1
|
Yang Y, Yu J, Hu X, Chen S, Zhao R, Huang C, Guo J, Tang T, Chen C, Lin Y, Wang Y, Liu T, Zheng H, Liao S, Chen J, Fu H, Liu T. Phase II Trial of Hypofractionated Radiotherapy and Immunochemotherapy in Primary Refractory Diffuse Large B-Cell Lymphoma: Preliminary Results and Insights from Digital Spatial Profiling. MedComm (Beijing) 2025; 6:e70225. [PMID: 40416596 PMCID: PMC12103654 DOI: 10.1002/mco2.70225] [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: 10/06/2024] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
This open-label, single-arm phase II study assessed the safety and efficacy of sequential hypofractionated radiotherapy (RT) followed by zimberelimab and R-GemOx (rituximab, gemcitabine, oxaliplatin) in patients with primary refractory diffuse large B-cell lymphoma (DLBCL). Fourteen patients were enrolled between June 2022 and December 2023, with 13 included in the analysis. RT doses of 36 and 24 Gy were delivered to the gross and target volumes in 12 fractions, followed by zimberelimab and R-GemOx. The overall response rate within the irradiated field was 92.3%, and a complete response (CR) was achieved by 61.5% of patients; however, 38.5% experienced disease progression. Treatment-related toxicities were manageable, primarily comprising mild leukocytopenia. Digital spatial profiling revealed 53 differentially expressed genes in CD20-rich lymphoma regions and 93 in CD3-rich T cell regions in non-CR patients. Reactome analysis identified key immune system pathways. T cell infiltration correlated with treatment efficacy, and multiplex immunohistochemistry validated immune pathways as potential therapeutic targets. This study demonstrated the promising role of RT combined with immunochemotherapy in refractory DLBCL and suggests immune pathways as critical targets to improve treatment outcomes.
Collapse
Affiliation(s)
- Yong Yang
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Jing Yu
- Department of Pulmonary OncologyHubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiao‐Mei Hu
- Department of PathologyFujian Medical University Union HospitalFuzhouChina
| | - Si‐Lin Chen
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Rui‐Zhi Zhao
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Cheng Huang
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Jiang‐Rui Guo
- Department of HematologyFujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on HematologyFuzhouChina
| | - Tian‐Lan Tang
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Cheng Chen
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Yu‐Ping Lin
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Ying Wang
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Tian‐Xiu Liu
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Hao Zheng
- Department of Radiation OncologyFujian Medical University Union Hospital, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies)FuzhouChina
| | - Si‐Qin Liao
- Department of PET/CTFujian Medical University Union HospitalFuzhouChina
| | - Jin‐Hua Chen
- Follow‐Up CenterFujian Medical University Union HospitalFuzhouChina
| | - Hai‐Ying Fu
- Department of HematologyThe Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, The Third People's Hospital of Fujian ProvinceFuzhouChina
| | - Ting‐Bo Liu
- Department of HematologyFujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on HematologyFuzhouChina
| |
Collapse
|
2
|
Ren Y, Tan H, Zhuang J, Cheng L, Yuan L, Ji L, Ke Y, Zhang X, Cheng Z, Li J, Liu P. Polatuzumab Vedotin, zanubrutinib and rituximab (Pola-ZR) achieved rapid and deep response in untreated frail and elderly DLBCL. Ann Hematol 2025:10.1007/s00277-025-06412-z. [PMID: 40377673 DOI: 10.1007/s00277-025-06412-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 05/12/2025] [Indexed: 05/18/2025]
Abstract
First-line treatment balancing efficacy and safety is urgently needed for frail and elderly diffuse large B-cell lymphoma (DLBCL) patients. We designed a triplet chemo-light regimen, Pola-ZR, in previously untreated frail and elderly DLBCL patients to assess the efficacy and safety in a prospective DLBCL cohort (NCT06203652). Polatuzumab vedotin was given 1.8 mg/KG intravenously on day 1, zanubrutinib 160 mg twice a day orally from day 1 to day 21, and rituximab 375 mg/m2 intravenously on day 1. Twenty-one days were a cycle. If assessed complete response (CR) after 6 cycles, patients would receive zanubrutinib alone for another 6 cycles. PET/CT or contrast-enhanced CT scan was scheduled every 3 cycles. The primary end point was overall response rate (ORR) after 6 cycles. Twenty-four patients were enrolled from 01 Apr 2023 to 20 Dec 2023. Median age was 73. Sixteen (66.7%) patients had an international prognostic index score of 3 to 5. After a median follow-up of 10.2 months, the CR rate and ORR after 6 cycles was 83% and 83%. Non-responders had high total metabolic tumour volume. Lung infection was the major safety concern. PJP prophylaxis was recommended. Pola-ZR regimen showed rapid and deep response with manageable safety profiles in both GCB and non-GCB subtypes.
Collapse
Affiliation(s)
- Yuhong Ren
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingli Zhuang
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Luya Cheng
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Ling Yuan
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Lili Ji
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yang Ke
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Xuejiao Zhang
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Zhixiang Cheng
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Jing Li
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| |
Collapse
|
3
|
Durmo R, Chauvie S, Minoia C, Bergesio F, Fallanca F, Peano S, Marcheselli L, Anastasia A, Boccomini C, Corradini P, Olivieri J, Arcaini L, Cavallo F, Ibatici A, Nassi L, Tarantino V, Pinto A, Stelitano C, Pulsoni A, Ricci F, Mancuso S, Cencini E, Di Renzo N, Mannarella C, Palmas A, Zinzani P, Bocci C, Rossi F, Carella AM, Federico M, Versari A, Guerra L, Luminari S. Total Metabolic Tumor Volume Is a Strong Independent Prognostic Factor in Follicular Lymphomas: Results From a Sub-Study of the FOLL12 Trial. Am J Hematol 2025. [PMID: 40366076 DOI: 10.1002/ajh.27711] [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: 12/09/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 05/15/2025]
Abstract
Discordant results have been generated regarding the prognostic role of Total Metabolic Tumor Volume (TMTV) in Follicular Lymphoma (FL). The use of prospective data and the adoption of the newly defined standardized SUV4 method for calculating TMTV may generate stronger evidence. We conducted a pre-planned post hoc analysis of the prospective multicenter randomized phase III FOLL12 trial for newly diagnosed high tumor burden FL (grade 1-3a), which mandated baseline staging with PET. Baseline PET/CT scans were reviewed centrally, and TMTV was calculated using the fixed threshold of SUV4. Kaplan-Meier and Cox regression were used for survival analysis. The primary study endpoint was Progression free Survival (PFS). A total of 689 FL patients were available for TMTV definition. Median TMTV was 161 mL (IQR 50 to 388 mL) and the best cutoff value was set at 180 mL. Patients with high TMTV had a significantly lower 5-year PFS compared to those with low TMTV: 59% (95% CI, 53-65%) vs. 74% (95% CI, 69-78%) HR 1.61 (95% CI, 1.24-2.09). Prognostic role of TMTV was independent of study arm, chemotherapy regimen, and FLIPI2. Combined with FLIPI-2, we identified three groups with different 5-yr PFS rates, with the lowest rates (51%) for patients with high TMTV and high FLIPI2. Combined TMTV and FLIPI model was also prognostic to predict the risk of early progression and of death. Applying the SUV4 standard method pre-treatment TMTV is confirmed as a strong and independent predictor of PFS in FL patients. Integrating TMTV with FLIPI-2 improves risk assessment.
Collapse
Affiliation(s)
- Rexhep Durmo
- Nuclear Medicine, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | | | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II' Bari, Bari, Italy
| | | | - Federico Fallanca
- IRCCS San Raffaele Scientific Institute, Nuclear Medicine, Milan, Italy
| | - Simona Peano
- Nuclear Medicine Division, S. Croce e Carle Hospital, Cuneo, Italy
| | | | | | - Carola Boccomini
- SC Ematologia, Dipartimento di Ematologia Ed Oncologia, AOU Città Della Salute e Della Scienza di Torino, Torino, Italy
| | | | | | - Luca Arcaini
- Department of Molecular Medicine, University of Pavia & Division of Hematology, Fondazione IRCCS Policlinico san Matteo, Pavia, Italy
| | - Federica Cavallo
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute Università di Torino, Torino, Italy
| | - Adalberto Ibatici
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Nassi
- Ematologia, Az Ospedaliera Careggi, Firenze, Italy
| | | | - Antonello Pinto
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, IRCCS-Fondazione 'G. Pascale', Naples, Italy
| | - Caterina Stelitano
- Hematology, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | | | | | | | - Emanuele Cencini
- UOC Ematologia, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | | | | | - Angelo Palmas
- Ematologia e Trapianto di Midollo Osseo Ospedale 'San Francesco' Nuoro, Matera, Italy
| | - Pierluigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Seràgnoli, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Caterina Bocci
- U.O.S.D Ematologia Con Autotrapianto Cellule Staminali Ospedale di Civitanova Marche, Civitanova Marche, Italy
| | | | | | - Massimo Federico
- Department Chimomo, Università di Modena and Reggio Emilia, Modena, Italy
| | - Annibale Versari
- Nuclear Medicine, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Luca Guerra
- Nuclear Medicine Unit, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- University of Milano Bicocca, Milan, Italy
| | - Stefano Luminari
- Department Chimomo, Università di Modena and Reggio Emilia, Modena, Italy
- Hematology, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
4
|
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
|
5
|
Cairns J, Frood R, Patel C, Scarsbrook A. The Role of AI in Lymphoma: An Update. Semin Nucl Med 2025; 55:377-386. [PMID: 40069036 DOI: 10.1053/j.semnuclmed.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 04/18/2025]
Abstract
Malignant lymphomas encompass a range of malignancies with incidence rising globally, particularly with age. In younger populations, Hodgkin and Burkitt lymphomas predominate, while older populations more commonly experience subtypes such as diffuse large B-cell, follicular, marginal zone, and mantle cell lymphomas. Positron emission tomography/computed tomography (PET/CT) using [18F] fluorodeoxyglucose (FDG) is the gold standard for staging, treatment response assessment, and prognostication in lymphoma. However, interpretation of PET/CT is complex, time-consuming, and reliant on expert imaging specialists, exacerbating challenges associated with workforce shortages worldwide. Artificial intelligence (AI) offers transformative potential across multiple aspects of PET/CT imaging in this setting. AI applications in appointment planning have demonstrated utility in reducing nonattendance rates and improving departmental efficiency. Advanced reconstruction techniques leveraging convolutional neural networks (CNNs) enable reduced injected activities of radiopharmaceutical and patient dose whilst maintaining diagnostic accuracy, particularly benefiting younger patients requiring multiple scans. Automated segmentation tools, predominantly using 3D U-Net architectures, have improved quantification of metrics such as total metabolic tumour volume (TMTV) and total lesion glycolysis (TLG), facilitating prognostication and treatment stratification. Despite these advancements, challenges remain, including variability in segmentation performance, impact on Deauville Score interpretation, and standardization of TMTV/TLG measurements. Emerging large language models (LLMs) also show promise in enhancing PET/CT reporting, converting free-text reports into structured formats, and improving patient communication. Further research is required to address limitations such as AI-induced errors, physiological uptake differentiation, and the integration of AI models into clinical workflows. With robust validation and harmonization, AI integration could significantly enhance lymphoma care, improving diagnostic precision, workflow efficiency, and patient outcomes.
Collapse
Affiliation(s)
- James Cairns
- Faculty of Medicine, University of Leeds, Leeds LS2 9JT, England; Department of Radiology, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, LS9 7TF, England
| | - Russell Frood
- Faculty of Medicine, University of Leeds, Leeds LS2 9JT, England; Department of Radiology, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, LS9 7TF, England
| | - Chirag Patel
- Department of Radiology, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, LS9 7TF, England
| | - Andrew Scarsbrook
- Faculty of Medicine, University of Leeds, Leeds LS2 9JT, England; Department of Radiology, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, LS9 7TF, England.
| |
Collapse
|
6
|
Ceriani L, Milan L, Chauvie S, Zucca E. Understandings 18 FDG PET radiomics and its application to lymphoma. Br J Haematol 2025. [PMID: 40230306 DOI: 10.1111/bjh.20074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
The early identification of lymphoma patients who fail front-line treatment is crucial for optimizing disease management. Positron emission tomography, a well-established tool for staging and response evaluation in lymphoma, is typically assessed visually or semiquantitatively, leaving much of its latent information unexploited. Radiomic analysis, which employs mathematical descriptors, can enable the extraction of quantitative features from baseline images that correlate with the disease's biological characteristics. Emerging radiomic features such as metabolic tumour volume, total lesion glycolysis and markers of disease dissemination and metabolic heterogeneity are proving to be powerful prognostic biomarkers in lymphoma. Texture analysis, the most advanced area of radiomics, offers highly complex features that require further standardization and validation before being adopted as reliable biomarkers. Combining radiomic features with clinical risk factors and genomic data holds promising potential for improving clinical risk prediction. This review explores the current state of radiomic analysis, progress towards its standardization and its incorporation into clinical practice and trial designs. The integration of radiomic markers with circulating tumour DNA may provide a comprehensive approach to developing baseline and dynamic risk scores, facilitating the testing of novel treatments and advancing personalized treatment of aggressive lymphomas.
