC11270790 DOI: 10.1186/s12885-024-12668-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND The metabolic tumour area (MTA) was found to be a promising predictor of prostate cancer. However, the role of MTA based on 18F-FDG PET/CT in diffuse large B-cell lymphoma (DLBCL) prognosis remains unclear. This study aimed to elucidate the prognostic significance of MTA and evaluate its incremental value to the National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI) for DLBCL patients treated with first-line R-CHOP regimens. METHODS A total of 280 consecutive patients with newly diagnosed DLBCL and baseline 18F-FDG PET/CT data were retrospectively evaluated. Lesions were delineated via a semiautomated segmentation method based on a 41% SUVmax threshold to estimate semiquantitative metabolic parameters such as total metabolic tumour volume (TMTV) and MTA. Receiver operating characteristic (ROC) curve analysis was used to determine the optimal cut-off values. Progression-free survival (PFS) and overall survival (OS) were the endpoints that were used to evaluate the prognosis. PFS and OS were estimated via Kaplan‒Meier curves and compared via the log-rank test. RESULTS Univariate analysis revealed that patients with high MTA, high TMTV and NCCN-IPI ≥ 4 were associated with inferior PFS and OS (P < 0.0001 for all). Multivariate analysis indicated that MTA remained an independent predictor of PFS and OS [hazard ratio (HR), 2.506; 95% confidence interval (CI), 1.337-4.696; P = 0.004; and HR, 1.823; 95% CI, 1.005-3.310; P = 0.048], whereas TMTV was not. Further analysis using the NCCN-IPI model as a covariate revealed that MTA and NCCN-IPI were still independent predictors of PFS (HR, 2.617; 95% CI, 1.494-4.586; P = 0.001; and HR, 2.633; 95% CI, 1.650-4.203; P < 0.0001) and OS (HR, 2.021; 95% CI, 1.201-3.401; P = 0.008; and HR, 3.869; 95% CI, 1.959-7.640; P < 0.0001; respectively). Furthermore, MTA was used to separate patients with high NCCN-IPI risk scores into two groups with significantly different outcomes. CONCLUSIONS Pre-treatment MTA based on 18F-FDG PET/CT and NCCN-IPI were independent predictor of PFS and OS in DLBCL patients treated with R-CHOP. MTA has additional predictive value for the prognosis of patients with DLBCL, especially in high-risk patients with NCCN-IPI ≥ 4. In addition, the combination of MTA and NCCN-IPI may be helpful in further improving risk stratification and guiding individualised treatment options. TRIAL REGISTRATION This research was retrospectively registered with the Ethics Committee of the Third Affiliated Hospital of Soochow University, and the registration number was approval No. 155 (approved date: 31 May 2022).
Collapse
Affiliation(s)
- Silu Cui
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
- Yangzhou University, Yangzhou, Jiangsu, China
| | - Wenchong Xin
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
- Department of Nuclear Medicine, Linyi People's Hospital, Linyi, Shandong, China
| | - Fei Wang
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China.
| | - Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Rong Niu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Feifei Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Yunmei Shi
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Bao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
14
|
Yousefirizi F, Klyuzhin IS, O JH, Harsini S, Tie X, Shiri I, Shin M, Lee C, Cho SY, Bradshaw TJ, Zaidi H, Bénard F, Sehn LH, Savage KJ, Steidl C, Uribe CF, Rahmim A. TMTV-Net: fully automated total metabolic tumor volume segmentation in lymphoma PET/CT images - a multi-center generalizability analysis. Eur J Nucl Med Mol Imaging 2024; 51:1937-1954. [PMID: 38326655 DOI: 10.1007/s00259-024-06616-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024]
Abstract
PURPOSE Total metabolic tumor volume (TMTV) segmentation has significant value enabling quantitative imaging biomarkers for lymphoma management. In this work, we tackle the challenging task of automated tumor delineation in lymphoma from PET/CT scans using a cascaded approach. METHODS Our study included 1418 2-[18F]FDG PET/CT scans from four different centers. The dataset was divided into 900 scans for development/validation/testing phases and 518 for multi-center external testing. The former consisted of 450 lymphoma, lung cancer, and melanoma scans, along with 450 negative scans, while the latter consisted of lymphoma patients from different centers with diffuse large B cell, primary mediastinal large B cell, and classic Hodgkin lymphoma cases. Our approach involves resampling PET/CT images into different voxel sizes in the first step, followed by training multi-resolution 3D U-Nets on each resampled dataset using a fivefold cross-validation scheme. The models trained on different data splits were ensemble. After applying soft voting to the predicted masks, in the second step, we input the probability-averaged predictions, along with the input imaging data, into another 3D U-Net. Models were trained with semi-supervised loss. We additionally considered the effectiveness of using test time augmentation (TTA) to improve the segmentation performance after training. In addition to quantitative analysis including Dice score (DSC) and TMTV comparisons, the qualitative evaluation was also conducted by nuclear medicine physicians. RESULTS Our cascaded soft-voting guided approach resulted in performance with an average DSC of 0.68 ± 0.12 for the internal test data from developmental dataset, and an average DSC of 0.66 ± 0.18 on the multi-site external data (n = 518), significantly outperforming (p < 0.001) state-of-the-art (SOTA) approaches including nnU-Net and SWIN UNETR. While TTA yielded enhanced performance gains for some of the comparator methods, its impact on our cascaded approach was found to be negligible (DSC: 0.66 ± 0.16). Our approach reliably quantified TMTV, with a correlation of 0.89 with the ground truth (p < 0.001). Furthermore, in terms of visual assessment, concordance between quantitative evaluations and clinician feedback was observed in the majority of cases. The average relative error (ARE) and the absolute error (AE) in TMTV prediction on external multi-centric dataset were ARE = 0.43 ± 0.54 and AE = 157.32 ± 378.12 (mL) for all the external test data (n = 518), and ARE = 0.30 ± 0.22 and AE = 82.05 ± 99.78 (mL) when the 10% outliers (n = 53) were excluded. CONCLUSION TMTV-Net demonstrates strong performance and generalizability in TMTV segmentation across multi-site external datasets, encompassing various lymphoma subtypes. A negligible reduction of 2% in overall performance during testing on external data highlights robust model generalizability across different centers and cancer types, likely attributable to its training with resampled inputs. Our model is publicly available, allowing easy multi-site evaluation and generalizability analysis on datasets from different institutions.
Collapse
Affiliation(s)
- Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| | - Ivan S Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Joo Hyun O
- College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Xin Tie
- Department of Radiology, University of WI-Madison, Madison, WI, USA
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Muheon Shin
- Department of Radiology, University of WI-Madison, Madison, WI, USA
| | - Changhee Lee
- Department of Radiology, University of WI-Madison, Madison, WI, USA
| | - Steve Y Cho
- Department of Radiology, University of WI-Madison, Madison, WI, USA
| | - Tyler J Bradshaw
- Department of Radiology, University of WI-Madison, Madison, WI, USA
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
- University Research and Innovation Center, Óbuda University, Budapest, Hungary
| | - François Bénard
- BC Cancer, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Laurie H Sehn
- BC Cancer, Vancouver, BC, Canada
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, Canada
| | - Kerry J Savage
- BC Cancer, Vancouver, BC, Canada
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, Canada
| | - Christian Steidl
- BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Carlos F Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
- BC Cancer, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
- Departments of Physics and Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| |
Collapse
|
15
|
Keijzer K, de Boer JW, van Doesum JA, Noordzij W, Huls GA, van Dijk LV, van Meerten T, Niezink AGH. Reducing and controlling metabolic active tumor volume prior to CAR T-cell infusion can improve survival outcomes in patients with large B-cell lymphoma. Blood Cancer J 2024; 14:41. [PMID: 38448432 PMCID: PMC10917787 DOI: 10.1038/s41408-024-01022-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024] Open
Abstract
Bridging therapy before CD19-directed chimeric antigen receptor (CAR) T-cell infusion is frequently applied in patients with relapsed or refractory Large B-cell lymphoma (r/r LBCL). This study aimed to assess the influence of quantified MATV and MATV-dynamics, between pre-apheresis (baseline) and pre-lymphodepleting chemotherapy (pre-LD) MATV, on CAR T-cell outcomes and toxicities in patients with r/r LBCL. MATVs were calculated semi-automatically at baseline (n = 74) and pre-LD (n = 68) in patients with r/r LBCL who received axicabtagene ciloleucel. At baseline, patients with a low MATV (< 190 cc) had a better time to progression (TTP) and overall survival (OS) compared to high MATV patients (p < 0.001). High MATV patients who remained stable or reduced upon bridging therapy showed a significant improvement in TTP (p = 0.041) and OS (p = 0.015), compared to patients with a high pre-LD MATV (> 480 cc). Furthermore, high MATV baseline was associated with severe cytokine release syndrome (CRS, p = 0.001). In conclusion, patients with low baseline MATV had the best TTP/OS and effective reduction or controlling MATV during bridging improved survival outcomes in patients with a high baseline MATV, providing rationale for the use of more aggressive bridging regimens.
Collapse
Affiliation(s)
- Kylie Keijzer
- Department of Hematology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Janneke W de Boer
- Department of Hematology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Jaap A van Doesum
- Department of Hematology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Gerwin A Huls
- Department of Hematology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Tom van Meerten
- Department of Hematology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Anne G H Niezink
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands.
| |
Collapse
|
16
|
Barrington SF, Cottereau AS, Zijlstra JM. Is 18F-FDG Metabolic Tumor Volume in Lymphoma Really Happening? J Nucl Med 2024; 65:jnumed.123.267022. [PMID: 38388515 PMCID: PMC10995527 DOI: 10.2967/jnumed.123.267022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Affiliation(s)
- Sally F Barrington
- King's College London and Guy's and St. Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom;
| | - Anne-Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, APHP, Paris Cité University, Paris, France; and
| | - Josée M Zijlstra
- Department of Hematology and Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| |
Collapse
|
17
|
Zhu YM, Peng P, Liu X, Qi SN, Wang SL, Fang H, Song YW, Liu YP, Jin J, Li N, Lu NN, Jing H, Tang Y, Chen B, Zhang WW, Zhai YR, Yang Y, Liang B, Zheng R, Li YX. Optimizing the prognostic capacity of baseline 18F-FDG PET/CT metabolic parameters in extranodal natural killer/T-cell lymphoma by using relative and absolute thresholds. Heliyon 2024; 10:e25184. [PMID: 38322946 PMCID: PMC10844272 DOI: 10.1016/j.heliyon.2024.e25184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Objectives To investigate the prognostic capacity of baseline 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) metabolic parameters in extranodal natural killer/T-cell lymphoma (ENKTCL), and the influence of relative thresholds (RT) and absolute thresholds (AT) selection on prognostic capacity. Materials and methods Metabolic tumor volume (MTV)-based parameters were defined using RTs (41 % or 25 % of maximum standardized uptake value [SUVmax]), ATs (SUV 2.5, 3.0, 4.0, or mean liver uptake) in 133 patients. Metabolic parameters were classified into avidity-related parameters (SUVmax, mean SUV [SUVmean], standard deviation of SUV [SUVsd]), volume-related parameters (RT-MTV), and avidity- and volume-related parameters (total lesion glycolysis [TLG] and AT-MTV). The prognostic capacity of the metabolic parameters and the effects of different threshold types (RT vs. AT) were evaluated. Results All metabolic parameters were moderately associated with prognosis. However, the area under the receiver operating characteristic curve of MTV and TLG was slightly higher than that of avidity-related parameters for predicting 5-year progression-free survival (PFS) (0.614-0.705 vs. 0.563-0.609) and overall survival (OS) (0.670-0.748 vs. 0.562-0.593). Correlations of MTV and avidity-related parameters differed between RTs (r < 0.06, P = 0.324-0.985) and ATs (r 0.56-0.84, P ≤ 0.001). AT-MTV was the optimal predictor for PFS and OS, while RT-TLG was the optimal predictor for PFS, and the combination of RT-MTV with SUVmax was the optimal predictor for OS. Conclusion The incorporation of volume and avidity significantly improved the prognostic capacity of PET in ENKTCL. Composite parameters that encompassed both avidity and volume were recommended.
