1
|
Novruzov E, Peters HA, Jannusch K, Kobbe G, Dietrich S, Fischer JC, Rox J, Antoch G, Giesel FL, Antke C, Baermann BN, Mamlins E. The predictive power of baseline metabolic and volumetric [ 18F]FDG PET parameters with different thresholds for early therapy failure and mortality risk in DLBCL patients undergoing CAR-T-cell therapy. Eur J Radiol Open 2025; 14:100619. [PMID: 39803388 PMCID: PMC11719856 DOI: 10.1016/j.ejro.2024.100619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025] Open
Abstract
Objective [18F]FDG imaging is an integral part of patient management in CAR-T-cell therapy for recurrent or therapy-refractory DLBCL. The calculation methods of predictive power of specific imaging parameters still remains elusive. With this retrospective study, we sought to evaluate the predictive power of the baseline metabolic parameters and tumor burden calculated with automated segmentation via different thresholding methods for early therapy failure and mortality risk in DLBCL patients. Materials and methods Eighteen adult patients were enrolled, who underwent CAR-T-cell therapy accompanied by at least one pretherapeutic and two posttherapeutic [18F]FDG PET scans within 30 and 90 days between December 2018 and October 2023. We performed single-click automatic segmentation within VOIs in addition to extracting the SUV parameters to calculate the MTVs and TLGs by applying thresholds based on the concepts of a fixed absolute threshold with an SUVmax > 4.0, a relative absolute threshold with an isocontour of > 40 % of the SUVmax, a background threshold involving the addition of the liver SUV value and its 2 SD values, and only the liver SUV value. Results For early therapy failure, baseline metabolic parameters such as the SUVmax, SUVpeak and SUVmean tended to have greater predictive power than did the baseline metabolic burden. However, the baseline metabolic burden was superior in the prediction of mortality risk regardless of the thresholding method used. Conclusion This study revealed that automated delineation methods of metabolic tumor burden using different thresholds do not differ in outcome substantially. Therefore, the current clinical standard with a fixed absolute threshold value of SUV > 4.0 seems to be a feasible option.
Collapse
Affiliation(s)
- Emil Novruzov
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Helena A. Peters
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Kai Jannusch
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Guido Kobbe
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Sascha Dietrich
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Johannes C. Fischer
- Institute for Transplantation Diagnostics and Cellular Therapy, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Jutta Rox
- Institute for Transplantation Diagnostics and Cellular Therapy, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Frederik L. Giesel
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
- Institute for Radiation Sciences, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| | - Ben-Niklas Baermann
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Eduards Mamlins
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf 40225, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Düsseldorf 40225, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany
| |
Collapse
|
2
|
Abenavoli EM, Linguanti F, Dercle L, Berti V, Lopci E. FDG-PET/CT Imaging in Chimeric Antigen Receptor-Engineered T-Cell Treatment in Patients with B-Cell Lymphoma: Current Evidence. PET Clin 2024; 19:505-513. [PMID: 38969565 DOI: 10.1016/j.cpet.2024.05.006] [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: 07/07/2024]
Abstract
The Food and Drug Administration and the European Medicines Agency have recently approved chimeric antigen receptor-engineered (CAR) T cells to treat several refractory/relapsed B-cell lymphomas. This comprehensive review aims to demonstrate the pivotal role that [18F]-FDG PET/computed tomographic (CT) imaging can play to enhance the care of patients treated with CAR T-cell therapy. To this end, this review deciphers evidence showing the diagnostic, prognostic, predictive, and theragnostic value of [18F]-FDG PET/CT-derived parameters.