Collapse
Affiliation(s)
- Luca Ceriani
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Lisa Milan
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Stephane Chauvie
- Medical Physics Division, Santa Croce e Carlo Hospital, Cuneo, Italy
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Haematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Medical Oncology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
Cui Y, Li Y, Hu W, Wu Z, Li S, Wang H. Evaluating ΔMTV%, ΔD max%, and %ΔSUV max of 18F-FDG PET/CT for mid-treatment efficacy and prognosis in diffuse large B-cell lymphoma. Discov Oncol 2025; 16:411. [PMID: 40146454 PMCID: PMC11950622 DOI: 10.1007/s12672-025-02126-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
PURPOSE To investigate the value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) imaging in interim therapeutic and prognostic evaluation of patients with diffuse large B-cell lymphoma (DLBCL). MATERIALS AND METHODS Data of 86 patients with pathologically confirmed DLBCL who underwent 18F-FDG PET/CT imaging before chemotherapy, radiotherapy, and after interim chemotherapy, were retrospectively analyzed. Receive operating characteristic (ROC) curve analysis was performed to assess the predictive capacity of changes and change rates in PET/CT imaging parameters [maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), total lesion glycolysis (TLG), and maximum tumor dissemination (Dmax)] for progression-free survival (PFS) and to identify optimal cutoff values. Kaplan-Meier survival curves were constructed, and the log-rank test was used to assess intergroup differences. Cox regression analysis was used to explore potential factors influencing PFS. RESULTS Among 86 patients [(45 men, 41 women, age: 57.8 ± 12.2 years)], the median PFS was 22.5 (14.5, 46) months. Until the last follow-up date, progression or recurrence occurred in 14 patients, while 9 patients died. The ROC curves indicated that the optimal cutoff values for predicting PFS were 99.10%, 99.72%, and 96.47% for ΔMTV%, ΔTLG%, and ΔDmax%, respectively (area under the curve = 0.786-0.849, all P < 0.05). Cox univariate analysis demonstrated that the alteration rates in metabolic and diffusion parameters before and after treatment, including SUVmax%, MTV%, TLG%, and Dmax%, were predictive of PFS (hazard ratio [HR] = 6.213-13.430, all P < 0.05). The Cox multivariate analysis demonstrated that ΔMTV% and ΔDmax% independently predicted PFS, with HRs of 10.727 (95% confidence interval [CI] = 1.928-56.672, P = 0.007) and 7.178 (95%CI = 1.514-34.041, P = 0.013), respectively. We established a new prediction model by combining the ΔMTV% and ΔDmax% parameters, and the results of the model showed statistically significant differences in PFS between the high, intermediate, and low-risk groups. The model predicted higher effects than individual indicators. CONCLUSION The rate of change in metabolic and diffusion parameters on interim PET/CT can predict the prognosis of patients with DLBCL.
Collapse
Affiliation(s)
- Yali Cui
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Yao Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Wenhao Hu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
- Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
8
|
Malmon S, Elsensohn MH, Thieblemont C, Morschhauser F, Casasnovas O, André M, Gouill SL, Tabaa YA, Durand PB, Bailly C, Edeline V, Vija L, Vercellino L, Ricci R, Kanoun S, Cottereau AS. Prognostic impact of metabolic tumor volume using the SUV4.0 segmentation threshold in 1,960 lymphoma patients from prospective LYSA trials. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07176-4. [PMID: 40108044 DOI: 10.1007/s00259-025-07176-4] [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: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE This study compared the prognostic value of total metabolic tumor volume (TMTV) in lymphoma measured with the recently proposed SUV4.0 segmentation threshold versus the 41% SUVmax across LYSA trials and its impact on intensity and dissemination PET features. METHODS A total of 1960 baseline PET/CT scans of Diffuse Large B cell lymphoma (DLBCL), follicular lymphoma (FL) and Hodgkin lymphoma (HL) patients were collected. After a semi-automatic preselection of region of interest, two different segmentation threshold were applied: 41% SUVmax (TMTV41%) and SUV > 4.0 (TMTV4.0). RESULTS The correlation between TMTV4.0 and TMTV41% was ρ = 0.90 for DLBCL, ρ = 0.65 for FL and ρ = 0.60 for HL. For SUVmax, SUVpeak, Dmax and Dbulk features, a strong correlation was observed with ρ > 0.95 whatever the lymphoma subtypes. The predictability of TMTV was high and comparable for the two methods with superimposable confidence intervals for the three subtypes. At the 90th percentile TMTV value, the predicted 7-year PFS was 51.13% with TMTV4.0 vs. 49.7% with TMTV41% for DLBCL patients, 45.5% vs. 39.8% for FL patients, and 82.6% vs. 80.5% for HL patients. A minority of patients showed a predicted PFS deviation > 10% between the two methods: 2.33% in DLBCL, 6.51% in FL and 1% in HL. CONCLUSION TMTV measured with the SUV4.0 threshold provides a comparable PFS prediction than the 41%SUVmax method supporting its routine adoption particularly in the diffuse large B cell lymphoma subtype.
Collapse
Affiliation(s)
| | | | - Catherine Thieblemont
- APHP, Hôpital Saint-Louis, Hemato-Oncology, Paris, France
- Université Paris Cité, Paris, France
| | - Franck Morschhauser
- Department of Hematology, Univ. Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche Sur les Formes Injectables et les Technologies Associées, Lille, F-59000, France
| | | | - Marc André
- Department of Hematology, CHU UCL Namur, Université Catholique de Louvain, Yvoir, Belgium
| | - Steven Le Gouill
- Service d'hématologie, Institut Curie, Saint Cloud, France
- Université de Versailles Saint-Quentin (UVSQ), Paris, France
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), U1288 Inserm/Institut Curie Centre de Recherche, Paris, France
| | - Yassine Al Tabaa
- Scintidoc Nuclear Medicine Center, Clinique Clémentville, Montpellier, France
| | - Paul Bland Durand
- Department of Nuclear Medicine, CHU H. Mondor, U-PEC, AP-HP, Créteil, France
| | - Clement Bailly
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, Nantes, 44000, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, Nantes, 44007, France
| | - Veronique Edeline
- Department of Nuclear Medicine, Hôpital La Pitié Salpetrière, Paris, France
| | - Lavinia Vija
- Nuclear Medicine Department, Oncopole Claudius Regaud, Toulouse, France
| | - Laetitia Vercellino
- Nuclear Medicine Department, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Paris, France
- INSERM UMR_S942, Université Paris Cité, Paris, 75006, France
| | - Romain Ricci
- LYSARC, Centre Hospitalier Lyon-Sud, 165 Chemin du Grand Revoyet Bâtiment 2D, Pierre-Bénite, 69310, France
| | - Salim Kanoun
- Centre de Recherche Clinique de Toulouse, Team 9, Toulouse, France
| | - Anne-Ségolène Cottereau
- Nuclear Medicine Department, AP-HP, Hôpital Cochin, Paris, France.
- Université Paris Cité, Paris, 75006, France.
| |
Collapse
|
9
|
Beck M, Blumenberg V, Bücklein VL, Bundschuh RA, Harrer DC, Hirschbühl K, Jung J, Kunz WG, Menhart K, Winkelmann M, Yakushev I, Illert AL, Eckstein M, Völkl S, Claus R, Hansmann L, Hecker JS, Kuwert T, Mackensen A, Subklewe M, Hellwig D, Müller F. Liver-FDG-uptake augments early PET/CT prognostic value for CD19-targeted CAR-T cell therapy in diffuse large B cell lymphoma. EJNMMI Res 2025; 15:25. [PMID: 40095158 PMCID: PMC11914545 DOI: 10.1186/s13550-025-01201-1] [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: 10/09/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Despite revolutionary efficacy of CD19-CAR-T cell therapy (CAR-T) in aggressive B cell lymphoma, many patients still relapse mostly early. In early failure, distinct drugs support CAR-T which makes reliable and early prediction of imminent relapse/refractoriness critical. A complete metabolic remission (CR) on Fluor-18-Deoxyglucose (FDG) Positron-Emission-Computed Tomography (PET) 30 days after CAR-T (PET30) strongly predicts progression-free survival (PFS), but still fails in a relevant proportion of patients. We aimed to identify additional routine parameters in PET evaluation to enhance CAR-T response prediction. RESULTS Thirty patients with aggressive B cell lymphoma treated with CAR-T were retrospectively analyzed. Pre-CAR-T, LDH was the strongest PFS-predictor also by multivariate analysis. Post-CAR-T, 10 out of 14 patients (71.4%) with PET30-CR remained in disease remission, while 12 out of 16 patients (75%) with incomplete metabolic remission (PET30-nCR) relapsed after CAR-T. 28.6% of patients with PET30-CR ultimately progressed. Change of liver FDG-uptake from baseline to day30 (Delta-Liver-SUVmean) was identified as an independent biomarker for response. PET30-nCR and a decrease of Delta-Liver-SUVmean were associated with a high risk of tumor progression (HR 4.79 and 3.99, respectively). The combination of PET30 and Delta-Liver-SUVmean identified patients at very low, at intermediate and at very high risk of relapse (PFS not reached, 7.5 months, 1.5 months, respectively). CONCLUSION Additionally to PET30 metabolic remission, longitudinal metabolic changes in Delta-Liver-SUVmean predicted CAR-T efficiency. Our results may guide early intervention studies aiming to enhance CAR-T particularly in the very high-risk patients.
Collapse
Affiliation(s)
- Michael Beck
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany.
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
| | - Viktoria Blumenberg
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Veit L Bücklein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | - Ralph A Bundschuh
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Nuclear Medicine, Faculty of Medicine, University Hospital of Augsburg, Augsburg, Germany
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Dennis C Harrer
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Klaus Hirschbühl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Johannes Jung
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Wolfgang G Kunz
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Karin Menhart
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Michael Winkelmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Igor Yakushev
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, School of Medicine, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Anna Lena Illert
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Eckstein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Pathology, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
| | - Simon Völkl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Rainer Claus
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Leo Hansmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Judith S Hecker
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Center for Translational Cancer Research, Technical University of Munich (TUM), TranslaTUM, Munich, Germany
| | - Torsten Kuwert
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
| | - Andreas Mackensen
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Marion Subklewe
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Dirk Hellwig
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Fabian Müller
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany.
| |
Collapse
|
10
|
Torkaman M, Jemaa S, Fredrickson J, Fernandez Coimbra A, De Crespigny A, Carano RAD. Comparative analysis of intestinal tumor segmentation in PET CT scans using organ based and whole body deep learning. BMC Med Imaging 2025; 25:52. [PMID: 39962481 PMCID: PMC11834234 DOI: 10.1186/s12880-025-01587-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND 18-Fluoro-deoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) is a valuable imaging tool widely used in the management of cancer patients. Deep learning models excel at segmenting highly metabolic tumors but face challenges in regions with complex anatomy and normal cell uptake, such as the gastro-intestinal tract. Despite these challenges, it remains important to achieve accurate segmentation of gastro-intestinal tumors. METHODS Here, we present an international multicenter comparative study between a novel organ-focused approach and a whole-body training method to evaluate the effectiveness of training data homogeneity in accurately identifying gastro-intestinal tumors. In the organ-focused method, the training data is limited to cases with intestinal tumors which makes the network trained with more homogeneous data and with stronger presence of intestinal tumor signals. The whole body approach extracts the intestinal tumors from the results of a model trained on the whole-body scans. Both approaches were trained using diffuse large B cell (DLBCL) patients from a large multi-center clinical trial (NCT01287741). RESULTS We report an improved mean(±std) Dice score of 0.78(±0.21) for the organ-based approach on the hold-out set, compared to 0.63(±0.30) for the whole-body approach, with the p-value of less than 0.0001. At the lesion level, the proposed organ-based approach also shows increased precision, recall, and F1-score. An independent trial was used to evaluate the generalizability of the proposed method to non-Hodgkin's lymphoma (NHL) patients with follicular lymphoma (FL). CONCLUSION Given the variability in structure and metabolism across tissues in the body, our quantitative findings suggest organ-focused training enhances intestinal tumor segmentation by leveraging tissue homogeneity in the training data, contrasting with the whole-body training approach, which, by its very nature, is a more heterogeneous data set.
Collapse
Affiliation(s)
| | | | | | | | | | - Richard A D Carano
- Genentech, Inc, South San Francisco, CA, USA
- F. Hoffman-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
11
|
Mendeville MS, Janssen J, Los-de Vries GT, van Dijk E, Richter J, Nijland M, Roemer MGM, Stathi P, Hijmering NJ, Bladergroen R, Pelaz DA, Diepstra A, Eertink CJ, Burggraaff CN, Kim Y, Lugtenburg PJ, van den Berg A, Tzankov A, Dirnhofer S, Dührsen U, Hüttmann A, Klapper W, Zijlstra JM, Ylstra B, de Jong D. Integrating genetic subtypes with PET scan monitoring to predict outcome in diffuse large B-cell lymphoma. Nat Commun 2025; 16:109. [PMID: 39747123 PMCID: PMC11696268 DOI: 10.1038/s41467-024-55614-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: 07/22/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Next Generation Sequencing-based subtyping and interim- and end of treatment positron emission tomography (i/eot-PET) monitoring have high potential for upfront and on-treatment risk assessment of diffuse large B-cell lymphoma patients. We performed Dana Farber Cancer Institute (DFCI) and LymphGen genetic subtyping for the HOVON84 (n = 208, EudraCT-2006-005174-42) and PETAL (n = 204, EudraCT-2006-001641-33) trials retrospectively combined with DFCI genetic data (n = 304). For all R-CHOP treated patients (n = 592), C5/MCD- and C2/A53-subtypes show significantly worse outcome independent of the international prognostic index. For all subtypes, adverse prognostic value of i/eot-PET-positive status is confirmed. Consistent with frequent primary refractory disease, only 67% C2 patients become eot-PET-negative versus 81-88% for other subtypes. Indicative of high relapse rates, outcome of C5 i/eot-PET-negative patients remains significantly worse in HOVON-84, which trend validates in the PETAL and SAKK38-07 trials (NCT00544219). These results show the added value of integrated genetic subtyping and PET monitoring for prognostic stratification and subtype-specific trial design.