Collapse
Affiliation(s)
- Ying-Ming Zhu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Pan Peng
- Department of Nuclear Medicine, National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing, China
| | - Xin Liu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital & Shenzhen Hospital, CAMS and PUMC, Shenzhen, China
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Ning-Ning Lu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Wen-Wen Zhang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yi-Rui Zhai
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yong Yang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bin Liang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Rong Zheng
- Department of Nuclear Medicine, National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing, China
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
18
|
Rojek AE, Kline JP, Feinberg N, Appelbaum DE, Pu Y, Derman BA, Jakubowiak A, Kosuri S, Liu H, Nawas MT, Smith SM, Bishop MR, Riedell PA. Optimization of Metabolic Tumor Volume as a Prognostic Marker in CAR T-Cell Therapy for Aggressive Large B-cell NHL. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:83-93. [PMID: 37827881 DOI: 10.1016/j.clml.2023.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a standard of care in relapsed/refractory (R/R) aggressive large B-cell non-Hodgkin lymphomas (B-NHL) though the majority of recipients do not receive durable disease benefit, prompting the need to better define risk factors for relapse/progression. OBJECTIVES We performed a single-center, retrospective analysis of patients treated with commercial CAR T-cell therapy to evaluate the impact of tumor burden, as measured by whole-body metabolic tumor volume (MTV) from 18F fluorodeoxyglucose PET imaging, on treatment outcomes. STUDY DESIGN Sixty-one patients treated with CAR T-cell therapy for R/R B-NHL between May 2016 and November 2021 were included. RESULTS Using a receiver operating characteristic curve-based MTV optimization cutoff of 450 mL, 1-year progression-free survival (PFS) was 22% for high MTV versus 54% for low MTV (P < .01), and 1-year overall survival (OS) was 37% and 73%, respectively (P = .01). In a subset of 46 patients, residual MTV of less than 106 mL at the day 30 (D30) disease assessment was associated with significantly improved outcomes (1-year OS 85% vs. 13%, P < .01). Incorporation of pretreatment MTV to the International Prognostic Index (IPI) scoring system significantly distinguished 2-year PFS and OS outcomes by 3 risk groups. CONCLUSIONS Our findings suggest that both pretreatment and D30 MTV are predictive of outcomes among R/R B-NHL patients treated with CAR T-cell therapy. These data indicate that efforts to reduce pretreatment tumor burden may improve longitudinal clinical outcomes. Furthermore, D30 postinfusion MTV quantification may aid clinicians in optimally identifying patients at high-risk for progression, and in whom closer disease monitoring should be considered. MTV also adds prognostic value to patients with high-risk IPI and holds promise for incorporation in novel risk scoring systems which can identify patients prior to CAR T-cell therapy at highest risk of adverse outcomes.
Collapse
Affiliation(s)
- Alexandra E Rojek
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL
| | - Justin P Kline
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Nicholas Feinberg
- Department of Radiology, Section of Nuclear Medicine, University of Chicago, Chicago, IL
| | - Daniel E Appelbaum
- Department of Radiology, Section of Nuclear Medicine, University of Chicago, Chicago, IL
| | - Yonglin Pu
- Department of Radiology, Section of Nuclear Medicine, University of Chicago, Chicago, IL
| | - Benjamin A Derman
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Andrzej Jakubowiak
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Satyajit Kosuri
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Hongtao Liu
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Mariam T Nawas
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Sonali M Smith
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL
| | - Michael R Bishop
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Peter A Riedell
- Department of Medicine, Section of Hematology-Oncology, University of Chicago, Chicago, IL; David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL.
| |
Collapse
|
19
|
Marchal E, Palard-Novello X, Lhomme F, Meyer ME, Manson G, Devillers A, Marolleau JP, Houot R, Girard A. Baseline [ 18F]FDG PET features are associated with survival and toxicity in patients treated with CAR T cells for large B cell lymphoma. Eur J Nucl Med Mol Imaging 2024; 51:481-489. [PMID: 37721580 DOI: 10.1007/s00259-023-06427-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/04/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T cells have established themselves as an effective treatment for refractory or relapsed large B cell lymphoma (LBCL). Recently, the sDmax, which corresponds to the distance separating the two farthest lesions standardized by the patient's body surface area, has appeared as a prognostic factor in LBCL. This study aimed to identify [18F]FDG-PET biomarkers associated with prognosis and predictive of adverse events in patients treated with CAR T cells. METHODS Patients were retrospectively included from two different university hospitals. They were being treated with CAR T cells for LBCL and underwent [18F]FDG-PET just before CAR T cell infusion. Lesions were segmented semi-automatically with a threshold of 41% of the maximal uptake. In addition to clinico-biological features, sDmax, total metabolic tumor volume (TMTV), SUVmax, and uptake intensity of healthy lymphoid organs and liver were collected. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. The occurrence of adverse events, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), was reported. RESULTS Fifty-six patients were included. The median follow-up was 9.7 months. Multivariate analysis showed that TMTV (cut-off of 36 mL) was an independent prognostic factor for PFS (p < 0.001) and that sDmax (cut-off of 0.15 m-1) was an independent prognostic factor for OS (p = 0.008). Concerning the occurrence of adverse events, a C-reactive protein level > 35 mg/L (p = 0.006) and a liver SUVmean > 2.5 (p = 0.027) before CAR T cells were associated with grade 2 to 4 CRS and a spleen SUVmean > 1.9 with grade 2 to 4 ICANS. CONCLUSION TMTV and sDmax had independent prognostic values, respectively, on PFS and OS. Regarding adverse events, the mean liver and spleen uptakes were associated with the occurrence of grade 2 to 4 CRS and ICANS, respectively. Integrating these biomarkers into the clinical workflow could be useful for early adaptation of patients management.
Collapse
Affiliation(s)
- E Marchal
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France.
| | - X Palard-Novello
- Department of Nuclear Medicine, University Rennes, CLCC Eugène Marquis, INSERM, LTSI-UMR 1099, Rennes, France
| | - F Lhomme
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - M E Meyer
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France
| | - G Manson
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - A Devillers
- Department of Nuclear Medicine, CLCC Eugène Marquis, Rennes, France
| | - J P Marolleau
- Department of Hematology, Amiens-Picardie University Hospital, Amiens, France
| | - R Houot
- Department of Clinical Hematology, Rennes University Hospital, Rennes, France
| | - A Girard
- Department of Nuclear Medicine, Amiens-Picardie University Hospital, Amiens, France
| |
Collapse
|
20
|
Lewis KL, Trotman J. Integration of PET in DLBCL. Semin Hematol 2023; 60:291-304. [PMID: 38326144 DOI: 10.1053/j.seminhematol.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 02/09/2024]
Abstract
F-fluorodeoxyglucose positron emission tomography-computerized tomography (18FDG-PET/CT) is the gold-standard imaging modality for staging and response assessment for most lymphomas. This review focuses on the utility of 18FDG-PET/CT, and its role in staging, prognostication and response assessment in diffuse large B-cell lymphoma (DLBCL), including emerging possibilities for future use.
Collapse
Affiliation(s)
| | - Judith Trotman
- Concord Repatriation General Hospital, Concord, NSW, Australia
| |
Collapse
|
21
|
Wang L, Zhang S, Xin J. Predicting diffuse large B-cell lymphoma outcomes with lesion-to-liver maximum standardized uptake value for interim-treatment and end-of-treatment positron emission tomography-computed tomography. Quant Imaging Med Surg 2023; 13:6789-6800. [PMID: 37869355 PMCID: PMC10585501 DOI: 10.21037/qims-23-251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/24/2023] [Indexed: 10/24/2023]
Abstract
Background 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography-computed tomography (PET-CT) has been used in response evaluation systems for malignant lymphomas and is an important tool for determining efficacy and prognosis. The Deauville 5-point scale (D-5PS) is an 18F-FDG PET-CT image-interpretation protocol for patients with lymphoma. Nevertheless, a number of limitations in visual image interpretation, such as interobserver disagreement and the increase of false-positive results, suggests that new parameters are needed. In this study, we aimed to evaluate the prognostic values of interim-treatment (I-) and end-of-treatment (EOT) PET-CT by comparing D-5PS to the semiquantitative lesion-to-liver maximum standardized uptake value ratio (RLL). Methods A total of 90 patients with diffuse large B-cell lymphoma (DLBCL) (45 I-PET and 45 EOT-PET) were analyzed, and the RLL was calculated. Patients were additionally evaluated using the D-5PS system. We determined the optimal cutoff value of RLL using receiver operating characteristic (ROC) analysis. Kaplan-Meier survival analysis was used to compare the outcome predictions, while multivariate Cox regression analysis was used to identify the predictive factors. Results Among the patients examined, 41 (20 I-PET and 21 EOT-PET) experienced progression, and 49 (25 I-PET, 24 EOT-PET) did not. The optimal cutoff values of the RLL for predicting disease progression were 1.37 for I-PET (sensitivity 75%, specificity 88%) and 2.03 for EOT-PET (sensitivity 45.5%, specificity 100%), while the cutoffs of the D-5PS were scores 4 for I-PET (sensitivity 80%, specificity 72%) and 5 for EOT-PET (sensitivity 40.9%, specificity 100%). The prognostic efficacy was higher for the RLL at interim than for the D-5PS [area under the curve (AUC) =0.848 vs. 0.741]. The EOT prognostic efficacy of both evaluation methods was essentially equivalent (AUC =0.785 vs. 0.725). Univariate and multivariate analyses showed that RLL and D-5PS were independent factors affecting DLBCL outcomes for both interim and EOT assessment. Conclusions RLL and D-5PS have independent predictive values for the interim and EOT evaluation of outcomes in patients with DLBCL. The RLL has better interim predictive ability than does D-5PS and can optimize D-5PS interpretation, thus improving interim outcome prediction.