Collapse
Affiliation(s)
- Matías S Mendeville
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Jurriaan Janssen
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - G Tjitske Los-de Vries
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Julia Richter
- Department of Pathology, Hematopathology Section and Lymph Node Registry University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Marcel Nijland
- Department of Hematology University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Margaretha G M Roemer
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Phylicia Stathi
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Nathalie J Hijmering
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Pathology, HOVON Pathology Facility and Biobank (HOP), Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Reno Bladergroen
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Diego A Pelaz
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Corinne J Eertink
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Coreline N Burggraaff
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Yongsoo Kim
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Alexandar Tzankov
- Institute of Pathology, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Stefan Dirnhofer
- Institute of Pathology, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Ulrich Dührsen
- Department of Hematology, University Hospital Essen, Essen, Germany
| | - Andreas Hüttmann
- Department of Hematology, University Hospital Essen, Essen, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Josée M Zijlstra
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands.
| | - Daphne de Jong
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Ababneh HS, Frigault MJ, Ng AK, Patel CG. Post-CAR T-Cell Therapy Failure Metabolic Parameters Predict Survival and Response in Large B-Cell Lymphoma. Hematol Oncol 2025; 43:e70025. [PMID: 39688000 DOI: 10.1002/hon.70025] [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: 05/13/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
18F-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) parameters have shown a significant prognostic role in relapsed/refractory large B-cell lymphoma (LBCL) patients undergoing CD19-targeted chimeric antigen receptor (CAR) T-cell therapy. While a substantial body of evidence exists on the prognostic value of PET/CT parameters in peri-CAR T setting, data available on the prognostic value of PET/CT parameters following CAR T-cell therapy failure is lacking. Therefore, we sought to analyze the PET/CT scans of LBCL patients who experienced post-CAR T relapsed/progressive disease and subsequently received salvage therapies. Thirty-three LBCL patients who had PET-CT scans done demonstrating post-CAR T failure and then received salvage therapies [as a first salvage modality: RT alone, nine patients; combined modality therapy (CMT), seven patients; systemic therapy (ST) alone, 17 patients] were analyzed. The median follow-up after CAR T-cell infusion was 11.7 months [interquartile range (IQR): 5.1-24.4 months], and the median follow-up after post-CAR T salvage therapy was 7.3 months (IQR: 2.7-19.1 months). The median timeframe for the PET scan showing post-CAR T failure was 2.4 months (IQR: 0.96-5.0 months). On univariable analysis from salvage therapy start date, high metabolic tumor volume (MTV) and total lesion glycolysis (TLG) were associated with inferior overall survival (OS) (Hazard ratio -HR = 8.4, p < 0.0001; HR = 3.2, p = 0.01, respectively). High MTV was associated with a non-significant trend of inferior progression-free survival (PFS) (HR = 3.5, p = 0.09). High maximum standardized uptake value (SUVmax) was not associated with inferior OS or inferior PFS. On multivariable analysis from salvage therapy start date, high MTV (HR = 4.6, 95% CI: 1.5-14.3, p = 0.009) was identified to be an independent prognostic factor for inferior OS. High International Prognostic Index (IPI) (≥ 3) at the time of salvage therapy (HR = 2.5, 95% CI: 1.1-5.6, p = 0.02) was significantly associated with inferior PFS. Our study shows that semiquantitative PET/CT metrics, especially MTV, are significant prognostic indicators of overall survival in this highly refractory population after CAR T-cell therapy failure, potentially refining prognostic and treatment approaches beyond conventional parameters like IPI.
Collapse
Affiliation(s)
- Hazim S Ababneh
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Division of Hematology & Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea K Ng
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chirayu G Patel
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Zhang W, Ye B, Song Y, Yang P, Si W, Jing H, Yang F, Yuan D, Wu Z, Lyu J, Peng K, Zhang X, Wang L, Li Y, Liu Y, Wu C, Hao X, Zhang Y, Qi W, Wang J, Dong F, Zhao Z, Jing H, Li Y. Integrating multi-omics features enables non-invasive early diagnosis and treatment response prediction of diffuse large B-cell lymphoma. Clin Transl Med 2025; 15:e70174. [PMID: 39776291 PMCID: PMC11705727 DOI: 10.1002/ctm2.70174] [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: 08/23/2024] [Revised: 11/13/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Multi-omics features of cell-free DNA (cfDNA) can effectively improve the performance of non-invasive early diagnosis and prognosis of cancer. However, multimodal characterization of cfDNA remains technically challenging. METHODS We developed a comprehensive multi-omics solution (COMOS) to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. The COMOS was tested on 214 plasma samples of diffuse large B-cell lymphoma (DLBCL) and matched healthy controls. RESULTS For early diagnosis, COMOS improved the area under the curve (AUC) value to .993 compared with the individual omics model, with a sensitivity of 95% at 98% specificity. Detection sensitivity achieved 91% at 99% specificity in early-stage patients, while the AUC values of the individual omics model were 0.942, 0.968, 0.989, 0.935, 0.921, 0.781 and 0.917, respectively, with lower sensitivity and specificity. In the treatment response cohort, COMOS yielded a superior sensitivity of 88% at 86% specificity (AUC, 0.903). COMOS has achieved excellent performance in early diagnosis and treatment response prediction. CONCLUSIONS Our study provides an effectively improved approach with high accuracy for the diagnosis and prognosis of DLBCL, showing great potential for future clinical application. KEY POINTS A comprehensive multi-omics solution to specifically obtain an extensive fragmentomics landscape, presented by breakpoint characteristics of nucleosomes, CpG islands, DNase clusters and enhancers, besides typical methylation, copy number alteration of cfDNA. Integrated model of cfDNA multi-omics could be used for non-invasive early diagnosis of DLBCL. Integrated model of cfDNA multi-omics could effectively evaluate the efficacy of R-CHOP before DLBCL treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/blood
- Female
- Male
- Middle Aged
- Aged
- Adult
- Early Detection of Cancer/methods
- Prognosis
- Cell-Free Nucleic Acids/blood
- Cell-Free Nucleic Acids/analysis
- Rituximab/therapeutic use
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Doxorubicin/therapeutic use
- Early Diagnosis
- Cyclophosphamide/therapeutic use
- Multiomics
Collapse
Affiliation(s)
- Weilong Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Yang Song
- BOE Technology Group Co., LtdBeijingChina
| | - Ping Yang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenzhe Si
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
| | | | - Fan Yang
- BOE Technology Group Co., LtdBeijingChina
| | - Dan Yuan
- BOE Technology Group Co., LtdBeijingChina
| | - Zhihong Wu
- BOE Technology Group Co., LtdBeijingChina
| | - Jiahao Lyu
- BOE Technology Group Co., LtdBeijingChina
| | - Kang Peng
- BOE Technology Group Co., LtdBeijingChina
| | - Xu Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Lingli Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Li
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yan Liu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Chaoling Wu
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Xiaoyu Hao
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yuqi Zhang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Wenxin Qi
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Jing Wang
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Fei Dong
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | | | - Hongmei Jing
- Department of HematologyLymphoma Research CenterPeking University Third HospitalBeijingChina
| | - Yanzhao Li
- BOE Technology Group Co., LtdBeijingChina
| |
Collapse
|
14
|
Dabrowska-Iwanicka A, Nowakowski GS. DLBCL: who is high risk and how should treatment be optimized? Blood 2024; 144:2573-2582. [PMID: 37922443 DOI: 10.1182/blood.2023020779] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/05/2023] Open
Abstract
ABSTRACT Diffuse large B-cell lymphoma (DLBCL), not otherwise specified, is the most common subtype of large B-cell lymphoma, with differences in prognosis reflecting heterogeneity in the pathological, molecular, and clinical features. Current treatment standard is based on multiagent chemotherapy, including anthracycline and monoclonal anti-CD20 antibody, which leads to cure in 60% of patients. Recent years have brought new insights into lymphoma biology and have helped refine the risk groups. The results of these studies inspired the design of new clinical trials with targeted therapies and response-adapted strategies and allowed to identify groups of patients potentially benefiting from new agents. This review summarizes recent progress in identifying high-risk patients with DLBCL using clinical and biological prognostic factors assessed at diagnosis and during treatment in the front-line setting, as well as new treatment strategies with the application of targeted agents and immunotherapy, including response-adapted strategies.
Collapse
Affiliation(s)
- Anna Dabrowska-Iwanicka
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | |
Collapse
|
15
|
Sun Z, Yang T, Ding C, Shi Y, Cheng L, Jia Q, Tao W. Clinical scoring systems, molecular subtypes and baseline [ 18F]FDG PET/CT image analysis for prognosis of diffuse large B-cell lymphoma. Cancer Imaging 2024; 24:168. [PMID: 39696503 DOI: 10.1186/s40644-024-00810-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous hematological malignancy resulting in a range of outcomes, and the early prediction of these outcomes has important implications for patient management. Clinical scoring systems provide the most commonly used prognostic evaluation criteria, and the value of genetic testing has also been confirmed by in-depth research on molecular typing. [18F]-fluorodeoxyglucose positron emission tomography / computed tomography ([18F]FDG PET/CT) is an invaluable tool for predicting DLBCL progression. Conventional baseline image-based parameters and machine learning models have been used in prognostic FDG PET/CT studies of DLBCL; however, numerous studies have shown that combinations of baseline clinical scoring systems, molecular subtypes, and parameters and models based on baseline FDG PET/CT image may provide better predictions of patient outcomes and aid clinical decision-making in patients with DLBCL.
Collapse
Affiliation(s)
- Zhuxu Sun
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Tianshuo Yang
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Chongyang Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Luyi Cheng
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Qingshen Jia
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University, Tianjin, China
| | - Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China.
| |
Collapse
|
16
|
Koumpis E, Georgoulis V, Papathanasiou K, Papoudou-Bai A, Kanavaros P, Kolettas E, Hatzimichael E. The Role of microRNA-155 as a Biomarker in Diffuse Large B-Cell Lymphoma. Biomedicines 2024; 12:2658. [PMID: 39767565 PMCID: PMC11673977 DOI: 10.3390/biomedicines12122658] [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: 10/19/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Diffuse Large B-cell Lymphoma (DLBCL) is the most common aggressive non-Hodgkin lymphoma (NHL). Despite the use of newer agents, such as polatuzumab vedotin, more than one-third of patients have ultimately relapsed or experienced refractory disease. MiRNAs are single-stranded, ~22-nucleotide-long RNAs that interact with their target RNA. They are significant regulators of post-transcriptional gene expression. One significant miRNA, miR-155, is involved in the pathophysiology of DLBCL and it is a critical modulator of hematopoiesis, inflammation, and immune responses. Targets of miR-155, such as histone deacetylase 4 (HDAC4), suppressor of cytokine signaling-1 (SOCS1) and immune cells, play a crucial role in DLBCL pathogenesis, since miR-155 regulates key pathways, transcription factors and cytokine expression and shapes the tumor microenvironment in DLBCL. In this review, we examine the role of miR-155 in DLBCL and its potential as a future diagnostic, prognostic, or predictive biomarker.
Collapse
Affiliation(s)
- Epameinondas Koumpis
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (E.K.); (V.G.); (K.P.)
| | - Vasileios Georgoulis
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (E.K.); (V.G.); (K.P.)
| | - Konstantina Papathanasiou
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (E.K.); (V.G.); (K.P.)
| | - Alexandra Papoudou-Bai
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Evangelos Kolettas
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, Institute of Biosciences, University Centre for Research and Innovation, University of Ioannina, 45110 Ioannina, Greece;
- Biomedical Research Institute, Foundation for Research and Technology, 45110 Ioannina, Greece
| | - Eleftheria Hatzimichael
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (E.K.); (V.G.); (K.P.)
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Hubbeling H, Leithner D, Silverman EA, Flynn J, Devlin S, Shah G, Fregonese B, Sanin BW, Bedmutha A, Tomas AA, Parascondola A, Saldia A, Landego I, Hajj C, Boardman AP, Dahi PB, Ghosh A, Giralt S, Lin RJ, Park J, Scordo M, Salles G, Yahalom J, Palomba ML, Schöder H, Perales MA, Shouval R, Imber BS. Metabolic Tumor Volume Response after Bridging Therapy Determines Chimeric Antigen Receptor T-Cell Outcomes in Large B-Cell Lymphoma. Clin Cancer Res 2024; 30:5083-5093. [PMID: 39259292 PMCID: PMC12001374 DOI: 10.1158/1078-0432.ccr-24-0830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE Greater disease burden is a well-established predictor of poorer outcomes following chimeric antigen receptor T-cell (CAR T) therapy. Although bridging therapy (BT) is widely used between leukapheresis and CAR T infusion, limited data have evaluated the impact of BT on CAR T outcomes. In this study, we hypothesized that the quantitative dynamics of radiomic cytoreduction during bridging are prognostic. EXPERIMENTAL DESIGN Patients with large B-cell lymphoma treated with CD19-CAR T from 2016 to 2022 were included in the study. Metabolic tumor volume (MTV) was determined for all patients on pre-leukapheresis PET and on post-BT/pre-infusion PET in those who received BT. Patients were stratified into "High" and "Low" disease burden using an MTV cutpoint of 65.4cc established by maximally selected log-rank statistic for progression-free survival (PFS). RESULTS Of 191 patients treated with CAR T, 144 (75%) received BT. In the BT cohort, 56% had a reduction in MTV post-BT. On multivariate analysis, the MTV trajectory across the bridging period remained significantly associated with PFS (P < 0.001); however, notably, patients with improved MTV (High->Low) had equivalent PFS compared with those with initially and persistently low MTV (Low->Low; HR for High->Low MTV: 2.74; 95% confidence interval, 0.82-9.18). There was a reduction in any grade immune effector cell-associated neurotoxicity syndrome in the High->Low MTV cohort as compared with the High->High MTV cohort (13% vs. 41%; P = 0.05). CONCLUSIONS This is the first study to use radiomics to quantify disease burden pre- and post-BT in a large real-world large B-cell lymphoma cohort. We demonstrate that effective BT can enable initially high-disease burden patients to achieve post-CAR T outcomes comparable with low-disease burden patients.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Immunotherapy, Adoptive/methods
- Aged
- Tumor Burden
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Adult
- Prognosis
- Treatment Outcome
- Antigens, CD19/immunology
- Young Adult
- Retrospective Studies
Collapse
Affiliation(s)
- Harper Hubbeling
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiation Oncology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Doris Leithner
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Emily A. Silverman
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Flynn
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean Devlin
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunjan Shah
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Beatrice Fregonese
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Beatriz W. Sanin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Akshay Bedmutha
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Alarcon Tomas
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allison Parascondola
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amethyst Saldia
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan Landego
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carla Hajj
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander P. Boardman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Parastoo B. Dahi
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnab Ghosh
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Giralt
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard J. Lin
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Michael Scordo
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M. Lia Palomba
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Heiko Schöder
- Department of Radiation Oncology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Miguel-Angel Perales
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Roni Shouval
- Department of Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Brandon S. Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
18
|
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
|
19
|
Chartier L, Belot A, Chaillol I, Elsensohn MH, Portugues C, Fournier M, Joubert C, Gat E, Pizot C, Fogarty P, Murairi T, Ammar RO, Paget J, Cherblanc F, Ricci R, Vercellino L, Kanoun S, Cottereau AS, Thieblemont C, Casasnovas O. Precautions to Consider in the Analysis of Prognostic and Predictive Indices. J Nucl Med 2024; 65:1672-1678. [PMID: 39486863 DOI: 10.2967/jnumed.123.267021] [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: 05/15/2024] [Accepted: 09/10/2024] [Indexed: 11/04/2024] Open
Abstract
Understanding the differences between prognostic and predictive indices is imperative for medical research advances. We have developed a new prognostic measure that will identify the strengths, limitations, and potential applications in clinical practice.
Collapse
Affiliation(s)
- Loïc Chartier
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France;
| | - Aurélien Belot
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Isabelle Chaillol
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | - Cédric Portugues
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | - Clémentine Joubert
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Elodie Gat
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Cécile Pizot
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Patrick Fogarty
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Tesla Murairi
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Romain Ould Ammar
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Jérôme Paget
- Biostatistics Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Fanny Cherblanc
- Medical Department, LYSARC, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Romain Ricci
- Imaging Department, LYSARC, Hôpital Henri-Mondor, Créteil, France
| | - Laetitia Vercellino
- Department of Nuclear Medicine, Hôpital Saint-Louis, AP-HP, INSERM UMR S942, Université Paris Cité, Paris, France
| | - Salim Kanoun
- Department of Hematology, Cancer Research Center of Toulouse, Team 9, INSERM Unité Mixte de Recherche 1037, Toulouse, France
| | | | - Catherine Thieblemont
- Assistance Publique-Hôpitaux de Paris, Université de Paris, and Hemato-Oncologie, Hôpital Saint-Louis, Paris, France; and
| | - Olivier Casasnovas
- Department of Hematology and INSERM 1231, CHU Dijon Bourgogne, Dijon, France
| |
Collapse
|
20
|
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
|
21
|
Kim SY, Chung HW, So Y, Lee MH, Lee EJ. Recent Updates of PET in Lymphoma: FDG and Beyond. Biomedicines 2024; 12:2485. [PMID: 39595051 PMCID: PMC11592097 DOI: 10.3390/biomedicines12112485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/12/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Lymphoma is one of the most common cancers worldwide, categorized into Hodgkin lymphoma and non-Hodgkin lymphoma. 18F-fluorodeoxyglucose positron emission tomography (FDG PET) has become an essential imaging tool for evaluating patients with lymphoma in terms of initial diagnosis, staging, prognosis, and treatment response assessment. Recent advancements in imaging technology and methodologies, along with the development of artificial intelligence, have revolutionized the evaluation of complex imaging data, enhancing the diagnostic and predictive power of PET in lymphoma. However, FDG is not cancer-specific, but it primarily reflects glucose metabolism, which has prompted the investigation of alternative PET tracers to address this limitation. Novel PET radiotracers, such as fibroblast activation protein inhibitors targeting the tumor microenvironment, have recently shown promising results in evaluating various malignancies compared to FDG PET. Furthermore, with the rapid advancements in immunotherapy and the favorable imaging properties of 89Zr, immunoPET has emerged as a promising modality, offering insights into the functional and molecular status of the immune system. ImmunoPET can also facilitate the development of new antibody therapeutics and radioimmunotherapy by providing pharmacokinetic and pharmacodynamic data. This review provides comprehensive insights into the current clinical applications of FDG PET in lymphoma, while also exploring novel PET imaging radiotracers beyond FDG, discussing their mechanisms of action and potential impact on patient management.
Collapse
Affiliation(s)
- Sung-Yong Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Hyun Woo Chung
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
- Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Young So
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
| | - Mark Hong Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Eun Jeong Lee
- Department of Nuclear Medicine, Seoul Medical Center, 156 Sinnae-ro, Jungnang-gu, Seoul 02053, Republic of Korea;
| |
Collapse
|
22
|
Chen J, Lin F, Dai Z, Chen Y, Fan Y, Li A, Zhao C. Survival prediction in diffuse large B-cell lymphoma patients: multimodal PET/CT deep features radiomic model utilizing automated machine learning. J Cancer Res Clin Oncol 2024; 150:452. [PMID: 39382750 PMCID: PMC11464575 DOI: 10.1007/s00432-024-05905-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/21/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE We sought to develop an effective combined model for predicting the survival of patients with diffuse large B-cell lymphoma (DLBCL) based on the multimodal PET-CT deep features radiomics signature (DFR-signature). METHODS 369 DLBCL patients from two medical centers were included in this study. Their PET and CT images were fused to construct the multimodal PET-CT images using a deep learning fusion network. Then the deep features were extracted from those fused PET-CT images, and the DFR-signature was constructed through an Automated machine learning (AutoML) model. Combined with clinical indexes from the Cox regression analysis, we constructed a combined model to predict the progression-free survival (PFS) and the overall survival (OS) of patients. In addition, the combined model was evaluated in the concordance index (C-index) and the time-dependent area under the ROC curve (tdAUC). RESULTS A total of 1000 deep features were extracted to build a DFR-signature. Besides the DFR-signature, the combined model integrating metabolic and clinical factors performed best in terms of PFS and OS. For PFS, the C-indices are 0.784 and 0.739 in the training cohort and internal validation cohort, respectively. For OS, the C-indices are 0.831 and 0.782 in the training cohort and internal validation cohort. CONCLUSIONS DFR-signature constructed from multimodal images improved the classification accuracy of prognosis for DLBCL patients. Moreover, the constructed DFR-signature combined with NCCN-IPI exhibited excellent potential for risk stratification of DLBCL patients.
Collapse
Affiliation(s)
- Jianxin Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Fengyi Lin
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Zhaoyan Dai
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yu Chen
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yawen Fan
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Ang Li
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Chenyu Zhao
- The Key Laboratory of Broadband Wireless Communication and Sensor Network Technology (Ministry of Education), Nanjing University of Posts and Telecommunications, Nanjing, China
| |
Collapse
|
23
|
Withofs N, Bonnet C, Hustinx R. 2-deoxy-2-[ 18F]FDG PET Imaging for Therapy Assessment in Hodgkin's and Non-Hodgkin Lymphomas. PET Clin 2024; 19:447-462. [PMID: 38945737 DOI: 10.1016/j.cpet.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography combined with computed tomography (PET/CT) has contributed to outcome improvement of patients with lymphoma. The use of [18F]FDG PET/CT for staging and response assessment is successfully applied both in routine clinical practice and in clinical trials. The challenges lie in enhancing the outcomes of lymphoma patients, particularly those with advanced or refractory/relapsed disease, and to minimize the long-term toxicity associated with treatments, including radiation therapy. The objective of this review article is to present contemporary data on the use of [18F]FDG PET/CT for treatment assessment of aggressive lymphomas.
Collapse
Affiliation(s)
- Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Quartier Hopital, Avenue de l'hopital 1, Liege, Belgium; GIGA-Nuclear Medicine Lab, University of Liege, CHU - B34 Quartier Hôpital, Avenue de l'Hôpital 11, Liège, BELGIQUE.
| | - Christophe Bonnet
- Department of Hematology, CHU of Liege, Quartier Hôpital, Avenue de l'hôpital 1, 4000 Liege 1, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Quartier Hopital, Avenue de l'hopital 1, Liege, Belgium; GIGA-Nuclear Medicine Lab, University of Liege, CHU - B34 Quartier Hôpital, Avenue de l'Hôpital 11, Liège, BELGIQUE
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Ikeda D, Oura M, Uehara A, Tabata R, Narita K, Takeuchi M, Machida Y, Matsue K. Prognostic relevance of tumor-infiltrating CD4 + cells and total metabolic tumor volume-based risk stratification in diffuse large B-cell lymphoma. Haematologica 2024; 109:2822-2832. [PMID: 38572548 PMCID: PMC11367203 DOI: 10.3324/haematol.2024.285038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
In order to elucidate the relationship between pretreatment radiomic parameters and the proportions of various tumor-infiltrating (TI) cells, we retrospectively analyzed the association of total metabolic tumor volume (TMTV) and TI cells on biopsied tumor lesions in 171 patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL). The surface markers of TI cells were analyzed by multicolor flow cytometry using a dissected single-cell suspension. In examining the correlation between TI cells and positron-emission tomography-derived parameters (maximum standardized uptake value [SUVmax], total metabolic tumor volume [TMTV], and total lesion glycolysis), intratumoral cell types minimally influenced the results, except for a weak negative correlation between CD4+ cells and SUVmax (R=-0.16, P=0.045). Even for the lesion fluorodeoxyglucose uptake at the biopsied site, CD19+ cells (indicative of malignant burden) showed only a weak correlation with the highest SUV (R=0.21, P=0.009), whereas CD3+ (R=-0.25, P=0.002) and CD4+ cells (R=-0.29, P<0.001) demonstrated a similarly weak inverse correlation. High TMTV and low TI CD4+ cells were independently associated with poor prognosis and their combination identified the most adverse population (3-year progression-free survival: 32.3%, 95% confidence interval [CI]: 19.4-53.7; 3-year overall survival: 48.4%, 95% CI: 33.6-69.6). Moreover, radiomic parameters incorporating the international prognostic index significantly improved the 3-year survival prediction (area under the curve: 0.76, P<0.05) compared to their standalone use. This study underscores the prognostic impact of TI CD4+ cells on DLBCL and suggests that integration of TMTV and TI cell analysis enhances the accuracy of prognostic prediction.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Male
- Female
- Middle Aged
- Prognosis
- Aged
- Adult
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Tumor Burden
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Aged, 80 and over
- Retrospective Studies
- Positron-Emission Tomography
- Risk Assessment
- Young Adult
- Fluorodeoxyglucose F18
Collapse
Affiliation(s)
- Daisuke Ikeda
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba.
| | - Mitsuaki Oura
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Atsushi Uehara
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Rikako Tabata
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Masami Takeuchi
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | | | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| |
Collapse
|
27
|
Wang J, Liu X, Wu Y, Zhong Q, Wu T, Yang Y, Chen B, Jing H, Tang Y, Jin J, Liu Y, Song Y, Fang H, Lu N, Li N, Zhai Y, Zhang W, Deng M, Wang S, Chen F, Yin L, Hu C, Qi S, Li Y. Association of overall survival benefit of radiotherapy with progression-free survival after chemotherapy for diffuse large B-cell lymphoma: A systematic review and meta-analysis. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:249-259. [PMID: 39281722 PMCID: PMC11401499 DOI: 10.1016/j.jncc.2024.04.002] [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: 02/07/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 09/18/2024] Open
Abstract
Objective To evaluate whether improved progression-free survival (PFS) from radiotherapy (RT) translates into an overall survival (OS) benefit for diffuse large B-cell lymphoma (DLBCL). Methods A systematic literature search identified randomized controlled trials (RCTs) and retrospective studies that compared combined-modality therapy (CMT) with chemotherapy (CT) alone. Weighted regression analyses were used to estimate the correlation between OS and PFS benefits. Cohen's kappa statistic assessed the consistency between DLBCL risk-models and PFS patterns. Furthermore, the benefit trend of RT was analyzed by fitting a linear regression model to the pooled hazard ratio (HR) according to the PFS patterns. Results For both 7 RCTs and 52 retrospective studies, correlations were found between PFS HR (HRPFS) and OS HR (HROS) at trial level (r = 0.639-0.876), and between PFS and OS rates at treatment-arm level, regardless of CT regimens (r = 0.882-0.964). Incorporating RT into CT increased about 18% of PFS, and revealed a different OS benefit profile. Patients were stratified into four CT-generated PFS patterns (>80%, >60-80%, >40-60%, and ≤40%), which was consistent with risk-stratified subgroups (kappa > 0.6). Absolute gain in OS from RT ranged from ≤5% at PFS >80% to about 21% at PFS ≤40%, with pooled HROS from 0.70 (95% CI, 0.51-0.97) to 0.48 (95% CI, 0.36-0.63) after rituximab-based CT. The OS benefit of RT was predominant in intermediate- and high-risk patients with PFS ≤ 80%. Conclusion We demonstrated a varied OS benefit profile of RT to inform treatment decisions and clinical trial design.