Collapse
Affiliation(s)
- Lu Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shixiong Zhang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Zhao S, Wang J, Jin C, Zhang X, Xue C, Zhou R, Zhong Y, Liu Y, He X, Zhou Y, Xu C, Zhang L, Qian W, Zhang H, Zhang X, Tian M. Stacking Ensemble Learning-Based [ 18F]FDG PET Radiomics for Outcome Prediction in Diffuse Large B-Cell Lymphoma. J Nucl Med 2023; 64:1603-1609. [PMID: 37500261 DOI: 10.2967/jnumed.122.265244] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/31/2023] [Indexed: 07/29/2023] Open
Abstract
This study aimed to develop an analytic approach based on [18F]FDG PET radiomics using stacking ensemble learning to improve the outcome prediction in diffuse large B-cell lymphoma (DLBCL). Methods: In total, 240 DLBCL patients from 2 medical centers were divided into the training set (n = 141), internal testing set (n = 61), and external testing set (n = 38). Radiomics features were extracted from pretreatment [18F]FDG PET scans at the patient level using 4 semiautomatic segmentation methods (SUV threshold of 2.5, SUV threshold of 4.0 [SUV4.0], 41% of SUVmax, and SUV threshold of mean liver uptake [PERCIST]). All extracted features were harmonized with the ComBat method. The intraclass correlation coefficient was used to evaluate the reliability of radiomics features extracted by different segmentation methods. Features from the most reliable segmentation method were selected by Pearson correlation coefficient analysis and the LASSO (least absolute shrinkage and selection operator) algorithm. A stacking ensemble learning approach was applied to build radiomics-only and combined clinical-radiomics models for prediction of 2-y progression-free survival and overall survival based on 4 machine learning classifiers (support vector machine, random forests, gradient boosting decision tree, and adaptive boosting). Confusion matrix, receiver-operating-characteristic curve analysis, and survival analysis were used to evaluate the model performance. Results: Among 4 semiautomatic segmentation methods, SUV4.0 segmentation yielded the highest interobserver reliability, with 830 (66.7%) selected radiomics features. The combined model constructed by the stacking method achieved the best discrimination performance. For progression-free survival prediction in the external testing set, the areas under the receiver-operating-characteristic curve and accuracy of the stacking-based combined model were 0.771 and 0.789, respectively. For overall survival prediction, the stacking-based combined model achieved an area under the curve of 0.725 and an accuracy of 0.763 in the external testing set. The combined model also demonstrated a more distinct risk stratification than the International Prognostic Index in all sets (log-rank test, all P < 0.05). Conclusion: The combined model that incorporates [18F]FDG PET radiomics and clinical characteristics based on stacking ensemble learning could enable improved risk stratification in DLBCL.
Collapse
Affiliation(s)
- Shuilin Zhao
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xiang Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chenxi Xue
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yuwei Liu
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xuexin He
- Department of Medical Oncology, Huashan Hospital of Fudan University, Shanghai, China
| | - Youyou Zhou
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Caiyun Xu
- Department of Nuclear Medicine, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Lixia Zhang
- Department of Nuclear Medicine, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Wenbin Qian
- Department of Hematology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China;
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China; and
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China;
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Ferrández MC, Golla SSV, Eertink JJ, de Vries BM, Wiegers SE, Zwezerijnen GJC, Pieplenbosch S, Schilder L, Heymans MW, Zijlstra JM, Boellaard R. Sensitivity of an AI method for [ 18F]FDG PET/CT outcome prediction of diffuse large B-cell lymphoma patients to image reconstruction protocols. EJNMMI Res 2023; 13:88. [PMID: 37758869 PMCID: PMC10533444 DOI: 10.1186/s13550-023-01036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Convolutional neural networks (CNNs), applied to baseline [18F]-FDG PET/CT maximum intensity projections (MIPs), show potential for treatment outcome prediction in diffuse large B-cell lymphoma (DLBCL). The aim of this study is to investigate the robustness of CNN predictions to different image reconstruction protocols. Baseline [18F]FDG PET/CT scans were collected from 20 DLBCL patients. EARL1, EARL2 and high-resolution (HR) protocols were applied per scan, generating three images with different image qualities. Image-based transformation was applied by blurring EARL2 and HR images to generate EARL1 compliant images using a Gaussian filter of 5 and 7 mm, respectively. MIPs were generated for each of the reconstructions, before and after image transformation. An in-house developed CNN predicted the probability of tumor progression within 2 years for each MIP. The difference in probabilities per patient was then calculated between both EARL2 and HR with respect to EARL1 (delta probabilities or ΔP). We compared these to the probabilities obtained after aligning the data with ComBat using the difference in median and interquartile range (IQR). RESULTS CNN probabilities were found to be sensitive to different reconstruction protocols (EARL2 ΔP: median = 0.09, interquartile range (IQR) = [0.06, 0.10] and HR ΔP: median = 0.1, IQR = [0.08, 0.16]). Moreover, higher resolution images (EARL2 and HR) led to higher probability values. After image-based and ComBat transformation, an improved agreement of CNN probabilities among reconstructions was found for all patients. This agreement was slightly better after image-based transformation (transformed EARL2 ΔP: median = 0.022, IQR = [0.01, 0.02] and transformed HR ΔP: median = 0.029, IQR = [0.01, 0.03]). CONCLUSION Our CNN-based outcome predictions are affected by the applied reconstruction protocols, yet in a predictable manner. Image-based harmonization is a suitable approach to harmonize CNN predictions across image reconstruction protocols.
Collapse
Affiliation(s)
- Maria C Ferrández
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Sandeep S V Golla
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bart M de Vries
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Louise Schilder
- Department of Internal Medicine, Amstelland Hospital, Amstelveen, The Netherlands
| | - Martijn W Heymans
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Xue H, Fang Q, Yao Y, Teng Y. 3D PET/CT tumor segmentation based on nnU-Net with GCN refinement. Phys Med Biol 2023; 68:185018. [PMID: 37549672 DOI: 10.1088/1361-6560/acede6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/07/2023] [Indexed: 08/09/2023]
Abstract
Objective. Whole-body positron emission tomography/computed tomography (PET/CT) scans are an important tool for diagnosing various malignancies (e.g. malignant melanoma, lymphoma, or lung cancer), and accurate segmentation of tumors is a key part of subsequent treatment. In recent years, convolutional neural network based segmentation methods have been extensively investigated. However, these methods often give inaccurate segmentation results, such as oversegmentation and undersegmentation. To address these issues, we propose a postprocessing method based on a graph convolutional network (GCN) to refine inaccurate segmentation results and improve the overall segmentation accuracy.Approach. First, nnU-Net is used as an initial segmentation framework, and the uncertainty in the segmentation results is analyzed. Certain and uncertain pixels are used to establish the nodes of a graph. Each node and its 6 neighbors form an edge, and 32 nodes are randomly selected as uncertain nodes to form edges. The highly uncertain nodes are used as the subsequent refinement targets. Second, the nnU-Net results of the certain nodes are used as labels to form a semisupervised graph network problem, and the uncertain part is optimized by training the GCN to improve the segmentation performance. This describes our proposed nnU-Net + GCN segmentation framework.Main results.We perform tumor segmentation experiments with the PET/CT dataset from the MICCIA2022 autoPET challenge. Among these data, 30 cases are randomly selected for testing, and the experimental results show that the false-positive rate is effectively reduced with nnU-Net + GCN refinement. In quantitative analysis, there is an improvement of 2.1% for the average Dice score, 6.4 for the 95% Hausdorff distance (HD95), and 1.7 for the average symmetric surface distance.Significance. The quantitative and qualitative evaluation results show that GCN postprocessing methods can effectively improve the tumor segmentation performance.
Collapse
Affiliation(s)
- Hengzhi Xue
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110004, People's Republic of China
| | - Qingqing Fang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110004, People's Republic of China
| | - Yudong Yao
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110004, People's Republic of China
- Department of Electrical and Computer Engineering, Steven Institute of Technology, Hoboken, NJ 07102, United States of America
| | - Yueyang Teng
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110004, People's Republic of China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, 110169, People's Republic of China
| |
Collapse
|
25
|
Constantino CS, Leocádio S, Oliveira FPM, Silva M, Oliveira C, Castanheira JC, Silva Â, Vaz S, Teixeira R, Neves M, Lúcio P, João C, Costa DC. Evaluation of Semiautomatic and Deep Learning-Based Fully Automatic Segmentation Methods on [ 18F]FDG PET/CT Images from Patients with Lymphoma: Influence on Tumor Characterization. J Digit Imaging 2023; 36:1864-1876. [PMID: 37059891 PMCID: PMC10407010 DOI: 10.1007/s10278-023-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
The objective is to assess the performance of seven semiautomatic and two fully automatic segmentation methods on [18F]FDG PET/CT lymphoma images and evaluate their influence on tumor quantification. All lymphoma lesions identified in 65 whole-body [18F]FDG PET/CT staging images were segmented by two experienced observers using manual and semiautomatic methods. Semiautomatic segmentation using absolute and relative thresholds, k-means and Bayesian clustering, and a self-adaptive configuration (SAC) of k-means and Bayesian was applied. Three state-of-the-art deep learning-based segmentations methods using a 3D U-Net architecture were also applied. One was semiautomatic and two were fully automatic, of which one is publicly available. Dice coefficient (DC) measured segmentation overlap, considering manual segmentation the ground truth. Lymphoma lesions were characterized by 31 features. Intraclass correlation coefficient (ICC) assessed features agreement between different segmentation methods. Nine hundred twenty [18F]FDG-avid lesions were identified. The SAC Bayesian method achieved the highest median intra-observer DC (0.87). Inter-observers' DC was higher for SAC Bayesian than manual segmentation (0.94 vs 0.84, p < 0.001). Semiautomatic deep learning-based median DC was promising (0.83 (Obs1), 0.79 (Obs2)). Threshold-based methods and publicly available 3D U-Net gave poorer results (0.56 ≤ DC ≤ 0.68). Maximum, mean, and peak standardized uptake values, metabolic tumor volume, and total lesion glycolysis showed excellent agreement (ICC ≥ 0.92) between manual and SAC Bayesian segmentation methods. The SAC Bayesian classifier is more reproducible and produces similar lesion features compared to manual segmentation, giving the best concordant results of all other methods. Deep learning-based segmentation can achieve overall good segmentation results but failed in few patients impacting patients' clinical evaluation.
Collapse
Affiliation(s)
- Cláudia S Constantino
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal.
| | - Sónia Leocádio
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Francisco P M Oliveira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Mariana Silva
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Carla Oliveira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Joana C Castanheira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Ângelo Silva
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Sofia Vaz
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Ricardo Teixeira
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Manuel Neves
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Paulo Lúcio
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Cristina João
- Hematology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| | - Durval C Costa
- Nuclear Medicine - Radiopharmacology Department, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| |
Collapse
|
26
|
Lim CH, Choi JY, Choi JH, Lee JH, Lee J, Lim CW, Kim Z, Woo SK, Park SB, Park JM. Development and External Validation of 18F-FDG PET-Based Radiomic Model for Predicting Pathologic Complete Response after Neoadjuvant Chemotherapy in Breast Cancer. Cancers (Basel) 2023; 15:3842. [PMID: 37568658 PMCID: PMC10417050 DOI: 10.3390/cancers15153842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The aim of our retrospective study is to develop and externally validate an 18F-FDG PET-derived radiomics model for predicting pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC) in breast cancer patients. A total of 87 breast cancer patients underwent curative surgery after NAC at Soonchunhyang University Seoul Hospital and were randomly assigned to a training cohort and an internal validation cohort. Radiomic features were extracted from pretreatment PET images. A radiomic-score model was generated using the LASSO method. A combination model incorporating significant clinical variables was constructed. These models were externally validated in a separate cohort of 28 patients from Soonchunhyang University Buscheon Hospital. The model performances were assessed using area under the receiver operating characteristic (AUC). Seven radiomic features were selected to calculate the radiomic-score. Among clinical variables, human epidermal growth factor receptor 2 status was an independent predictor of pCR. The radiomic-score model achieved good discriminability, with AUCs of 0.963, 0.731, and 0.729 for the training, internal validation, and external validation cohorts, respectively. The combination model showed improved predictive performance compared to the radiomic-score model alone, with AUCs of 0.993, 0.772, and 0.906 in three cohorts, respectively. The 18F-FDG PET-derived radiomic-based model is useful for predicting pCR after NAC in breast cancer.