Collapse
Affiliation(s)
- Jingnan Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Xin Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yunpeng Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Qiuzi Zhong
- Beijing Hospital, National Geriatric Medical Center, Beijing, China
| | - Tao Wu
- Affiliated Hospital of Guizhou Medical University, Guizhou Cancer Hospital, Guiyang, Guizhou, China
| | - Yong Yang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yueping Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yongwen Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Ningning Lu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yirui Zhai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Wenwen Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Min Deng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Shulian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Fan Chen
- Department of Radiation Oncology, Affiliated Hospital of Qinghai University, Qinghai, China
| | - Lin Yin
- Department of Radiation Oncology, Affiliated Hospital of Qinghai University, Qinghai, China
| | - Chen Hu
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Shunan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yexiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| |
Collapse
|
28
|
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
|
29
|
Wang S, Mouliere F, Pegtel DM, Chamuleau MED. Turning the tide in aggressive lymphoma: liquid biopsy for risk-adapted treatment strategies. Trends Mol Med 2024; 30:660-672. [PMID: 38692937 DOI: 10.1016/j.molmed.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Diffuse large B cell lymphoma (DLBCL) exhibits significant biological and clinical heterogeneity that presents challenges for risk stratification and disease surveillance. Existing tools for risk stratification, including the international prognostic index (IPI), tissue molecular analyses, and imaging, have limited accuracy in predicting outcomes. The therapeutic landscape for aggressive lymphoma is rapidly evolving, and there is a pressing need to identify patients at risk of refractory or relapsed (R/R) disease in the context of personalized therapy. Liquid biopsy, a minimally invasive method for cancer signal detection, has been explored to address these challenges. We review advances in liquid biopsy strategies focusing on circulating nucleic acids in DLBCL patients and highlight their clinical potential. We also provide recommendations for biomarker-guided trials to support risk-adapted treatment modalities.
Collapse
Affiliation(s)
- Steven Wang
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Florent Mouliere
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Cancer Research UK National Biomarker Centre, University of Manchester, Wilmslow Road, Manchester, UK
| | - D Michiel Pegtel
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Martine E D Chamuleau
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Knaup H, Weindler J, van Heek L, Voltin CA, Fuchs M, Borchmann P, Dietlein M, Kobe C, Roth K. PET/CT Reconstruction and Its Impact on [Measures of] Metabolic Tumor Volume. Acad Radiol 2024; 31:3020-3025. [PMID: 38155023 DOI: 10.1016/j.acra.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
RATIONALE AND OBJECTIVES In oncological imaging, the use of metabolic tumor volume (MTV) for further prognostic differentiation and the development of risk adapted strategies appears promising. The aim of this analysis was to evaluate ultra-high definition (UHD) and ordered subset expectation maximization (OSEM) PET/CT reconstructions for their potential impact on different methods of MTV measurement. MATERIALS AND METHODS We analyzed positron emission tomography combined with computed tomography (PET/CT) scans of 40 Hodgkin lymphoma patients before first-line treatment who had undergone fluorodeoxyglucose (FDG) PET/CT. The MTVs were determined taking an SUV of 4.0 (MTV4.0) as a fixed threshold or 41% of the single hottest voxel (MTV41%) as an adaptive threshold for automated lymphoma delineation in both UHD and OSEM reconstructions. We then compared the absolute and relative differences between MTV4.0 and MTV41% in UHD and OSEM reconstructions. The relative distribution of MTV4.0 and MTV41% in relation to the reconstruction method applied was recorded and respective differences were tested for statistical significance using the paired sample t-test. RESULTS A comparison of MTV4.0 and MTV41% showed smaller relative and absolute differences in MTV between different reconstruction settings for the MTV4.0 method. Conversely, the absolute as well as the relative differences between MTVs obtained from different reconstructions settings were significantly greater when the MTV41% method was applied (p < 0001). CONCLUSION MTV4.0 brings higher robustness between different reconstruction settings, while with MTV41% the deviation between volumes obtained with different reconstruction settings is greater. For clinical routine and for multicenter settings, the MTV4.0 therefore appears most promising.
Collapse
Affiliation(s)
- Henry Knaup
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Jasmin Weindler
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Lutz van Heek
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Conrad-Amadeus Voltin
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Michael Fuchs
- German Hodgkin Study Group, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany (M.F., P.B.)
| | - Peter Borchmann
- German Hodgkin Study Group, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany (M.F., P.B.)
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| | - Carsten Kobe
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.).
| | - Katrin Roth
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Str. 62, Cologne, 50937, Germany (H.K., J.W., L.V.H., C.A.V., M.D., C.K., K.R.)
| |
Collapse
|
31
|
Lee W, Oh M, Kim JS, Sung M, Hong K, Kwak BJ, Park Y, Jun E, Song KB, Hwang DW, Lee JH, Yoo C, Kim KP, Park I, Jeong JH, Chang HM, Ryoo BY, Lee JB, Kim SC. Metabolic tumor burden as a prognostic indicator after neoadjuvant chemotherapy in pancreatic cancer. Int J Surg 2024; 110:4074-4082. [PMID: 38537071 PMCID: PMC11254192 DOI: 10.1097/js9.0000000000001389] [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: 12/20/2023] [Accepted: 03/11/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND There is no standardized assessment for evaluating response although neoadjuvant chemotherapy (NAT) is widely accepted for borderline resectable or locally advanced pancreatic cancer (BRPC or LAPC). This study was aimed to evaluate NAT response using positron emission tomography (PET) with 2-deoxy-2-[fluorine-18]fluoro-D-glucose ( 18 F-FDG-PET/CT) parameters alongside carbohydrate antigen (CA) 19-9 levels. METHODS Patients who underwent surgery after NAT for BRPC and LAPC between 2017 and 2021 were identified. The study assessed the prognostic value of PET-derived parameters after NAT, determining cutoff values using the K-adaptive partitioning method. It created four groups based on the elevation or normalization of PET parameters and CA19-9 levels, comparing survival between these groups. RESULTS Of 200 eligible patients, FOLFIRINOX and gemcitabine-based NAT was administered in 166 and 34 patients, respectively (mean NAT cycles, 8.3). In a multivariate analysis, metabolic tumor volume (MTV) demonstrated the most robust performance in assessing response [hazard ratio (HR) 3.11, 95% confidence interval (CI) 1.73-5.58, P <0.001] based on cutoff value of 2.4. Patients with decreased MTV had significantly better survival than those with elevated MTV among individuals with CA19-9 levels less than 37 IU/l (median survival; 35.5 vs. 20.9 months, P <0.001) and CA19-9 levels at least 37 IU/l (median survival; 34.3 vs. 17.8 months, P =0.03). In patients suspected to be Lewis antigen negative, the predictive performance of MTV was found to be limited ( P =0.84). CONCLUSION Elevated MTV is an influential prognostic factor for worse survival, regardless of post-NAT CA19-9 levels. These results could be helpful in identifying patients with a poor prognosis despite normalization of CA19-9 levels after NAT.
Collapse
Affiliation(s)
- Woohyung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine
| | - Minkyu Sung
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Kwangpyo Hong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Bong Jun Kwak
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Yejong Park
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Eunsung Jun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Ki Byung Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Dae Wook Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Jae Hoon Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Kyu-pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Heung-Moon Chang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine
| | - Jung Bok Lee
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
Locke FL, Oluwole OO, Kuruvilla J, Thieblemont C, Morschhauser F, Salles G, Rowe SP, Vardhanabhuti S, Winters J, Filosto S, To C, Cheng P, Schupp M, Korn R, Kersten MJ. Axicabtagene ciloleucel vs standard of care in second-line large B-cell lymphoma: outcomes by metabolic tumor volume. Blood 2024; 143:2464-2473. [PMID: 38557775 PMCID: PMC11208295 DOI: 10.1182/blood.2023021620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
ABSTRACT Metabolic tumor volume (MTV) assessed using 2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography, a measure of tumor burden, is a promising prognostic indicator in large B-cell lymphoma (LBCL). This exploratory analysis evaluated relationships between baseline MTV (categorized as low [median or less] vs high [greater than median]) and clinical outcomes in the phase 3 ZUMA-7 study (NCT03391466). Patients with LBCL relapsed within 12 months of or refractory to first-line chemoimmunotherapy were randomized 1:1 to axicabtagene ciloleucel (axi-cel; autologous anti-CD19 chimeric antigen receptor T-cell therapy) or standard care (2-3 cycles of chemoimmunotherapy followed by high-dose chemotherapy with autologous stem cell transplantation in patients who had a response). All P values are descriptive. Within high- and low-MTV subgroups, event-free survival (EFS) and progression-free survival (PFS) were superior with axi-cel vs standard care. EFS in patients with high MTV (vs low MTV) was numerically shorter with axi-cel and was significantly shorter with standard care. PFS was shorter in patients with high MTV vs low MTV in both the axi-cel and standard-care arms, and median MTV was lower in patients in ongoing response at data cutoff vs others. Median MTV was higher in patients treated with axi-cel who experienced grade ≥3 neurologic events or cytokine release syndrome (CRS) than in patients with grade 1/2 or no neurologic events or CRS, respectively. Baseline MTV less than or equal to median was associated with better clinical outcomes in patients receiving axi-cel or standard care for second-line LBCL. The trial was registered at www.clinicaltrials.gov as #NCT03391466.
Collapse
Affiliation(s)
- Frederick L. Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| | - Olalekan O. Oluwole
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Cancer Center, Nashville, TN
| | - John Kuruvilla
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Franck Morschhauser
- Department of Hematology, University of Lille, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | - Paul Cheng
- Kite, a Gilead Company, Santa Monica, CA
| | | | | | - Marie José Kersten
- Amsterdam University Medical Center (location University of Amsterdam), Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Cherng HJJ, Herrera A. Circulating Tumor DNA in Diffuse Large B-Cell Lymphoma: from Bench to Bedside? Curr Treat Options Oncol 2024; 25:659-678. [PMID: 38656685 DOI: 10.1007/s11864-024-01201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
OPINION STATEMENT Diffuse large B-cell lymphoma (DLBCL) is a curable disease with variable outcomes due to underlying heterogeneous clinical and molecular features-features that are insufficiently characterized with our current tools. Due to these limitations, treatment largely remains a "one-size-fits-all" approach. Circulating tumor DNA (ctDNA) is a novel biomarker in cancers that is increasingly utilized for risk stratification and response assessment. ctDNA is readily detectable from the plasma of patients with DLBCL but has not yet been incorporated into clinical care to guide treatment. Here, we describe how ctDNA sequencing represents a promising technology in development to personalize the care of patients with DLBCL. We will review the different types of ctDNA assays being studied and the rapidly growing body of evidence supporting the utility of ctDNA in different treatment settings in DLBCL. Risk stratification by estimation of tumor burden and liquid genotyping, molecular response assessment during treatment, and monitoring for measurable residual disease (MRD) to identify therapy resistance and predict clinical relapse are all potential applications of ctDNA. It is time for clinical trials in DLBCL to utilize ctDNA as an integral biomarker for patient selection, response-adapted designs, and surrogate endpoints. As more ctDNA assays become commercially available for routine use, clinicians should consider liquid biopsy when treatment response is equivocal on imaging. Incorporating MRD may also guide decision-making if patients experience severe treatment toxicities. Though important barriers remain, we believe that ctDNA will soon be ready to transition from bench to bedside to individualize treatment for our patients with DLBCL.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/genetics
- Humans
- Circulating Tumor DNA/blood
- Biomarkers, Tumor/blood
- Liquid Biopsy/methods
- Disease Management
- Translational Research, Biomedical
- Precision Medicine/methods
- Prognosis
- Clinical Decision-Making
- Disease Susceptibility
Collapse
Affiliation(s)
- Hua-Jay J Cherng
- Lymphoma Service, Division of Hematology & Oncology, Columbia University Irving Medical Center, 177 Fort Washington Avenue, 6GN-Rm 435, New York, NY, 10032, USA.
| | - Alex Herrera
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
34
|
Ikeda D, Oura M, Uehara A, Tabata R, Narita K, Takeuchi M, Machida Y, Matsue K. Real-world applicability of the International Metabolic Prognostic Index in DLBCL: a validation cohort study. Blood Adv 2024; 8:1893-1897. [PMID: 38359408 PMCID: PMC11021887 DOI: 10.1182/bloodadvances.2023012165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Affiliation(s)
- Daisuke Ikeda
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Mitsuaki Oura
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Atsushi Uehara
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Rikako Tabata
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Masami Takeuchi
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| | - Youichi Machida
- Department of Radiology, Kameda Medical Center, Chiba, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba, Japan
| |
Collapse
|
35
|
Parihar AS, Pant N, Subramaniam RM. Quarter-Century PET/CT Transformation of Oncology: Lymphoma. PET Clin 2024; 19:281-290. [PMID: 38403384 DOI: 10.1016/j.cpet.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The clinical landscape of lymphomas has changed dramatically over the last 2 decades, including significant progress made in the understanding and utilization of imaging modalities and the available treatment options for both indolent and aggressive lymphomas. Since the introduction of hybrid PET/CT scanners in 2001, the indications of 18F-fluorodeoxyglucose (FDG) PET/CT in the management of lymphomas have grown rapidly. In today's clinical practice, FDG PET/CT is used in successful management of the vast majority patients with lymphomas.
Collapse
Affiliation(s)
- Ashwin Singh Parihar
- Mallinckrodt Institute of Radiology; Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
| | | | - Rathan M Subramaniam
- Faculty of Medicine, Nursing, Midwifery & Health Sciences, The University of Notre Dame Australia, Sydney, Australia; Department of Radiology, Duke University, Durham, NC, USA; Department of Medicine, University of Otago Medical School, Dunedin, New Zealand
| |
Collapse
|
36
|
Voltin CA, Paccagnella A, Winkelmann M, Heger JM, Casadei B, Beckmann L, Herrmann K, Dekorsy FJ, Kutsch N, Borchmann P, Fanti S, Kunz WG, Subklewe M, Kobe C, Zinzani PL, Stelljes M, Roth KS, Drzezga A, Noppeney R, Rahbar K, Reinhardt HC, von Tresckow B, Seifert R, Albring JC, Blumenberg V, Farolfi A, Flossdorf S, Gödel P, Hanoun C. Multicenter development of a PET-based risk assessment tool for product-specific outcome prediction in large B-cell lymphoma patients undergoing CAR T-cell therapy. Eur J Nucl Med Mol Imaging 2024; 51:1361-1370. [PMID: 38114616 PMCID: PMC10957657 DOI: 10.1007/s00259-023-06554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE The emergence of chimeric antigen receptor (CAR) T-cell therapy fundamentally changed the management of individuals with relapsed and refractory large B-cell lymphoma (LBCL). However, real-world data have shown divergent outcomes for the approved products. The present study therefore set out to evaluate potential risk factors in a larger cohort. METHODS Our analysis set included 88 patients, treated in four German university hospitals and one Italian center, who had undergone 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography (PET) before CAR T-cell therapy with tisagenlecleucel or axicabtagene ciloleucel. We first determined the predictive value of conventional risk factors, treatment lines, and response to bridging therapy for progression-free survival (PFS) through forward selection based on Cox regression. In a second step, the additive potential of two common PET parameters was assessed. Their optimal dichotomizing thresholds were calculated individually for each CAR T-cell product. RESULTS Extra-nodal involvement emerged as the most relevant of the conventional tumor and patient characteristics. Moreover, we found that inclusion of metabolic tumor volume (MTV) further improves outcome prediction. The hazard ratio for a PFS event was 1.68 per unit increase of our proposed risk score (95% confidence interval [1.20, 2.35], P = 0.003), which comprised both extra-nodal disease and lymphoma burden. While the most suitable MTV cut-off among patients receiving tisagenlecleucel was 11 mL, a markedly higher threshold of 259 mL showed optimal predictive performance in those undergoing axicabtagene ciloleucel treatment. CONCLUSION Our analysis demonstrates that the presence of more than one extra-nodal lesion and higher MTV in LBCL are associated with inferior outcome after CAR T-cell treatment. Based on an assessment tool including these two factors, patients can be assigned to one of three risk groups. Importantly, as shown by our study, metabolic tumor burden might facilitate CAR T-cell product selection and reflect the individual need for bridging therapy.
Collapse
Affiliation(s)
- Conrad-Amadeus Voltin
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Andrea Paccagnella
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Michael Winkelmann
- Department of Radiology, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Jan-Michel Heger
- Department of Internal Medicine I, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Lymphoma Working Group (CLWG), Cologne, Germany
| | - Beatrice Casadei
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- 'L. e A. Seràgnoli' Institute of Hematology, Scientific Institute for Research, Hospitalization, and Healthcare (IRCCS) 'Azienda Ospedaliero-Universitaria Di Bologna', University of Bologna, Bologna, Italy
| | - Laura Beckmann
- Department of Internal Medicine I, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
| | - Franziska J Dekorsy
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Nadine Kutsch
- Department of Internal Medicine I, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Lymphoma Working Group (CLWG), Cologne, Germany
| | - Peter Borchmann
- Department of Internal Medicine I, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Lymphoma Working Group (CLWG), Cologne, Germany
| | - Stefano Fanti
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Division of Nuclear Medicine, Scientific Institute for Research, Hospitalization, and Healthcare (IRCCS) 'Azienda Ospedaliero-Universitaria Di Bologna', University of Bologna, Bologna, Italy
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, Comprehensive Cancer Center Munich (CCCM), University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, Gene Center Munich, Ludwig Maximilian University Munich, Munich, Germany
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF) Partner Site Munich, Munich, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Pier Luigi Zinzani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- 'L. e A. Seràgnoli' Institute of Hematology, Scientific Institute for Research, Hospitalization, and Healthcare (IRCCS) 'Azienda Ospedaliero-Universitaria Di Bologna', University of Bologna, Bologna, Italy
| | - Matthias Stelljes
- Department of Medicine A-Hematology, Oncology, and Pneumology, West German Cancer Center (WTZ) Network Partner Site, University Hospital Münster, University of Münster, Münster, Germany
| | - Katrin S Roth
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Richard Noppeney
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, University of Münster, Münster, Germany
| | - H Christian Reinhardt
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bastian von Tresckow
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Nuclear Medicine, University Hospital Münster, University of Münster, Münster, Germany
| | - Jörn C Albring
- Department of Medicine A-Hematology, Oncology, and Pneumology, West German Cancer Center (WTZ) Network Partner Site, University Hospital Münster, University of Münster, Münster, Germany
| | - Viktoria Blumenberg
- Department of Medicine III, Comprehensive Cancer Center Munich (CCCM), University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, Gene Center Munich, Ludwig Maximilian University Munich, Munich, Germany
- German Cancer Consortium (DKTK) and Bavarian Center for Cancer Research (BZKF) Partner Site Munich, Munich, Germany
| | - Andrea Farolfi
- Division of Nuclear Medicine, Scientific Institute for Research, Hospitalization, and Healthcare (IRCCS) 'Azienda Ospedaliero-Universitaria Di Bologna', University of Bologna, Bologna, Italy
| | - Sarah Flossdorf
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Philipp Gödel
- Department of Internal Medicine I, Center for Integrated Oncology Aachen-Bonn-Cologne-Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Lymphoma Working Group (CLWG), Cologne, Germany
| | - Christine Hanoun
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
37
|
Song GY, Jung SH, Ahn SY, Kim M, Ahn JS, Lee JJ, Kim HJ, Moon JB, Yoo SW, Kwon SY, Min JJ, Bom HS, Kang SR, Yang DH. Prognostic Significance Of Sequential 18f-fdg Pet/Ct During Frontline Treatment Of Peripheral T Cell Lymphomas. Korean J Intern Med 2024; 39:327-337. [PMID: 38268194 PMCID: PMC10918377 DOI: 10.3904/kjim.2023.323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/03/2023] [Accepted: 10/19/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND/AIMS The prognostic significance of 18F-fluorodeoxyglucose (FDG)-positron emission tomography-computed tomography (PET/CT) in peripheral T-cell lymphomas (PTCLs) are controversial. We explored the prognostic impact of sequential 18F-FDG PET/CT during frontline chemotherapy of patients with PTCLs. METHODS In total, 143 patients with newly diagnosed PTCLs were included. Sequential 18F-FDG PET/CTs were performed at the time of diagnosis, during chemotherapy, and at the end of chemotherapy. The baseline total metabolic tumor volume (TMTV) was calculated using the the standard uptake value with a threshold method of 2.5. RESULTS A baseline TMTV of 457.0 cm3 was used to categorize patients into high and low TMTV groups. Patients with a requirehigh TMTV had shorter progression-free survival (PFS) and overall survival (OS) than those with a low TMTV (PFS, 9.8 vs. 26.5 mo, p = 0.043; OS, 18.9 vs. 71.2 mo, p = 0.004). The interim 18F-FDG PET/CT response score was recorded as 1, 2-3, and 4-5 according to the Deauville criteria. The PFS and OS showed significant differences according to the interim 18F-FDG PET/CT response score (PFS, 120.7 vs. 34.1 vs. 5.1 mo, p < 0.001; OS, not reached vs. 61.1 mo vs. 12.1 mo, p < 0.001). CONCLUSION The interim PET/CT response based on visual assessment predicts disease progression and survival outcome in PTCLs. A high baseline TMTV is associated with a poor response to anthracycline-based chemotherapy in PTCLs. However, TMTV was not an independent predictor for PFS in the multivariate analysis.
Collapse
Affiliation(s)
- Ga-Young Song
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Seo-Yeon Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Mihee Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Jang Bae Moon
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Su Woong Yoo
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Seong Young Kwon
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Hee-Seung Bom
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Sae-Ryung Kang
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun,
Korea
| |
Collapse
|
38
|
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
|
39
|
Girum KB, Cottereau AS, Vercellino L, Rebaud L, Clerc J, Casasnovas O, Morschhauser F, Thieblemont C, Buvat I. Tumor Location Relative to the Spleen Is a Prognostic Factor in Lymphoma Patients: A Demonstration from the REMARC Trial. J Nucl Med 2024; 65:313-319. [PMID: 38071535 PMCID: PMC10858380 DOI: 10.2967/jnumed.123.266322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/23/2023] [Indexed: 02/03/2024] Open
Abstract
Baseline [18F]FDG PET/CT radiomic features can improve the survival prediction in patients with diffuse large B-cell lymphoma (DLBCL). The purpose of this study was to investigate whether characterizing tumor locations relative to the spleen location in baseline [18F]FDG PET/CT images predicts survival in patients with DLBCL and improves the predictive value of total metabolic tumor volume (TMTV) and age-adjusted international prognostic index (IPI). Methods: This retrospective study included 301 DLBCL patients from the REMARC (NCT01122472) cohort. Physicians delineated the tumor regions, whereas the spleen was automatically segmented using an open-access artificial intelligence algorithm. We systematically measured the distance between the centroid of the spleen and all other lesions, defining the SD of these distances as the lesion spread (SpreadSpleen). We calculated the maximum distance between the spleen and another lesion (Dspleen) for each patient and normalized it with the body surface area, resulting in standardized Dspleen (sDspleen). The predictive value of each PET/CT feature for progression-free survival (PFS) and overall survival (OS) was evaluated through univariate and multivariate time-dependent Cox models and Kaplan-Meier analysis. Results: In total, 282 patients (mean age, 68.33 ± 5.41 y; 164 men) were evaluated. The artificial intelligence algorithm successfully segmented the spleen in 96% of the patients. SpreadSpleen, Dspleen, and sDspleen were correlated neither with TMTV (Pearson ρ < 0.23) nor with IPI (Pearson ρ < 0.15). When median values were used as the cutoff, SpreadSpleen, Dspleen, and sDspleen all significantly classified patients into 2 risk groups for PFS and OS (P < 0.001). They complemented TMTV and IPI to classify the patients into 3 risk groups for PFS and OS (P < 0.001). Integrating SpreadSpleen, Dspleen, or sDspleen into a Cox model on the basis of TMTV, IPI, and TMTV combined with IPI significantly improved the concordance index for PFS and OS (P < 0.05). Conclusion: Baseline PET/CT features that characterize tumor spread and dissemination relative to the spleen strongly predicted survival in patients with DLBCL. Integrating these features with TMTV and IPI further improved survival prediction.
Collapse
Affiliation(s)
- Kibrom B Girum
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France
| | - Anne-Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, Paris Descartes University, Paris, France
| | | | - Louis Rebaud
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France
- Research and Clinical Collaborations, Siemens Medical Solutions USA, Knoxville, Tennessee
| | - Jérôme Clerc
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, Paris Descartes University, Paris, France
| | | | - Franck Morschhauser
- Research Group on Injectable Forms and Associated Technologies, Department of Hematology, Claude Huriez Hospital, University Lille, Lille, France; and
| | | | - Irène Buvat
- LITO Laboratory, U1288 Inserm, Institut Curie, University Paris-Saclay, Orsay, France;
| |
Collapse
|
40
|
Huang C, Tang TL, Qiu YY, Lin YP, Chen SL, Zhao RZ, Shi GQ, Liao SQ, Chen JH, Fu HY, Liu JZ, Xu BH, Liu TB, Yang Y. Hypofractionated radiotherapy for refractory or relapsed aggressive B-cell lymphoma in the rituximab era. BMC Cancer 2024; 24:72. [PMID: 38218811 PMCID: PMC10788030 DOI: 10.1186/s12885-024-11837-2] [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: 08/02/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is an effective and available local treatment for patients with refractory or relapsed (R/R) aggressive B-cell lymphomas. However, the value of hypofractionated RT in this setting has not been confirmed. METHODS We retrospectively analyzed patients with R/R aggressive B-cell lymphoma who received hypofractionated RT between January 2020 and August 2022 at a single institution. The objective response rate (ORR), overall survival (OS), progression-free survival (PFS) and acute side effects were analyzed. RESULTS A total of 30 patients were included. The median dose for residual disease was 36 Gy, at a dose per fraction of 2.3-5 Gy. After RT, the ORR and complete response (CR) rates were 90% and 80%, respectively. With a median follow-up of 10 months (range, 2-27 months), 10 patients (33.3%) experienced disease progression and three died. The 1-year OS and PFS rates for all patients were 81.8% and 66.3%, respectively. The majority (8/10) of post-RT progressions involved out-of-field relapses. Patients with relapsed diseases, no response to systemic therapy, multiple lesions at the time of RT, and no response to RT were associated with out-of-field relapses. PFS was associated with response to RT (P = 0.001) and numbers of residual sites (P < 0.001). No serious non-hematological adverse effects (≥ grade 3) associated with RT were reported. CONCLUSION These data suggest that hypofractionated RT was effective and tolerable for patients with R/R aggressive B-cell lymphoma, especially for those that exhibited localized residual disease.