Collapse
Affiliation(s)
- Chae Hong Lim
- Department of Nuclear Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Joon Ho Choi
- Department of Nuclear Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea
| | - Jun-Hee Lee
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Jihyoun Lee
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea
| | - Cheol Wan Lim
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea
| | - Zisun Kim
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea
| | - Sang-Keun Woo
- Division of Applied RI, Korea Institutes of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Soo Bin Park
- Department of Nuclear Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
| | - Jung Mi Park
- Department of Nuclear Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea
| |
Collapse
|
27
|
Eertink JJ, Zwezerijnen GJC, Heymans MW, Pieplenbosch S, Wiegers SE, Dührsen U, Hüttmann A, Kurch L, Hanoun C, Lugtenburg PJ, Barrington SF, Mikhaeel NG, Ceriani L, Zucca E, Czibor S, Györke T, Chamuleau MED, Hoekstra OS, de Vet HCW, Boellaard R, Zijlstra JM, PETRA Consortium. Baseline PET radiomics outperforms the IPI risk score for prediction of outcome in diffuse large B-cell lymphoma. Blood 2023; 141:3055-3064. [PMID: 37001036 PMCID: PMC10646814 DOI: 10.1182/blood.2022018558] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/31/2023] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
The objective of this study is to externally validate the clinical positron emission tomography (PET) model developed in the HOVON-84 trial and to compare the model performance of our clinical PET model using the international prognostic index (IPI). In total, 1195 patients with diffuse large B-cell lymphoma (DLBCL) were included in the study. Data of 887 patients from 6 studies were used as external validation data sets. The primary outcomes were 2-year progression-free survival (PFS) and 2-year time to progression (TTP). The metabolic tumor volume (MTV), maximum distance between the largest lesion and another lesion (Dmaxbulk), and peak standardized uptake value (SUVpeak) were extracted. The predictive values of the IPI and clinical PET model (MTV, Dmaxbulk, SUVpeak, performance status, and age) were tested. Model performance was assessed using the area under the curve (AUC), and diagnostic performance, using the positive predictive value (PPV). The IPI yielded an AUC of 0.62. The clinical PET model yielded a significantly higher AUC of 0.71 (P < .001). Patients with high-risk IPI had a 2-year PFS of 61.4% vs 51.9% for those with high-risk clinical PET, with an increase in PPV from 35.5% to 49.1%, respectively. A total of 66.4% of patients with high-risk IPI were free from progression or relapse vs 55.5% of patients with high-risk clinical PET scores, with an increased PPV from 33.7% to 44.6%, respectively. The clinical PET model remained predictive of outcome in 6 independent first-line DLBCL studies, and had higher model performance than the currently used IPI in all studies.
Collapse
Affiliation(s)
- J. J. Eertink
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - G. J. C. Zwezerijnen
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M. W. Heymans
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - S. Pieplenbosch
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S. E. Wiegers
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - U. Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - A. Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - L. Kurch
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Leipzig, Leipzig, Germany
| | - C. Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - P. J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - S. F. Barrington
- King’s College London and Guy’s and St Thomas’ PET Centre, School of Biomedical Engineering and Imaging Sciences, King’s Health Partners, King’s College London, London, United Kingdom
| | - N. G. Mikhaeel
- Department of Clinical Oncology, Guy’s Cancer Centre and School of Cancer and Pharmaceutical Sciences, King’s College London University, London, United Kingdom
| | - L. Ceriani
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - E. Zucca
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, IOSI - Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - S. Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - T. Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - M. E. D. Chamuleau
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - O. S. Hoekstra
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H. C. W. de Vet
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - R. Boellaard
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J. M. Zijlstra
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - PETRA Consortium
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Leipzig, Leipzig, Germany
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- King’s College London and Guy’s and St Thomas’ PET Centre, School of Biomedical Engineering and Imaging Sciences, King’s Health Partners, King’s College London, London, United Kingdom
- Department of Clinical Oncology, Guy’s Cancer Centre and School of Cancer and Pharmaceutical Sciences, King’s College London University, London, United Kingdom
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, IOSI - Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Jin H, Jin M, Lim CH, Choi JY, Kim SJ, Lee KH. Metabolic bulk volume predicts survival in a homogeneous cohort of stage II/III diffuse large B-cell lymphoma patients undergoing R-CHOP treatment. Front Oncol 2023; 13:1186311. [PMID: 37384292 PMCID: PMC10293666 DOI: 10.3389/fonc.2023.1186311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/24/2023] [Indexed: 06/30/2023] Open
Abstract
Purpose Accurate risk stratification can improve lymphoma management, but current volumetric 18F-fluorodeoxyglucose (FDG) indicators require time-consuming segmentation of all lesions in the body. Herein, we investigated the prognostic values of readily obtainable metabolic bulk volume (MBV) and bulky lesion glycolysis (BLG) that measure the single largest lesion. Methods The study subjects were a homogeneous cohort of 242 newly diagnosed stage II or III diffuse large B-cell lymphoma (DLBCL) patients who underwent first-line R-CHOP treatment. Baseline PET/CT was retrospectively analyzed for maximum transverse diameter (MTD), total metabolic tumor volume (TMTV), total lesion glycolysis (TLG), MBV, and BLG. Volumes were drawn using 30% SUVmax as threshold. Kaplan-Meier survival analysis and the Cox proportional hazards model assessed the ability to predict overall survival (OS) and progression-free survival (PFS). Results During a median follow-up period of 5.4 years (maximum of 12.7 years), events occurred in 85 patients, including progression, relapse, and death (65 deaths occurred at a median of 17.6 months). Receiver operating characteristic (ROC) analysis identified an optimal TMTV of 112 cm3, MBV of 88 cm3, TLG of 950, and BLG of 750 for discerning events. Patients with high MBV were more likely to have stage III disease; worse ECOG performance; higher IPI risk score; increased LDH; and high SUVmax, MTD, TMTV, TLG, and BLG. Kaplan-Meier survival analysis showed that high TMTV (p = 0.005 and < 0.001), MBV (both p < 0.001), TLG (p < 0.001 and 0.008), and BLG (p = 0.018 and 0.049) were associated with significantly worse OS and PFS. On Cox multivariate analysis, older age (> 60 years; HR, 2.74; 95% CI, 1.58-4.75; p < 0.001) and high MBV (HR, 2.74; 95% CI, 1.05-6.54; p = 0.023) were independent predictors of worse OS. Older age (hazard ratio [HR], 2.90; 95% CI, 1.74-4.82; p < 0.001) and high MBV (HR, 2.36; 95% CI, 1.15-6.54; p = 0.032) were also independent predictors of worse PFS. Furthermore, among subjects ≤60 years, high MBV remained the only significant independent predictor of worse OS (HR, 4.269; 95% CI, 1.03-17.76; p = 0.046) and PFS (HR, 6.047; 95% CI, 1.73-21.11; p = 0.005). Among subjects with stage III disease, only greater age (HR, 2.540; 95% CI, 1.22-5.30; p = 0.013) and high MBV (HR, 6.476; 95% CI, 1.20-31.9; p = 0.030) were significantly associated with worse OS, while greater age was the only independent predictor of worse PFS (HR, 6.145; 95% CI, 1.10-4.17; p = 0.024). Conclusions MBV easily obtained from the single largest lesion may provide a clinically useful FDG volumetric prognostic indicator in stage II/III DLBCL patients treated with R-CHOP.
Collapse
Affiliation(s)
- Hyun Jin
- Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Myung Jin
- Department of Electrical and Computer Engineering, Seoul, Republic of Korea
| | - Chae Hong Lim
- Department of Nuclear Medicine, Soonchunhyang University School of Medicine, Seoul, Republic of Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok-Jin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Barrington SF. Advances in positron emission tomography and radiomics. Hematol Oncol 2023; 41 Suppl 1:11-19. [PMID: 37294959 PMCID: PMC10775708 DOI: 10.1002/hon.3137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
Positron emission tomography is established for staging and response evaluation in lymphoma using visual evaluation and semi-quantitative analysis. Radiomic analysis involving quantitative imaging features at baseline, such as metabolic tumor volume and markers of disease dissemination and changes in the standardized uptake value during treatment are emerging as powerful biomarkers. The combination of radiomic features with clinical risk factors and genomic analysis offers the potential to improve clinical risk prediction. This review discusses the state of current knowledge, progress toward standardization of tumor delineation for radiomic analysis and argues that radiomic features, molecular markers and circulating tumor DNA should be included in clinical trial designs to enable the development of baseline and dynamic risk scores that could further advance the field to facilitate testing of novel treatments and personalized therapy in aggressive lymphomas.
Collapse
Affiliation(s)
- Sally F. Barrington
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Campus, Kings College LondonLondonUK
| |
Collapse
|
30
|
Michaud L, Bantilan K, Mauguen A, Moskowitz CH, Zelenetz AD, Schöder H. Prognostic Value of 18F-FDG PET/CT in Diffuse Large B-Cell Lymphoma Treated with a Risk-Adapted Immunochemotherapy Regimen. J Nucl Med 2023; 64:536-541. [PMID: 36549918 PMCID: PMC10071786 DOI: 10.2967/jnumed.122.264740] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Early identification of patients with diffuse large B-cell lymphoma (DLBCL) who are likely to experience disease recurrence or refractory disease after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) would be useful for improving risk-adapted treatment strategies. We aimed to assess the prognostic value of 18F-FDG PET/CT parameters at baseline, interim, and end of treatment (EOT). Methods: We analyzed the prognostic impact of 18F-FDG PET/CT in 166 patients with DLBCL treated with a risk-adapted immunochemotherapy regimen. Scans were obtained at baseline, after 4 cycles of R-CHOP or 3 cycles of RR-CHOP (double dose of R) and 1 cycle of CHOP alone (interim) and 6 wk after completing therapy (EOT). Progression-free survival (PFS) and overall survival (OS) were estimated using Kaplan-Meier and the impact of clinical/PET factors assessed with Cox models. We also assessed the predictive ability of the recently proposed International Metabolic Prognostic Index (IMPI). Results: The median follow-up was 7.9 y. International Prognostic Index (IPI), baseline metabolic tumor volume (MTV), and change in maximum SUV (ΔSUVmax) at interim scans were statistically significant predictors for OS. Baseline MTV, interim ΔSUVmax, and EOT Deauville score were statistically significant predictors of PFS. Combining interim PET parameters demonstrated that patients with Deauville 4-5 and positive ΔSUVmax ≤ 70% at restaging (∼10% of the cohort) had extremely poor prognosis. The IMPI had limited discrimination and slightly overestimated the event rate in our cohort. Conclusion: Baseline MTV and interim ΔSUVmax predicted both PFS and OS with this sequential immunochemotherapy program. Combining interim Deauville score with interim ΔSUVmax may identify an extremely high-risk DLBCL population.
Collapse
Affiliation(s)
- Laure Michaud
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kurt Bantilan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Craig H Moskowitz
- Department of Medicine, University of Miami Health System, Miami, Florida
| | - Andrew D Zelenetz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
| |
Collapse
|
31
|
Czibor S, Carr R, Redondo F, Auewarakul CU, Cerci JJ, Paez D, Fanti S, Györke T. Prognostic parameters on baseline and interim [ 18 F]FDG-PET/computed tomography in diffuse large B-cell lymphoma patients. Nucl Med Commun 2023; 44:291-301. [PMID: 36705233 PMCID: PMC9994851 DOI: 10.1097/mnm.0000000000001664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE 2-[ 18 F]fluoro-2-deoxy- d -glucose PET/computed tomography ([ 18 F]FDG-PET/CT) is a widely used imaging method in the management of diffuse large B-cell lymphomas (DLBCL). Our aim was to investigate the prognostic performance of different PET biomarkers in a multicenter setting. METHODS We investigated baseline volumetric values [metabolic tumor volume (MTV) and total lesion glycolysis (TLG), also normalized for body weight] segmented with three different methods [>SUV4 (glob4); 41% isocontour (41pc), and a gradient-based lesion growing algorithm (grad)] and interim parameters [Deauville score, maximal standardized uptake value (ΔSUVmax), modified qPET, and ratio PET (rPET)] alongside clinical parameters (stage, revised International Prognostic Index), using 24-month progression-free survival as the clinical endpoint. Receiver operating characteristics analyses were performed to define optimal cutoff points for the continuous PET parameters. RESULTS A total of 107 diffuse large B-cell lymphoma patients were included (54 women; mean age: 53.7 years). MTV and TLG calculations showed good correlation among glob4, 41pc, and grad methods; however, optimal cutoff points were markedly different.Significantly different PFS was observed between low- and high-risk groups according to baseline MTV, body weight-adjusted (bwa) MTV, TLG, bwaTLG, as well as interim parameters Deauville score, ΔSUVmax, mqPET, and rPET. Univariate Cox regression analyses showed hazard ratios (HRs) lowest for bwaMTVglob4 (HR = 2.3) and highest for rPET (HR = 9.09). In a multivariate Cox-regression model, rPET was shown to be an independent predictor of PFS ( P = 0.041; HR = 9.15). Combined analysis showed that ΔSUVmax positive patients with high MTV formed a group with distinctly poor PFS (35.3%). CONCLUSION Baseline MTV and TLG values and optimal cutoff points achieved with different segmentation methods varied markedly and showed a limited prognostic impact. Interim PET/CT parameters provided more accurate prognostic information with semiquantitative 'Deauville-like' parameters performing best in the present study.
Collapse
Affiliation(s)
- Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Robert Carr
- Department of Hematology, Guy's and St. Thomas' Hospital, King's College London, London, UK
| | | | - Chirayu U Auewarakul
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Juliano J Cerci
- PET/CT Department at Quanta Diagnóstico e Terapia, Curitiba, Brazil
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Application, International Atomic Energy Agency, Vienna, Austria
| | - Stefano Fanti
- Metropolitan Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| |
Collapse
|
32
|
Dai J, Wang H, Xu Y, Chen X, Tian R. Clinical application of AI-based PET images in oncological patients. Semin Cancer Biol 2023; 91:124-142. [PMID: 36906112 DOI: 10.1016/j.semcancer.2023.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Based on the advantages of revealing the functional status and molecular expression of tumor cells, positron emission tomography (PET) imaging has been performed in numerous types of malignant diseases for diagnosis and monitoring. However, insufficient image quality, the lack of a convincing evaluation tool and intra- and interobserver variation in human work are well-known limitations of nuclear medicine imaging and restrict its clinical application. Artificial intelligence (AI) has gained increasing interest in the field of medical imaging due to its powerful information collection and interpretation ability. The combination of AI and PET imaging potentially provides great assistance to physicians managing patients. Radiomics, an important branch of AI applied in medical imaging, can extract hundreds of abstract mathematical features of images for further analysis. In this review, an overview of the applications of AI in PET imaging is provided, focusing on image enhancement, tumor detection, response and prognosis prediction and correlation analyses with pathology or specific gene mutations in several types of tumors. Our aim is to describe recent clinical applications of AI-based PET imaging in malignant diseases and to focus on the description of possible future developments.
Collapse
Affiliation(s)
- Jiaona Dai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang City 421001, China
| | - Xiyang Chen
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
33
|
Korsholm K, Overbeck N, Dias AH, Loft A, Andersen FL, Fischer BM. Impact of Reduced Image Noise on Deauville Scores in Patients with Lymphoma Scanned on a Long-Axial Field-of-View PET/CT-Scanner. Diagnostics (Basel) 2023; 13:diagnostics13050947. [PMID: 36900090 PMCID: PMC10000539 DOI: 10.3390/diagnostics13050947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Total body and long-axial field-of-view (LAFOV) PET/CT represent visionary innovations in imaging enabling either improved image quality, reduction in injected activity-dose or decreased acquisition time. An improved image quality may affect visual scoring systems, including the Deauville score (DS), which is used for clinical assessment of patients with lymphoma. The DS compares SUVmax in residual lymphomas with liver parenchyma, and here we investigate the impact of reduced image noise on the DS in patients with lymphomas scanned on a LAFOV PET/CT. METHODS Sixty-eight patients with lymphoma underwent a whole-body scan on a Biograph Vision Quadra PET/CT-scanner, and images were evaluated visually with regard to DS for three different timeframes of 90, 300, and 600 s. SUVmax and SUVmean were calculated from liver and mediastinal blood pool, in addition to SUVmax from residual lymphomas and measures of noise. RESULTS SUVmax in liver and in mediastinal blood pool decreased significantly with increasing acquisition time, whereas SUVmean remained stable. In residual tumor, SUVmax was stable during different acquisition times. As a result, the DS was subject to change in three patients. CONCLUSIONS Attention should be drawn towards the eventual impact of improvements in image quality on visual scoring systems such as the DS.
Collapse
Affiliation(s)
- Kirsten Korsholm
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
- Correspondence:
| | - Nanna Overbeck
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
| | - André H. Dias
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Flemming Littrup Andersen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Barbara Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
34
|
Xu H, Ma J, Yang G, Xiao S, Li W, Sun Y, Sun Y, Wang Z, Zhao H. Prognostic value of metabolic tumor volume and lesion dissemination from baseline PET/CT in patients with diffuse large B-cell lymphoma: further risk stratification of the group with low-risk and high-risk NCCN-IPI. Eur J Radiol 2023; 163:110798. [PMID: 37030099 DOI: 10.1016/j.ejrad.2023.110798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE The purpose of this study was to determine the prognostic value of metabolic tumor volume and lesion dissemination from baseline PET/CT in patients with diffuse large B-cell lymphoma (DLBCL) and the prognostic value of them in the National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI) subgroups. METHODS A total of 113 patients who underwent 18F-FDG PET/CT examination in our institution were retrospectively collected. The MTV was measured by iterative adaptive algorithm. The location of the lesion was obtained according to its three-dimensional coordinates, and Dmax was obtained. SDmax is derived from Dmax standardized by body surface area (BSA). The X-tile method was used to determine the optimal cut-off values for MTV, Dmax and SDmax. Cox regression analysis was used to perform univariate and multivariate analyses. Patient survival rates were derived from Kaplan-Meier curves and compared using the log-rank test. RESULTS The median follow-up time was 24 months. The median of MTV was 196.86 cm3 (range 2.54-2925.37 cm3), and the optimal cut-off value was 489 cm3. The median of SDmax was 0.25 m-1 (range 0.12-0.51 m-1), and the best cut-off value was 0.31 m-1. MTV and SDmax were independent prognostic factors of PFS (all P < 0.001). Combined with MTV and SDmax, the patients were divided into three groups, and the difference of PFS among the groups was statistically significant (P < 0.001), and was able to stratify the risk of NCCN-IPI patients in the low-risk (NCCN-IPI < 4) and high-risk (NCCN-IPI ≥ 4) groups (P = 0.001 and P = 0.031). CONCLUSION MTV and SDmax are independent prognostic factors for PFS in DCBCL patients, which describe tumor burden and tumor dissemination characteristics, respectively. The combination of the two could facilitate risk stratification between the low-risk and high-risk NCCN-IPI groups.
Collapse
Affiliation(s)
- Hong Xu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jie Ma
- Department of Nuclear Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guangjie Yang
- Department of Nuclear Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shuxin Xiao
- Department of Lymphoma, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenwen Li
- Department of Hematology, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Yue Sun
- Department of Blood Transfusion, Affiliated Hospital of Jining Medical College, Jining, Shandong, China
| | - Yujiao Sun
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhenguang Wang
- Department of Nuclear Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hongguo Zhao
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
35
|
Keijzer K, Niezink AG, de Boer JW, van Doesum JA, Noordzij W, van Meerten T, van Dijk LV. Semi-automated 18F-FDG PET segmentation methods for tumor volume determination in Non-Hodgkin lymphoma patients: a literature review, implementation and multi-threshold evaluation. Comput Struct Biotechnol J 2023; 21:1102-1114. [PMID: 36789266 PMCID: PMC9900370 DOI: 10.1016/j.csbj.2023.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
In the treatment of Non-Hodgkin lymphoma (NHL), multiple therapeutic options are available. Improving outcome predictions are essential to optimize treatment. The metabolic active tumor volume (MATV) has shown to be a prognostic factor in NHL. It is usually retrieved using semi-automated thresholding methods based on standardized uptake values (SUV), calculated from 18F-Fluorodeoxyglucose Positron Emission Tomography (18F-FDG PET) images. However, there is currently no consensus method for NHL. The aim of this study was to review literature on different segmentation methods used, and to evaluate selected methods by using an in house created software tool. A software tool, MUltiple SUV Threshold (MUST)-segmenter was developed where tumor locations are identified by placing seed-points on the PET images, followed by subsequent region growing. Based on a literature review, 9 SUV thresholding methods were selected and MATVs were extracted. The MUST-segmenter was utilized in a cohort of 68 patients with NHL. Differences in MATVs were assessed with paired t-tests, and correlations and distributions figures. High variability and significant differences between the MATVs based on different segmentation methods (p < 0.05) were observed in the NHL patients. Median MATVs ranged from 35 to 211 cc. No consensus for determining MATV is available based on the literature. Using the MUST-segmenter with 9 selected SUV thresholding methods, we demonstrated a large and significant variation in MATVs. Identifying the most optimal segmentation method for patients with NHL is essential to further improve predictions of toxicity, response, and treatment outcomes, which can be facilitated by the MUST-segmenter.
Collapse
Key Words
- 18F-FDG PET
- AT, adaptive thresholding methods
- CAR, chimeric antigen receptor
- CT, computed tomography
- DICOM, Digital Imaging and Communications in Medicine
- DLBCL, Diffuse large B-cell lymphoma
- EANM, European Association of Nuclear Medicine
- EARL, EANM Research Ltd.