Collapse
Affiliation(s)
- Cheng Huang
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Tian-Lan Tang
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Yan-Yan Qiu
- Department of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, P. R. China
| | - Yu-Ping Lin
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Si-Lin Chen
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Rui-Zhi Zhao
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Gui-Qing Shi
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Si-Qin Liao
- Department of PET/CT, Fujian Medical University Union Hospital, Fuzhou, P. R. China
| | - Jin-Hua Chen
- Follow-Up Center, Fujian Medical University Union Hospital, Fuzhou, P. R. China
| | - Hai-Ying Fu
- Department of Hematology, The Third Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, The Third People's Hospital of Fujian Province, Fuzhou, P. R. China
| | - Jian-Zhi Liu
- Department of Otorhinolaryngology, Fujian Medical University Union Hospital, Fuzhou, P. R. China
| | - Ben-Hua Xu
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China
| | - Ting-Bo Liu
- Department of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, P. R. China.
| | - Yong Yang
- Department of Radiation Oncology, Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies ), Fujian Medical University Union Hospital, Fuzhou, 350001, P. R. China.
| |
Collapse
|
41
|
Alderuccio JP, Reis IM, Hamadani M, Nachiappan M, Leslom S, Kahl BS, Ai WZ, Radford J, Solh M, Ardeshna KM, Hess BT, Lunning MA, Zinzani PL, Stathis A, Carlo-Stella C, Lossos IS, Caimi PF, Han S, Yang F, Kuker RA, Moskowitz CH. PET/CT Biomarkers Enable Risk Stratification of Patients with Relapsed/Refractory Diffuse Large B-cell Lymphoma Enrolled in the LOTIS-2 Clinical Trial. Clin Cancer Res 2024; 30:139-149. [PMID: 37855688 PMCID: PMC10872617 DOI: 10.1158/1078-0432.ccr-23-1561] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/11/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE Significant progress has occurred in developing quantitative PET/CT biomarkers in diffuse large B-cell lymphoma (DLBCL). Total metabolic tumor volume (MTV) is the most extensively studied, enabling assessment of FDG-avid tumor burden associated with outcomes. However, prior studies evaluated the outcome of cytotoxic chemotherapy or chimeric antigen receptor T-cell therapy without data on recently approved FDA agents. Therefore, we aimed to assess the prognosis of PET/CT biomarkers in patients treated with loncastuximab tesirine. EXPERIMENTAL DESIGN We centrally reviewed screening PET/CT scans of patients with relapsed/refractory DLBCL enrolled in the LOTIS-2 (NCT03589469) study. MTV was obtained by computing individual volumes using the SUV ≥4.0 threshold. Other PET/CT metrics, clinical factors, and the International Metabolic Prognostic Index (IMPI) were evaluated. Logistic regression was used to assess the association between biomarkers and treatment response. Cox regression was used to determine the effect of biomarkers on time-to-event outcomes. We estimated biomarker prediction as continuous and binary variables defined by cutoff points. RESULTS Across 138 patients included in this study, MTV with a cutoff point of 96 mL was the biomarker associated with the highest predictive performance in univariable and multivariable models to predict failure to achieve complete metabolic response (OR, 5.42; P = 0.002), progression-free survival (HR, 2.68; P = 0.002), and overall survival (HR, 3.09; P < 0.0001). IMPI demonstrated an appropriate performance, however, not better than MTV alone. CONCLUSIONS Pretreatment MTV demonstrated robust risk stratification, with those patients demonstrating high MTV achieving lower responses and survival to loncastuximab tesirine in relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Isildinha M. Reis
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mehdi Hamadani
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Muthiah Nachiappan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Salman Leslom
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Brad S. Kahl
- Washington University, St. Louis, MO, United States
| | - Weiyun Z. Ai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - John Radford
- NIHR Clinical Research Facility, University of Manchester and the Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Melhem Solh
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, United States
| | - Kirit M. Ardeshna
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Brian T. Hess
- Medical University of South Carolina, Charleston, SC, United States
| | - Matthew A. Lunning
- University of Nebraska Medical Center- Fred and Pamela Buffett Cancer Center, Omaha, NE, United States
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and Department of Oncology and Hematology, Humanitas Research Hospital–IRCCS, Milano, Italy
| | - Izidore S. Lossos
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Paolo F. Caimi
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States
| | - Sunwoo Han
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Fei Yang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Russ A. Kuker
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Craig H. Moskowitz
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
42
|
Ferdinandus J, van Heek L, Roth K, Dietlein M, Eich HT, Baues C, Borchmann P, Kobe C. Patterns of PET-positive residual tissue at interim restaging and risk of treatment failure in advanced-stage Hodgkin's lymphoma: an analysis of the randomized phase III HD18 trial by the German Hodgkin Study Group. Eur J Nucl Med Mol Imaging 2024; 51:490-495. [PMID: 37735258 PMCID: PMC10774157 DOI: 10.1007/s00259-023-06431-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Response-adapted treatment using early interim functional imaging with PET after two cycles of chemotherapy (PET-2) for advanced-stage Hodgkin's lymphoma (AS-HL) is the standard of care in several countries. However, the distribution of residual metabolic disease in PET-2 and the prognostic relevance of multiple involved regions have not been reported to date. METHODS We retrospectively analyzed data from all PET-2-positive patients included in HD18. Residual tissue was visually compared with reference regions according to the Deauville score (DS). PET-2 positivity was defined as residual tissue with uptake above the liver (DS4). PFS was defined as the time from staging until progression, relapse, or death from any cause, or to the day when information was last received on the patient's disease status and analyzed using Kaplan-Meier and Cox regressions. Comparisons were made between patients with 1-2 and >2 positive regions in PET-2 as well as patients without PET-2-positive regions randomized into comparator arms of HD18. RESULTS Between 2008 and 2014, 1964 patients with newly diagnosed AS-HL were recruited in HD18 and randomized following their PET-2 scan. Of these, 480 patients had a positive PET-2 and were eligible for this analysis. Upper and lower mediastinum in almost half of all patients: 230 (47.9%) and 195 (40.6%), respectively. 372 (77.5%) of patients have 1-2 positive regions in PET-2. 5y-PFS for patients with 1-2 regions was 91.7% (CI95: 88.7-94.6) vs. 81.8% (CI95: 74.2-90.1) for those with >2 regions with a corresponding hazard ratio (HR) of 2.2 (CI95: 1.2-4.0). Compared with patients without PET-2-positive disease receiving 6-8 cycles of chemotherapy, patients with 1-2 had a higher risk for a PFS event (HR 1.35; CI95 0.81-2.28), but it was not statistically significant (p=0.25). Patients with >2 PET-2-positive lesions had a significantly higher risk (HR 2.95; CI95: 1.62-5.37; p<0.001). CONCLUSION PET-2-positive residuals of AS-HL are mostly located in the mediastinum, and a majority of patients have few affected regions. The risk of progression was twofold higher in patients with more than two positive regions in PET-2.
Collapse
Affiliation(s)
- Justin Ferdinandus
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Gleueler Straße 269-273, 50935, Cologne, Germany.
- German Hodgkin Study Group (GHSG), Cologne, Germany.
| | - Lutz van Heek
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Katrin Roth
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Markus Dietlein
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Hans-Theodor Eich
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Christian Baues
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Radiotherapy and Cyberknife Center, University Hospital Cologne, Cologne, Germany
- Department of Radiooncology, Marienhospital Herne, Ruhr University Bochum, Bochum, Germany
| | - Peter Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Gleueler Straße 269-273, 50935, Cologne, Germany
- German Hodgkin Study Group (GHSG), Cologne, Germany
| | - Carsten Kobe
- German Hodgkin Study Group (GHSG), Cologne, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
43
|
Cottereau AS, Rebaud L, Trotman J, Feugier P, Nastoupil LJ, Bachy E, Flinn IW, Haioun C, Ysebaert L, Bartlett NL, Tilly H, Casasnovas O, Ricci R, Portugues C, Buvat I, Meignan M, Morschhauser F. Metabolic tumor volume predicts outcome in patients with advanced stage follicular lymphoma from the RELEVANCE trial. Ann Oncol 2024; 35:130-137. [PMID: 37898239 DOI: 10.1016/j.annonc.2023.10.121] [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: 07/15/2023] [Revised: 09/22/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND We investigated the prognostic value of baseline positron emission tomography (PET) parameters for patients with treatment-naïve follicular lymphoma (FL) in the phase III RELEVANCE trial, comparing the immunomodulatory combination of lenalidomide and rituximab (R2) versus R-chemotherapy (R-chemo), with both regimens followed by R maintenance therapy. PATIENTS AND METHODS Baseline characteristics of the entire PET-evaluable population (n = 406/1032) were well balanced between treatment arms. The maximal standard uptake value (SUVmax) and the standardized maximal distance between tow lesions (SDmax) were extracted, the standardized distance between two lesions the furthest apart, were extracted. The total metabolic tumor volume (TMTV) was computed using the 41% SUVmax method. RESULTS With a median follow-up of 6.5 years, the 6-year progression-free survival (PFS) was 57.8%, the median TMTV was 284 cm3, SUVmax was 11.3 and SDmax was 0.32 m-1, with no significant difference between arms. High TMTV (>510 cm3) and FLIPI were associated with an inferior PFS (P = 0.013 and P = 0.006, respectively), whereas SUVmax and SDmax were not (P = 0.08 and P = 0.12, respectively). In multivariable analysis, follicular lymphoma international prognostic index (FLIPI) and TMTV remained significantly associated with PFS (P = 0.0119 and P = 0.0379, respectively). These two adverse factors combined stratified the overall population into three risk groups: patients with no risk factors (40%), with one factor (44%), or with both (16%), with a 6-year PFS of 67.7%, 54.5%, and 41.0%, respectively. No significant interaction between treatment arms and TMTV or FLIPI (P = 0.31 or P = 0.59, respectively) was observed. The high-risk group (high TMTV and FLIPI 3-5) had a similar PFS in both arms (P = 0.45) with a median PFS of 68.4% in the R-chemo arm versus 71.4% in the R2 arm. CONCLUSIONS Baseline TMTV is predictive of PFS, independently of FLIPI, in patients with advanced FL even in the context of antibody maintenance.
Collapse
Affiliation(s)
- A S Cottereau
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, Université Paris Cité, Paris.
| | - L Rebaud
- LITO Laboratory, UMR 1288 Inserm, Institut Curie, Université Paris-Saclay, Orsay; Siemens Healthcare SAS, Saint Denis, France
| | - J Trotman
- Department of Hematology, Concord Repatriation General Hospital, University of Sydney, Sydney, Australia
| | - P Feugier
- Department of Hematology, University Hospital of Nancy and INSERM 1256 University of Lorraine, Vandœuvre-lès-Nancy, France
| | - L J Nastoupil
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E Bachy
- EA LIB (Lymphoma Immuno-Biology), University Claude Bernard Lyon 1, Lyon, France
| | - I W Flinn
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, USA
| | - C Haioun
- Lymphoïd Malignancies Unit, Henri Mondor Hospital, AP-HP, Créteil
| | - L Ysebaert
- Department of Hematology, IUC Toulouse-Oncopole Toulouse, Toulouse, France
| | - N L Bartlett
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, USA
| | - H Tilly
- Imaging Department, Centre Henri Becquerel, Rouen; QuantIF-LITIS, EA 4108, IRIB, University of Rouen, Rouen
| | - O Casasnovas
- Department of Hematology, F Mitterrand Hospital, Dijon; Inserm 1231, University of Dijon
| | - R Ricci
- LYSARC, Centre Hospitalier Lyon-Sud, Pierre-Bénite
| | - C Portugues
- LYSARC, Centre Hospitalier Lyon-Sud, Pierre-Bénite
| | - I Buvat
- LITO Laboratory, UMR 1288 Inserm, Institut Curie, Université Paris-Saclay, Orsay
| | - M Meignan
- Lysa Imaging, Henri Mondor University Hospital, AP-HP, University Paris East, Creteil
| | - F Morschhauser
- Department of Hematology, University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France
| |
Collapse
|
44
|
Ababneh HS, Ng AK, Abramson JS, Soumerai JD, Takvorian RW, Frigault MJ, Patel CG. Metabolic parameters predict survival and toxicity in chimeric antigen receptor T-cell therapy-treated relapsed/refractory large B-cell lymphoma. Hematol Oncol 2024; 42:e3231. [PMID: 37795759 DOI: 10.1002/hon.3231] [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: 06/05/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has revolutionized treatment for patients with relapsed/refractory large B-cell lymphoma (LBCL). However, data available concerning the impact of the prognostic value of quantitative 18F-fluorodeoxyglucose positron emission tomography-computed tomography (FDG PET/CT) parameters on the CAR T-related outcomes and toxicities are limited. Therefore, we aimed to evaluate the predictive value of pre- and post-CAR T metabolic parameters on survival and toxicities following CAR T-cell therapy. Fifty-nine patients with PET/CT scans done pre-and post-CAR T infusion were retrospectively identified and analyzed in a single institution database of LBCL patients treated with commercial CD19-targeted CAR T-cell therapy. The median follow-up was 10.7 months [interquartile range (IQR): 2.6-25.5 months]. The overall response (complete response-CR and partial response) and CR rates post-CAR T were 76% (n = 45) and 53% (n = 31), respectively. On univariate analysis, low pre-CAR T total lesion glycolysis (TLG) and metabolic tumor volume (MTV) predicted improved overall response post-CAR T (OR = 4.7, p = 0.01, OR = 9.5, p = 0.03, respectively) and CR post-CAR T (OR = 12.4, p = 0.0004, OR = 10.9, p = 0.0001, respectively). High TLG pre-CAR T was correlated with cytokine release syndrome (CRS, OR = 3.25, p = 0.04). High MTV pre-CAR T was correlated with developing immune effector cell neurotoxicity syndrome (ICANS) events (OR = 4.3, p = 0.01), and high SUV pre-CAR T was associated with grade 3-4 neurological events (OR = 12, p = 0.01). High MTV/TLG/SUVmax post-CAR T were significantly associated with inferior Overall survival (OS). On multivariate analysis, high TLG pre-CAR T (HR = 2.4, p = 0.03), age ≥60 (HR = 2.7, p = 0.03), and bulky disease (≥5 cm) at the time of apheresis (HR = 2.5, p = 0.02) were identified to be independent prognostic factors for inferior PFS. High MTV post-CAR T was identified as the most prognostic factor associated with inferior OS.