- FDG, fluorodeoxyglucose
- HL, Hodgkin lymphoma
- IMG, robustness across image reconstruction methods
- IQR, interquartile range
- LBCL, Large B-cell lymphoma
- LDH, lactate dehydrogenase
- MAN, clinician based evaluation using manual segmentations
- MATV, Metabolic active tumor volume
- MIP, Maximum Intensity Projection
- MUST, Multiple SUV Thresholding
- Metabolic tumor volume
- NHL, Non-Hodgkin lymphoma
- Non-Hodgkin lymphoma
- OBS, robustness across observers
- OS, overall survival
- PD-L1, programmed cell death ligand-1
- PET segmentation
- PET, positron emission tomography
- PFS, progression free survival
- PROG, progression vs non-progression
- PTCL, Peripheral T-cell lymphoma
- PTLD, Post-transplant lymphoproliferative disorder
- QS, quality scores
- SOFT, robustness across software
- SUV thresholding
- SUV, standardized uptake value
- Segmentation software
- TCL, T-cell lymphoma
- UMCG, University Medical Center Groningen
- VOI, volume of interest
- cc, cubic centimeter
Collapse
Affiliation(s)
- Kylie Keijzer
- Department of Hematology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands,Department of Radiation Oncology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Anne G.H. Niezink
- Department of Radiation Oncology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Janneke W. de Boer
- Department of Hematology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Jaap A. van Doesum
- Department of Hematology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Tom van Meerten
- Department of Hematology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands
| | - Lisanne V. van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, the Netherlands,Corresponding author.
| |
Collapse
|
36
|
Karimdjee M, Delaby G, Huglo D, Baillet C, Willaume A, Dujardin S, Bailliez A. Evaluation of a convolution neural network for baseline total tumor metabolic volume on [ 18F]FDG PET in diffuse large B cell lymphoma. Eur Radiol 2023; 33:3386-3395. [PMID: 36600126 DOI: 10.1007/s00330-022-09375-1] [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: 07/15/2022] [Revised: 10/20/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
OBJECTIVES New PET data-processing tools allow for automatic lesion selection and segmentation by a convolution neural network using artificial intelligence (AI) to obtain total metabolic tumor volume (TMTV) and total lesion glycolysis (TLG) routinely at the clinical workstation. Our objective was to evaluate an AI implemented in a new version of commercial software to verify reproducibility of results and time savings in a daily workflow. METHODS Using the software to obtain TMTV and TLG, two nuclear physicians applied five methods to retrospectively analyze data for 51 patients. Methods 1 and 2 were fully automated with exclusion of lesions ≤ 0.5 mL and ≤ 0.1 mL, respectively. Methods 3 and 4 were fully automated with physician review. Method 5 was semi-automated and used as reference. Time and number of clicks to complete the measurement were recorded for each method. Inter-instrument and inter-observer variation was assessed by the intra-class coefficient (ICC) and Bland-Altman plots. RESULTS Between methods 3 and 5, for the main user, the ICC was 0.99 for TMTV and 1.0 for TLG. Between the two users applying method 3, ICC was 0.97 for TMTV and 0.99 for TLG. Mean processing time (± standard deviation) was 20 s ± 9.0 for method 1, 178 s ± 125.7 for method 3, and 326 s ± 188.6 for method 5 (p < 0.05). CONCLUSION AI-enabled lesion detection software offers an automated, fast, reliable, and consistently performing tool for obtaining TMTV and TLG in a daily workflow. KEY POINTS • Our study shows that artificial intelligence lesion detection software is an automated, fast, reliable, and consistently performing tool for obtaining total metabolic tumor volume and total lesion glycolysis in a daily workflow.
Collapse
Affiliation(s)
- Mourtaza Karimdjee
- Nuclear Medicine Department, CHU Lille University Hospital, Lille, France.
| | - Gauthier Delaby
- Nuclear Medicine Department, CHU Lille University Hospital, Lille, France
| | - Damien Huglo
- Nuclear Medicine Department, CHU Lille University Hospital, Lille, France
| | - Clio Baillet
- Nuclear Medicine Department, CHU Lille University Hospital, Lille, France
| | - Alexandre Willaume
- Hematology Department, Group of Hospitals of the Catholic Institute of Lille, Lille, France
| | - Simon Dujardin
- Nuclear Medicine Department, CHU Lille University Hospital, Lille, France
| | - Alban Bailliez
- Nuclear Medicine Department, Group of Hospitals of the Catholic Institute of Lille, Lille, France
| |
Collapse
|
37
|
Lue KH, Chen YH, Wu YF, Liu SH. Influence of the methodological aspects of the dichotomization of total metabolic tumor volume measured through baseline fluorine-18 fluorodeoxyglucose PET on survival prediction in lymphoma. Nucl Med Commun 2023; 44:74-80. [PMID: 36514929 DOI: 10.1097/mnm.0000000000001640] [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: 12/15/2022]
Abstract
OBJECTIVE The total metabolic tumor volume (TMTV) measured from fluorine-18 fluorodeoxyglucose (18F-FDG) PET can be useful for determining the prognosis of patients with lymphoma. Stratifying patients into high- and low-TMTV risk groups requires a cutoff point, which is determined through the dichotomization method. This study investigated whether different TMTV dichotomization methods influenced survival prediction in patients with lymphoma. METHODS We retrospectively enrolled 129 patients with lymphoma who had undergone baseline 18F-FDG PET. TMTV was calculated using a fixed standardized uptake value threshold of 4.0. A total of six methods were employed to determine the optimal TMTV cutoff point using receiver-operating characteristic curve analyses, X-Tile bioinformatics software, and the Cutoff Finder web application. The prognostic performance of each method in survival prediction was examined. RESULTS The median (interquartile range) TMTV was 123 cm3 (21-335 cm3). The optimal TMTV cutoff values for predicting progression-free survival (PFS) and overall survival (OS) were in the range of 144-748 cm3. The cutoff points were used to dichotomize patients into two groups with distinct prognoses. All TMTV dichotomizations were significantly predictive of PFS and OS. The survival curves showed significant differences between the high- and low-TMTV groups. The C-indices of the survival models did not significantly differ in any of the dichotomizations. CONCLUSION The prognostic significance of TMTV was maintained regardless of the methodological aspects of dichotomization. However, the optimal TMTV cutoff point varied according to the chosen dichotomization method. Care should be taken when establishing an optimal TMTV cutoff point for clinical use.
Collapse
Affiliation(s)
- Kun-Han Lue
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology
| | - Yu-Hung Chen
- School of Medicine, College of Medicine, Tzu Chi University
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
| | - Yi-Feng Wu
- School of Medicine, College of Medicine, Tzu Chi University
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shu-Hsin Liu
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
| |
Collapse
|
38
|
Zwezerijnen GJC, Eertink JJ, Ferrández MC, Wiegers SE, Burggraaff CN, Lugtenburg PJ, Heymans MW, de Vet HCW, Zijlstra JM, Boellaard R. Reproducibility of [18F]FDG PET/CT liver SUV as reference or normalisation factor. Eur J Nucl Med Mol Imaging 2023; 50:486-493. [PMID: 36166080 PMCID: PMC9816285 DOI: 10.1007/s00259-022-05977-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/15/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Although visual and quantitative assessments of [18F]FDG PET/CT studies typically rely on liver uptake value as a reference or normalisation factor, consensus or consistency in measuring [18F]FDG uptake is lacking. Therefore, we evaluate the variation of several liver standardised uptake value (SUV) measurements in lymphoma [18F]FDG PET/CT studies using different uptake metrics. METHODS PET/CT scans from 34 lymphoma patients were used to calculate SUVmaxliver, SUVpeakliver and SUVmeanliver as a function of (1) volume-of-interest (VOI) size, (2) location, (3) imaging time point and (4) as a function of total metabolic tumour volume (MTV). The impact of reconstruction protocol on liver uptake is studied on 15 baseline lymphoma patient scans. The effect of noise on liver SUV was assessed using full and 25% count images of 15 lymphoma scans. RESULTS Generally, SUVmaxliver and SUVpeakliver were 38% and 16% higher compared to SUVmeanliver. SUVmaxliver and SUVpeakliver increased up to 31% and 15% with VOI size while SUVmeanliver remained unchanged with the lowest variability for the largest VOI size. Liver uptake metrics were not affected by VOI location. Compared to baseline, liver uptake metrics were 15-18% and 9-18% higher at interim and EoT PET, respectively. SUVliver decreased with larger total MTVs. SUVmaxliver and SUVpeakliver were affected by reconstruction protocol up to 62%. SUVmax and SUVpeak moved 22% and 11% upward between full and 25% count images. CONCLUSION SUVmeanliver was most robust against VOI size, location, reconstruction protocol and image noise level, and is thus the most reproducible metric for liver uptake. The commonly recommended 3 cm diameter spherical VOI-based SUVmeanliver values were only slightly more variable than those seen with larger VOI sizes and are sufficient for SUVmeanliver measurements in future studies. TRIAL REGISTRATION EudraCT: 2006-005,174-42, 01-08-2008.
Collapse
Affiliation(s)
- Gerben J C Zwezerijnen
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | - Maria C Ferrández
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Coreline N Burggraaff
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | | | - Martijn W Heymans
- Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Henrica C W de Vet
- Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Huang Z, Guo Y, Zhang N, Huang X, Decazes P, Becker S, Ruan S. Multi-scale feature similarity-based weakly supervised lymphoma segmentation in PET/CT images. Comput Biol Med 2022; 151:106230. [PMID: 36306574 DOI: 10.1016/j.compbiomed.2022.106230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/28/2022] [Accepted: 10/16/2022] [Indexed: 12/27/2022]
Abstract
Accurate lymphoma segmentation in PET/CT images is important for evaluating Diffuse Large B-Cell Lymphoma (DLBCL) prognosis. As systemic multiple lymphomas, DLBCL lesions vary in number and size for different patients, which makes DLBCL labeling labor-intensive and time-consuming. To reduce the reliance on accurately labeled datasets, a weakly supervised deep learning method based on multi-scale feature similarity is proposed for automatic lymphoma segmentation. Weak labeling was performed by randomly dawning a small and salient lymphoma volume for the patient without accurate labels. A 3D V-Net is used as the backbone of the segmentation network and image features extracted in different convolutional layers are fused with the Atrous Spatial Pyramid Pooling (ASPP) module to generate multi-scale feature representations of input images. By imposing multi-scale feature consistency constraints on the predicted tumor regions as well as the labeled tumor regions, weakly labeled data can also be effectively used for network training. The cosine similarity, which has strong generalization, is exploited here to measure feature distances. The proposed method is evaluated with a PET/CT dataset of 147 lymphoma patients. Experimental results show that when using data, half of which have accurate labels and the other half have weak labels, the proposed method performed similarly to a fully supervised segmentation network and achieved an average Dice Similarity Coefficient (DSC) of 71.47%. The proposed method is able to reduce the requirement for expert annotations in deep learning-based lymphoma segmentation.
Collapse
Affiliation(s)
- Zhengshan Huang
- Department of Biomedical Engineering, School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, China
| | - Yu Guo
- Department of Biomedical Engineering, School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, China.
| | - Ning Zhang
- Department of Biomedical Engineering, School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, China
| | - Xian Huang
- Department of Biomedical Engineering, School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, China
| | - Pierre Decazes
- LITIS, University of Rouen Normandy, Rouen, France; Department of Nuclear Medicine, Henri Becquerel Cancer Centre, Rouen, France
| | - Stephanie Becker
- LITIS, University of Rouen Normandy, Rouen, France; Department of Nuclear Medicine, Henri Becquerel Cancer Centre, Rouen, France
| | - Su Ruan
- LITIS, University of Rouen Normandy, Rouen, France
| |
Collapse
|
40
|
Baseline radiomics features and MYC rearrangement status predict progression in aggressive B-cell lymphoma. Blood Adv 2022; 7:214-223. [PMID: 36306337 PMCID: PMC9841040 DOI: 10.1182/bloodadvances.2022008629] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/21/2023] Open
Abstract
We investigated whether the outcome prediction of patients with aggressive B-cell lymphoma can be improved by combining clinical, molecular genotype, and radiomics features. MYC, BCL2, and BCL6 rearrangements were assessed using fluorescence in situ hybridization. Seventeen radiomics features were extracted from the baseline positron emission tomography-computed tomography of 323 patients, which included maximum standardized uptake value (SUVmax), SUVpeak, SUVmean, metabolic tumor volume (MTV), total lesion glycolysis, and 12 dissemination features pertaining to distance, differences in uptake and volume between lesions, respectively. Logistic regression with backward feature selection was used to predict progression after 2 years. The predictive value of (1) International Prognostic Index (IPI); (2) IPI plus MYC; (3) IPI, MYC, and MTV; (4) radiomics; and (5) MYC plus radiomics models were tested using the cross-validated area under the curve (CV-AUC) and positive predictive values (PPVs). IPI yielded a CV-AUC of 0.65 ± 0.07 with a PPV of 29.6%. The IPI plus MYC model yielded a CV-AUC of 0.68 ± 0.08. IPI, MYC, and MTV yielded a CV-AUC of 0.74 ± 0.08. The highest model performance of the radiomics model was observed for MTV combined with the maximum distance between the largest lesion and another lesion, the maximum difference in SUVpeak between 2 lesions, and the sum of distances between all lesions, yielding an improved CV-AUC of 0.77 ± 0.07. The same radiomics features were retained when adding MYC (CV-AUC, 0.77 ± 0.07). PPV was highest for the MYC plus radiomics model (50.0%) and increased by 20% compared with the IPI (29.6%). Adding radiomics features improved model performance and PPV and can, therefore, aid in identifying poor prognosis patients.