Collapse
Affiliation(s)
- Hazim S Ababneh
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea K Ng
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeremy S Abramson
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob D Soumerai
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronald W Takvorian
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew J Frigault
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chirayu G Patel
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Pitarch G, Rotstein Habarnau Y, Chirico R, Konowalik B, Pérez A, Valda A, Bastianello M. Exploring the applicability of a lesion segmentation method on [ 18F]fluorothymidine PET/CT images in diffuse large B-cell lymphoma. Eur J Hybrid Imaging 2023; 7:28. [PMID: 38143262 PMCID: PMC10749290 DOI: 10.1186/s41824-023-00184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/31/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND AND PURPOSE The determination of the total metabolic tumour volume based on [18F]fluorothymidine ([18F]FLT) PET/CT images in diffuse large B-cell lymphoma has a potential clinical value for detecting early relapse in this type of heterogeneous lymphoproliferative tumours. Tumour segmentation is a key step in this process. For this purpose, our objective was to determine a segmentation threshold of [18F]FLT PET/CT images, based on a reference tissue uptake, on a cohort of patients with diffuse large B-cell lymphoma (DLBCL) that have been scanned at different stages of the treatment. METHODS We enrolled 23 adult patients with DLBCL confirmed in II-IV stages without nervous system compromise. All patients were scanned using [18F]FLT PET/CT at the time of diagnosis (baseline PET), interim PET (iPET), and at the end of treatment (fPET). The administered activity was 1.8-2.6 MBq/kg body weight, performed 60-70 min after injection and without use of contrast-enhanced CT. First, we assessed the [18F]FLT uptake stability in liver and bone marrow along the patient follow-up. For the lesion segmentation, three threshold values were assessed. RESULTS Both, liver, and bone marrow can be indistinctly taken as reference tissue. The SUV threshold for a voxel to be considered as belonging to a lesion is expressed in terms of a percentage relative to the patient's uptake in the reference tissue. Found thresholds were: for liver, 62%, 33%, 27%; and for bone marrow, 35%, 21% and 22%, for baseline, iPET and fPET stages, respectively. The relative threshold throughout the treatment has a decreasing tendency along the stages. CONCLUSION Based on the results obtained with [18F]FLT PET/CT during staging and follow-up in patients with DLBCL, reference values were obtained for each stage referring to liver and bone marrow uptake that could be used in clinical practice oncology.
Collapse
Affiliation(s)
- Germán Pitarch
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Yamila Rotstein Habarnau
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Roxana Chirico
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Brenda Konowalik
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| | - Amalia Pérez
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Alejandro Valda
- Centro Universitario de Imágenes Médicas, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Buenos Aires, Argentina.
| | - María Bastianello
- Sección de Imágenes Moleculares y Terapia Metabólica, Hospital Universitario CEMIC, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
46
|
Johansson P, Alig S, Richter J, Hanoun C, Rekowski J, Dürig J, Ylstra B, de Jong D, Klapper W, Alizadeh AA, Dührsen U, Hüttmann A. Outcome prediction by interim positron emission tomography and IgM monoclonal gammopathy in diffuse large B-cell lymphoma. Ann Hematol 2023; 102:3445-3455. [PMID: 37566280 PMCID: PMC10640472 DOI: 10.1007/s00277-023-05393-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
In diffuse large B-cell lymphoma (DLBCL), a positive interim positron emission tomography (PET) scan predicts treatment failure, but the proportion of high-risk patients thus identified is small. To improve prediction, we combined the interim PET result with the presence or absence of an associated IgM gammopathy. Of 108 DLBCL patients participating in a prospective trial, nine (8%) were interim PET positive and 19 (18%) had an IgM gammopathy. The monoclonal protein was not associated with distinguishing genetic features, and its light chain restriction was not always concordant with the light chain restriction of the lymphoma. The information provided by interim PET and IgM gammopathy was combined to dichotomize the population into sizeable high-risk (1-2 adverse factors) and low-risk groups (no adverse factor) with widely different outcomes (population size, 25% vs. 75%; 3-year risk of progression, 51% vs. 10%; 3-year overall survival, 64% vs. 95%). Multivariable analyses including established risk factors revealed the interim PET result and the IgM gammopathy status to be the only factors significantly associated with outcome. Information about interim PET response and IgM gammopathy may be useful in studies testing risk-adapted treatment strategies.
Collapse
Affiliation(s)
- Patricia Johansson
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
- Institute of Cell Biology (Cancer Research), Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Stefan Alig
- Department of Medicine, Divisions of Oncology and Hematology, Stanford University, Stanford, CA, USA
| | - Julia Richter
- Department of Hematopathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Jan Rekowski
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Dürig
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daphne de Jong
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wolfram Klapper
- Department of Hematopathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ash A Alizadeh
- Department of Medicine, Divisions of Oncology and Hematology, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Institute for Stem Cell Biology & Regenerative Medicine, Stanford, CA, USA
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany.
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| |
Collapse
|
47
|
Wendler T, Kreissl MC, Schemmer B, Rogasch JMM, De Benetti F. Artificial Intelligence-powered automatic volume calculation in medical images - available tools, performance and challenges for nuclear medicine. Nuklearmedizin 2023; 62:343-353. [PMID: 37995707 PMCID: PMC10667065 DOI: 10.1055/a-2200-2145] [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: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Volumetry is crucial in oncology and endocrinology, for diagnosis, treatment planning, and evaluating response to therapy for several diseases. The integration of Artificial Intelligence (AI) and Deep Learning (DL) has significantly accelerated the automatization of volumetric calculations, enhancing accuracy and reducing variability and labor. In this review, we show that a high correlation has been observed between Machine Learning (ML) methods and expert assessments in tumor volumetry; Yet, it is recognized as more challenging than organ volumetry. Liver volumetry has shown progression in accuracy with a decrease in error. If a relative error below 10 % is acceptable, ML-based liver volumetry can be considered reliable for standardized imaging protocols if used in patients without major anomalies. Similarly, ML-supported automatic kidney volumetry has also shown consistency and reliability in volumetric calculations. In contrast, AI-supported thyroid volumetry has not been extensively developed, despite initial works in 3D ultrasound showing promising results in terms of accuracy and reproducibility. Despite the advancements presented in the reviewed literature, the lack of standardization limits the generalizability of ML methods across diverse scenarios. The domain gap, i. e., the difference in probability distribution of training and inference data, is of paramount importance before clinical deployment of AI, to maintain accuracy and reliability in patient care. The increasing availability of improved segmentation tools is expected to further incorporate AI methods into routine workflows where volumetry will play a more prominent role in radionuclide therapy planning and quantitative follow-up of disease evolution.
Collapse
Affiliation(s)
- Thomas Wendler
- Clinical Computational Medical Imaging Research, Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, Germany
- Institute of Digital Medicine, Universitätsklinikum Augsburg, Germany
- Computer-Aided Medical Procedures and Augmented Reality School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | | | | | - Julian Manuel Michael Rogasch
- Department of Nuclear Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin,Germany
| | - Francesca De Benetti
- Computer-Aided Medical Procedures and Augmented Reality School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| |
Collapse
|
48
|
Wang F, Cui S, Lu L, Shao X, Yan F, Liu Y, He B, Wang J, Cao Y, Yue Y, Wang Y, Gu W. Dissemination feature based on PET/CT is a risk factor for diffuse large B cell lymphoma patients outcome. BMC Cancer 2023; 23:1165. [PMID: 38030989 PMCID: PMC10687880 DOI: 10.1186/s12885-023-11333-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/24/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND 18F-FDG PET/CT provides precise information about dissemination of lymphoma lesions. Dmax, defined as distance between the two lesions that were farthest apart by PET/CT, was found to be a promising predictor of Diffuse large B-cell lymphoma (DLBCL) outcome in a small size of clinical trial data. We analyzed the impact of Dmax on the outcome of a large real-world DLBCL cohort. METHODS Data of newly diagnosed DLBCL at the Third Affiliated Hospital of Soochow University were retrospectively collected. Baseline Dmax, clinical data and survival information were recorded. A metabolic parameter, metabolic bulk volume (MBV), was also measured to verify the independent impact of Dmax. RESULTS Optimal cut-off values for Dmax and MBV were 45.34 cm and 21.65 cm3. With a median follow-up of 32 months, Dmax significantly impacted progression-free survival (PFS) and overall survival (OS) in 253 DLBCL patients. For Dmaxlow and Dmaxhigh groups, estimated 3-year OS were 87.0% and 53.8% (p < 0.001), while 3-year PFS were 77.3% and 37.3% (p < 0.001). And for MBVlow and MBVhighgroups, 3-year OS were 84.5% and 58.8% (p < 0.001), and 3-year PFS were 68.7% and 50.4% (p = 0.003). Multivariate analysis identified Dmax and Eastern Cooperative Oncology Group performance status (ECOG PS) independently associated with PFS and OS, while MBV only independently associated with OS. A Dmax revised prognostic index (DRPI) combining Dmax and ECOG PS identified an ultra-risk DLBCL population with 3-year PFS of 31.7% and 3-year OS of 38.5%. The area under the curve (AUC) showed that this model performed better than International prognostic Index (IPI). CONCLUSION Dmax is a new and promising indicator to investigate dissemination of lymphoma lesions associated with the outcome of DLBCL. It significantly contributes to stratification of patients with disparate outcomes. TRIAL REGISTRATION This research has been retrospectively registered in the Ethics Committee institutional of the Third Affiliated Hospital of Soochow University, and the registration number was approval No. 155 (approved date: 31 May 2022).
Collapse
Affiliation(s)
- Fei Wang
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Silu Cui
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Luo Lu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Feng Yan
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yaqi Liu
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Bai He
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jianfeng Wang
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yanhua Yue
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
49
|
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
|
50
|
Li M, Lu H, Fan J, Dai M, Su C. A nomogram prognostic model for diffuse large B-cell lymphoma based on SUVmax and GNRI in elderly patients. EJHAEM 2023; 4:1030-1041. [PMID: 38024603 PMCID: PMC10660607 DOI: 10.1002/jha2.794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 12/01/2023]
Abstract
To establish a nomogram for elderly patients with diffuse large B-cell lymphoma (DLBCL) based on nutritional and imaging features. The data of 221 elderly pretreatment DLBCL patients were retrospectively analyzed. All cases were randomly separated into the training group and validation group. A nomogram was built based on the results of multivariate analysis. A nomogram was established based on maximum standardized uptake value (SUVmax), geriatric nutritional risk index (GNRI), and lactate dehydrogenase. The concordance index (C-index) of the nomogram was 0.772 for the training group and 0.729 for the validation group, and similar results were found in the area under the curve (AUC). The calibration curve showed favorable consistency between prediction and real survival. The decision curve analysis (DCA) also showed that the nomogram had favorable clinical effectiveness. The new risk-stratification model divided patients into three groups with obvious survival. The C-index and AUCs for the new model were greater than those of IPI and NCCN-IPI. The DCA curve suggested that the new model had better clinical effectiveness than the IPI and NCCN-IPI. The nomogram prognostic model based on SUVmax and GNRI performed superior to NCCN-IPI and equal to IPI for risk stratification of elderly DLBCL patients.
Collapse
Affiliation(s)
- Maoqin Li
- Department of HematologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouP. R. China
| | - Haihao Lu
- Department of HematologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouP. R. China
| | - Jiaoyang Fan
- Department of HematologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouP. R. China
| | - Min Dai
- Department of HematologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouP. R. China
| | - Chang Su
- Department of HematologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouP. R. China
| |
Collapse
|