Collapse
|
41
|
Metabolic Imaging in B-Cell Lymphomas during CAR-T Cell Therapy. Cancers (Basel) 2022; 14:cancers14194700. [PMID: 36230629 PMCID: PMC9562671 DOI: 10.3390/cancers14194700] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Chimeric antigen receptor–engineered T cells are an innovative therapy in hematologic malignancies, especially in patients with refractory/relapsed B-cell lymphomas. Few studies have analyzed the role of [18F]FDG PET/CT in this field; this review aims to illustrate the literature data and the major findings related to [18F]FDG PET/CT use during CAR-T cell therapy in B-cell lymphomas, focusing on the prognostic value of metabolic parameters, as well as on response assessment. Furthermore, this work shows in detail the specific adverse events during CAR-T cell therapy and the role of [18F]FDG PET/CT imaging in their occurrence. Abstract Chimeric antigen receptor–engineered (CAR) T cells are emerging powerful therapies for patients with refractory/relapsed B-cell lymphomas. [18F]FDG PET/CT plays a key role during staging and response assessment in patients with lymphoma; however, the evidence about its utility in CAR-T therapies for lymphomas is limited. This review article aims to provide an overview of the role of PET/CT during CAR-T cell therapy in B-cell lymphomas, focusing on the prognostic value of metabolic parameters, as well as on response assessment. Data from the literature report on the use of [18F]FDG PET/CT at the baseline with two scans performed before treatment started focused on the time of decision (TD) PET/CT and time of transfusion (TT) PET/CT. Metabolic tumor burden is the most studied parameter associated with disease progression and overall survival, making us able to predict the occurrence of adverse effects. Instead, for post-therapy evaluation, 1 month (M1) PET/CT seems the preferable time slot for response assessment and in this setting, the Deauville 5-point scale (DS), volumetric analyses, SUVmax, and its variation between different time points (∆SUVmax) have been evaluated, confirming the usefulness of M1 PET/CT, especially in the case of pseudoprogression. Additionally, an emerging role of PET/CT brain scans is reported for the evaluation of neurotoxicity related to CAR-T therapies. Overall, PET/CT results to be an accurate method in all phases of CAR-T treatment, with particular interest in assessing treatment response. Moreover, PET parameters have been reported to be reliable predictors of outcome and severe toxicity.
Collapse
|
42
|
Zheng S, Liu T, Li L, Liu Q, Yang L, Zhang Q, Lu X. Tumor-infiltrating lymphocyte signature in epithelial and stromal compartments of an esophageal squamous cell carcinoma acidic microenvironment mediated by MCT4. Pathol Res Pract 2022; 236:153954. [PMID: 35667197 DOI: 10.1016/j.prp.2022.153954] [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: 02/13/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022]
Abstract
Tumor-infiltrating lymphocytes (TILs), including but not limited to neutrophils, M2 macrophages, cytotoxic CD8 T cells and dendritic cells, will play a role in the acidic tumor microenvironment mediated by monocarboxylate transporter 4 (MCT4) in esophageal squamous cell carcinoma (ESCC). However, the roles they play and their significance in ESCC remain less clear. To understand the clinicopathological and prognostic significance of neutrophils, M2 macrophages, CD8 T cells and dendritic cells in the tumor acidic microenvironment mediated by MCT4, we investigated the distribution of these TILs in the epithelial and stromal compartments of ESCC by means of multiplexed immunohistochemistry on a tissue microarray containing 87 paired dots of ESCC and its adjacent normal tissue (ANT) and an additional 6 cases of unpaired ESCC dots. The density of cells stained with MCT4 in the epithelium was significantly associated with overall survival. Dendritic cells stained with S100 in epithelial compartmentalization were found to markedly correlate with clinical stage and tumor invasion depth. No other significant association could be identified in terms of prognostic and clinicopathological significance. The potential correlation between the number of cells stained with MCT4 versus the number of TILs was also explored, showing that only in epithelial cells were there significant and positive correlations identified between the number of cells stained with MCT4 versus the number of neutrophils stained with CD15, M2 macrophages stained with CD163 and CD8 T cells stained by CD8a. However, no significant correlation was found along the stromal line. Together, the data we described here, although somewhat discouraging, showed that in epithelial cells from which ESCC originated, acidicity mediated by MCT4 may be responsible for lactate release and may have an effect on the infiltration of TILs we assessed.
Collapse
Affiliation(s)
- Shutao Zheng
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Tao Liu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China; Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Lu Li
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Qing Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Lifei Yang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Qiqi Zhang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Xiaomei Lu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China.
| |
Collapse
|
43
|
Ferrández MC, Eertink JJ, Golla SSV, Wiegers SE, Zwezerijnen GJC, Pieplenbosch S, Zijlstra JM, Boellaard R. Combatting the effect of image reconstruction settings on lymphoma [ 18F]FDG PET metabolic tumor volume assessment using various segmentation methods. EJNMMI Res 2022; 12:44. [PMID: 35904645 PMCID: PMC9338209 DOI: 10.1186/s13550-022-00916-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
Background [18F]FDG PET-based metabolic tumor volume (MTV) is a promising prognostic marker for lymphoma patients. The aim of this study is to assess the sensitivity of several MTV segmentation methods to variations in image reconstruction methods and the ability of ComBat to improve MTV reproducibility. Methods Fifty-six lesions were segmented from baseline [18F]FDG PET scans of 19 lymphoma patients. For each scan, EARL1 and EARL2 standards and locally clinically preferred reconstruction protocols were applied. Lesions were delineated using 9 semiautomatic segmentation methods: fixed threshold based on standardized uptake value (SUV), (SUV = 4, SUV = 2.5), relative threshold (41% of SUVmax [41M], 50% of SUVpeak [A50P]), majority vote-based methods that select voxels detected by at least 2 (MV2) and 3 (MV3) out of the latter 4 methods, Nestle thresholding, and methods that identify the optimal method based on SUVmax (L2A, L2B). MTVs from EARL2 and locally clinically preferred reconstructions were compared to those from EARL1. Finally, different versions of ComBat were explored to harmonize the data.
Results MTVs from the SUV4.0 method were least sensitive to the use of different reconstructions (MTV ratio: median = 1.01, interquartile range = [0.96–1.10]). After ComBat harmonization, an improved agreement of MTVs among different reconstructions was found for most segmentation methods. The regular implementation of ComBat (‘Regular ComBat’) using non-transformed distributions resulted in less accurate and precise MTV alignments than a version using log-transformed datasets (‘Log-transformed ComBat’). Conclusion MTV depends on both segmentation method and reconstruction methods. ComBat reduces reconstruction dependent MTV variability, especially when log-transformation is used to account for the non-normal distribution of MTVs. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00916-9.
Collapse
Affiliation(s)
- Maria C Ferrández
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Sandeep S V Golla
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Mikhaeel NG, Heymans MW, Eertink JJ, de Vet HC, Boellaard R, Dührsen U, Ceriani L, Schmitz C, Wiegers SE, Hüttmann A, Lugtenburg PJ, Zucca E, Zwezerijnen GJ, Hoekstra OS, Zijlstra JM, Barrington SF. Proposed New Dynamic Prognostic Index for Diffuse Large B-Cell Lymphoma: International Metabolic Prognostic Index. J Clin Oncol 2022; 40:2352-2360. [PMID: 35357901 PMCID: PMC9287279 DOI: 10.1200/jco.21.02063] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/23/2022] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Baseline metabolic tumor volume (MTV) is a promising biomarker in diffuse large B-cell lymphoma (DLBCL). Our aims were to determine the best statistical relationship between MTV and survival and to compare MTV with the International Prognostic Index (IPI) and its individual components to derive the best prognostic model. METHODS PET scans and clinical data were included from five published studies in newly diagnosed diffuse large B-cell lymphoma. Transformations of MTV were compared with the primary end points of 3-year progression-free survival (PFS) and overall survival (OS) to derive the best relationship for further analyses. MTV was compared with IPI categories and individual components to derive the best model. Patients were grouped into three groups for survival analysis using Kaplan-Meier analysis; 10% at highest risk, 30% intermediate risk, and 60% lowest risk, corresponding with expected clinical outcome. Validation of the best model was performed using four studies as a test set and the fifth study for validation and repeated five times. RESULTS The best relationship for MTV and survival was a linear spline model with one knot located at the median MTV value of 307.9 cm3. MTV was a better predictor than IPI for PFS and OS. The best model combined MTV with age as continuous variables and individual stage as I-IV. The MTV-age-stage model performed better than IPI and was also better at defining a high-risk group (3-year PFS 46.3% v 58.0% and 3-year OS 51.5% v 66.4% for the new model and IPI, respectively). A regression formula was derived to estimate individual patient survival probabilities. CONCLUSION A new prognostic index is proposed using MTV, age, and stage, which outperforms IPI and enables individualized estimates of patient outcome.
Collapse
Affiliation(s)
- N. George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London University, London, United Kingdom
| | - Martijn W. Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Jakoba J. Eertink
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Henrica C.W. de Vet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Ronald Boellaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Luca Ceriani
- Department of Oncology, IOSI—Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK—Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Christine Schmitz
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sanne E. Wiegers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pieternella J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Emanuele Zucca
- Department of Oncology, IOSI—Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK—Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Gerben J.C. Zwezerijnen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Otto S. Hoekstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Josée M. Zijlstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sally F. Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, Kings College London, London, United Kingdom
| |
Collapse
|
45
|
Mikhaeel NG, Heymans MW, Eertink JJ, de Vet HCW, Boellaard R, Dührsen U, Ceriani L, Schmitz C, Wiegers SE, Hüttmann A, Lugtenburg PJ, Zucca E, Zwezerijnen GJC, Hoekstra OS, Zijlstra JM, Barrington SF. Proposed New Dynamic Prognostic Index for Diffuse Large B-Cell Lymphoma: International Metabolic Prognostic Index. J Clin Oncol 2022; 40:2352-2360. [PMID: 35357901 DOI: 10.1200/jco.21.02063:jco2102063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023] Open
Abstract
PURPOSE Baseline metabolic tumor volume (MTV) is a promising biomarker in diffuse large B-cell lymphoma (DLBCL). Our aims were to determine the best statistical relationship between MTV and survival and to compare MTV with the International Prognostic Index (IPI) and its individual components to derive the best prognostic model. METHODS PET scans and clinical data were included from five published studies in newly diagnosed diffuse large B-cell lymphoma. Transformations of MTV were compared with the primary end points of 3-year progression-free survival (PFS) and overall survival (OS) to derive the best relationship for further analyses. MTV was compared with IPI categories and individual components to derive the best model. Patients were grouped into three groups for survival analysis using Kaplan-Meier analysis; 10% at highest risk, 30% intermediate risk, and 60% lowest risk, corresponding with expected clinical outcome. Validation of the best model was performed using four studies as a test set and the fifth study for validation and repeated five times. RESULTS The best relationship for MTV and survival was a linear spline model with one knot located at the median MTV value of 307.9 cm3. MTV was a better predictor than IPI for PFS and OS. The best model combined MTV with age as continuous variables and individual stage as I-IV. The MTV-age-stage model performed better than IPI and was also better at defining a high-risk group (3-year PFS 46.3% v 58.0% and 3-year OS 51.5% v 66.4% for the new model and IPI, respectively). A regression formula was derived to estimate individual patient survival probabilities. CONCLUSION A new prognostic index is proposed using MTV, age, and stage, which outperforms IPI and enables individualized estimates of patient outcome.
Collapse
Affiliation(s)
- N George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London University, London, United Kingdom
| | - Martijn W Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Jakoba J Eertink
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Henrica C W de Vet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Ronald Boellaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Luca Ceriani
- Department of Oncology, IOSI-Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK-Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Christine Schmitz
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sanne E Wiegers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Emanuele Zucca
- Department of Oncology, IOSI-Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK-Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Gerben J C Zwezerijnen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Otto S Hoekstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Josée M Zijlstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, Kings College London, London, United Kingdom
| |
Collapse
|
46
|
Guzmán Ortiz S, Mucientes Rasilla J, Vargas Núñez J, Royuela A, Rodríguez Carrillo J, Dotor de Lama A, Navarro Matilla M, Mitjavila Casanovas M. Evaluación del valor pronóstico de los parámetros volumétricos metabólicos calculados con la 18F-FDG PET/TC y su valor añadido a las características moleculares en pacientes con linfoma B difuso de células grandes. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Kostakoglu L, Mattiello F, Martelli M, Sehn LH, Belada D, Ghiggi C, Chua N, González-Barca E, Hong X, Pinto A, Shi Y, Tatsumi Y, Bolen C, Knapp A, Sellam G, Nielsen T, Sahin D, Vitolo U, Trněný M. Total metabolic tumor volume as a survival predictor for patients with diffuse large B-cell lymphoma in the GOYA study. Haematologica 2022; 107:1633-1642. [PMID: 34407602 PMCID: PMC9244811 DOI: 10.3324/haematol.2021.278663] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/04/2021] [Indexed: 11/14/2022] Open
Abstract
This retrospective analysis of the phase III GOYA study investigated the prognostic value of baseline metabolic tumor volume parameters and maximum standardized uptake values for overall and progression-free survival (PFS) in treatment-naïve diffuse large B-cell lymphoma. Baseline total metabolic tumor volume (determined for tumors >1 mL using a threshold of 1.5 times the mean liver standardized uptake value +2 standard deviations), total lesion glycolysis, and maximum standardized uptake value positron emission tomography data were dichotomized based on receiver operating characteristic analysis and divided into quartiles by baseline population distribution. Of 1,418 enrolled patients, 1,305 had a baseline positron emission tomography scan with detectable lesions. Optimal cut-offs were 366 cm3 for total metabolic tumor volume and 3,004 g for total lesion glycolysis. High total metabolic tumor volume and total lesion glycolysis predicted poorer PFS, with associations retained after adjustment for baseline and disease characteristics (high total metabolic tumor volume hazard ratio: 1.71, 95% confidence interval [CI]: 1.35- 2.18; total lesion glycolysis hazard ratio: 1.46; 95% CI: 1.15-1.86). Total metabolic tumor volume was prognostic for PFS in subgroups with International Prognostic Index scores 0-2 and 3-5, and those with different cell-of-origin subtypes. Maximum standardized uptake value had no prognostic value in this setting. High total metabolic tumor volume associated with high International Prognostic Index or non-germinal center B-cell classification identified the highest-risk cohort for unfavorable prognosis. In conclusion, baseline total metabolic tumor volume and total lesion glycolysis are independent predictors of PFS in patients with diffuse large B-cell lymphoma after first-line immunochemotherapy.
Collapse
Affiliation(s)
- Lale Kostakoglu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA.
| | | | - Maurizio Martelli
- Department of translational and precision medicine, Sapienza University, Rome
| | - Laurie H Sehn
- BC Cancer Centre for Lymphoid Cancer and the University of British Columbia, Vancouver, BC
| | - David Belada
- 4th Department of Internal Medicine-Hematology, Charles University, Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | | | - Neil Chua
- Cross Cancer Institute, University of Alberta, Edmonton, AB
| | - Eva González-Barca
- Institut Català d'Oncologia, Institut d'Investigació Biomédica de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat, Barcelona
| | - Xiaonan Hong
- Fudan University Shanghai Cancer Center, Shanghai
| | - Antonio Pinto
- Hematology-Oncology, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Naples
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yoichi Tatsumi
- Department of Patient Safety and Management, Kindai University Hospital and Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka
| | | | | | | | | | | | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin
| | - Marek Trněný
- First Department of Medicine, Charles University General Hospital, Prague, Czech Republic
| |
Collapse
|
48
|
Huang L, Ruan S, Decazes P, Denœux T. Lymphoma segmentation from 3D PET-CT images using a deep evidential network. Int J Approx Reason 2022. [DOI: 10.1016/j.ijar.2022.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Morland D, Triumbari EKA, Maiolo E, Cuccaro A, Treglia G, Hohaus S, Annunziata S. Healthy Organs Uptake on Baseline 18F-FDG PET/CT as an Alternative to Total Metabolic Tumor Volume to Predict Event-Free Survival in Classical Hodgkin's Lymphoma. Front Med (Lausanne) 2022; 9:913866. [PMID: 35814740 PMCID: PMC9256984 DOI: 10.3389/fmed.2022.913866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeHealthy organs uptake, including cerebellar and liver SUVs have been reported to be inversely correlated to total metabolic tumor volume (TMTV), a controversial predictor of event-free survival (EFS) in classical Hodgkin's Lymphoma (cHL). The objective of this study was to estimate TMTV by using healthy organs SUV measurements and assess the performance of this new index (UF, Uptake Formula) to predict EFS in cHL.MethodsPatients with cHL were retrospectively included. SUV values and TMTV derived from baseline 18F-FDG PET/CT were harmonized using ComBat algorithm across PET/CT systems. UF was estimated using ANOVA analysis. Optimal thresholds of TMTV and UF were calculated and tested using Cox models.Results163 patients were included. Optimal UF model of TMTV included age, lymphoma maximum SUVmax, hepatic SUVmean and cerebellar SUVmax (R2 14.0% - p < 0.001). UF > 236.8 was a significant predictor of EFS (HR: 2.458 [1.201–5.030], p = 0.01) and was not significantly different from TMTV > 271.0 (HR: 2.761 [1.183–5.140], p = 0.001). UF > 236.8 remained significant in a bivariate model including IPS score (p = 0.02) and determined two populations with different EFS (63.7 vs. 84.9%, p = 0.01).ConclusionThe Uptake Formula, a new index including healthy organ SUV values, shows similar performance to TMTV in predicting EFS in Hodgkin's Lymphoma. Validation cohorts will be needed to confirm this new prognostic parameter.
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, Roma, Italy
- Service de Médecine Nucléaire, Institut Godinot, Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l'Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, Reims, France
- *Correspondence: David Morland
| | - Elizabeth Katherine Anna Triumbari
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Elena Maiolo
- Unità di Ematologia, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Annarosa Cuccaro
- Unità di Ematologia, ASL Toscana N/O Spedali Riuniti Livorno, Livorno, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Stefan Hohaus
- Unità di Ematologia, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Section of Hematology, Department of Radiological Sciences, Radiotherapy and Hematology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| |
Collapse
|
50
|
Yhim H, Eshet Y, Metser U, Lajkosz K, Cooper M, Prica A, Kukreti V, Bhella S, Lang N, Xu W, Rodin D, Hodgson D, Tsang R, Crump M, Kuruvilla J, Kridel R. Risk stratification for relapsed/refractory classical Hodgkin lymphoma integrating pretransplant Deauville score and residual metabolic tumor volume. Am J Hematol 2022; 97:583-591. [PMID: 35170780 DOI: 10.1002/ajh.26500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/31/2021] [Accepted: 02/07/2022] [Indexed: 11/09/2022]
Abstract
Pretransplant Deauville score (DS) is an imaging biomarker used for risk stratification in relapsed/refractory classical Hodgkin lymphoma (cHL). However, the prognostic value of residual metabolic tumor volume (rMTV) in patients with DS 4-5 has been less well characterized. We retrospectively assessed 106 patients with relapsed/refractory cHL who underwent autologous stem cell transplantation. Pretransplant DS was determined as 1-3 (59%) and 4-5 (41%), with a markedly inferior event-free survival (EFS) in patients with DS 4-5 (hazard ratio [HR], 3.14; p = .002). High rMTV41% (rMTVhigh , ≥4.4 cm3 ) predicted significantly poorer EFS in patients with DS 4-5 (HR, 3.70; p = .014). In a multivariable analysis, we identified two independent factors predicting treatment failure: pretransplant DS combined with rMTV41% and disease status (primary refractory vs. relapsed). These two factors allow to stratify patients into three groups with divergent 2-year EFS: 89% for low-risk (51%; relapsed disease and either pretransplant DS 1-3 or DS 4-5/rMTVlow ; HR 1), 65% for intermediate-risk (28%; refractory disease and either DS 1-3 or DS 4-5/rMTVlow ; HR 3.26), and 45% for high-risk (21%; DS 4-5/rMTVhigh irrespective of disease status; HR 7.61) groups. Pretransplant DS/rMTV41% combination and disease status predict the risk of post-transplant treatment failure and will guide risk-stratified approaches in relapsed/refractory cHL.
Collapse
Affiliation(s)
- Ho‐Young Yhim
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
- Department of Internal Medicine Jeonbuk National University Medical School and Research Institute of Clinical Medicine of Jeonbuk National University‐Biomedical Research Institute of Jeonbuk National University Hospital Jeonju Republic of Korea
| | - Yael Eshet
- Joint Department of Medical Imaging, Princess Margaret Cancer Centre University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto Toronto Ontario Canada
| | - Ur Metser
- Joint Department of Medical Imaging, Princess Margaret Cancer Centre University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto Toronto Ontario Canada
| | - Katherine Lajkosz
- Department of Biostatistics, Princess Margaret Cancer Centre, Dalla Lana School of Public Health University of Toronto Toronto Ontario Canada
| | - Matthew Cooper
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
- Faculty of Medicine Dalhousie University Halifax Nova Scotia Canada
| | - Anca Prica
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - Vishal Kukreti
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - Sita Bhella
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - Noémie Lang
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, Dalla Lana School of Public Health University of Toronto Toronto Ontario Canada
| | - Danielle Rodin
- Radiation Medicine Program Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
- Department of Radiation Oncology University of Toronto Toronto Ontario Canada
| | - David Hodgson
- Radiation Medicine Program Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
- Department of Radiation Oncology University of Toronto Toronto Ontario Canada
| | - Richard Tsang
- Radiation Medicine Program Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
- Department of Radiation Oncology University of Toronto Toronto Ontario Canada
| | - Michael Crump
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - John Kuruvilla
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| | - Robert Kridel
- Division of Medical Oncology and Hematology Princess Margaret Cancer Centre – University Health Network Toronto Ontario Canada
| |
Collapse
|