Collapse
Affiliation(s)
| | - Flavia Linguanti
- Nuclear Medicine Department, Ospedale San Donato, Via Pietro Nenni 20, Arezzo 52100, Italy; Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, 622 West 168th Street, New York, NY 10032, USA
| | - Valentina Berti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Largo Brambilla 3, Florence 50134, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milano CAP 20089, Italy.
| |
Collapse
|
3
|
Ploch W, Sadowski K, Olejarz W, Basak GW. Advancement and Challenges in Monitoring of CAR-T Cell Therapy: A Comprehensive Review of Parameters and Markers in Hematological Malignancies. Cancers (Basel) 2024; 16:3339. [PMID: 39409959 PMCID: PMC11475293 DOI: 10.3390/cancers16193339] [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: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment for relapsed/refractory B-cell lymphomas. Despite its success, this therapy is accompanied by a significant frequency of adverse events, including cytokine release syndrome (CRS), immune-effector-cell-associated neurotoxicity syndrome (ICANS), or cytopenias, reaching even up to 80% of patients following CAR-T cell therapy. CRS results from the uncontrolled overproduction of proinflammatory cytokines, which leads to symptoms such as fever, headache, hypoxia, or neurological complications. CAR-T cell detection is possible by the use of flow cytometry (FC) or quantitative polymerase chain reaction (qPCR) assays, the two primary techniques used for CAR-T evaluation in peripheral blood, bone marrow (BM), and cerebrospinal fluid (CSF). State-of-the-art imaging technologies play a crucial role in monitoring the distribution and persistence of CAR-T cells in clinical trials. Still, they can also be extended with the use of FC and digital PCR (dPCR). Monitoring the changes in cell populations during disease progression and treatment gives an important insight into how the response to CAR-T cell therapy develops on a cellular level. It can help improve the therapeutic design and optimize CAR-T cell therapy to make it more precise and personalized, which is crucial to overcoming the problem of tumor relapse.
Collapse
Affiliation(s)
- Weronika Ploch
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
4
|
Al-Ibraheem A, Abdlkadir AS, Al-Adhami DA, Sathekge M, Bom HHS, Ma’koseh M, Mansour A, Abdel-Razeq H, Al-Rabi K, Estrada-Lobato E, Al-Hussaini M, Matalka I, Abdel Rahman Z, Fanti S. The prognostic utility of 18F-FDG PET parameters in lymphoma patients under CAR-T-cell therapy: a systematic review and meta-analysis. Front Immunol 2024; 15:1424269. [PMID: 39286245 PMCID: PMC11402741 DOI: 10.3389/fimmu.2024.1424269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Background Chimeric antigen receptor (CAR) T-cell therapy has attracted considerable attention since its recent endorsement by the Food and Drug Administration, as it has emerged as a promising immunotherapeutic modality within the landscape of oncology. This study explores the prognostic utility of [18F]Fluorodeoxyglucose positron emission tomography ([18F]FDG PET) in lymphoma patients undergoing CAR T-cell therapy. Through meta-analysis, pooled hazard ratio (HR) values were calculated for specific PET metrics in this context. Methods PubMed, Scopus, and Ovid databases were explored to search for relevant topics. Dataset retrieval from inception until March 12, 2024, was carried out. The primary endpoints were impact of specific PET metrics on overall survival (OS) and progression-free survival (PFS) before and after treatment. Data from the studies were extracted for a meta-analysis using Stata 17.0. Results Out of 27 studies identified for systematic review, 15 met the criteria for meta-analysis. Baseline OS analysis showed that total metabolic tumor volume (TMTV) had the highest HR of 2.66 (95% CI: 1.52-4.66), followed by Total-body total lesion glycolysis (TTLG) at 2.45 (95% CI: 0.98-6.08), and maximum standardized uptake values (SUVmax) at 1.30 (95% CI: 0.77-2.19). TMTV and TTLG were statistically significant (p < 0.0001), whereas SUVmax was not (p = 0.33). For PFS, TMTV again showed the highest HR at 2.65 (95% CI: 1.63-4.30), with TTLG at 2.35 (95% CI: 1.40-3.93), and SUVmax at 1.48 (95% CI: 1.08-2.04), all statistically significant (p ≤ 0.01). The ΔSUVmax was a significant predictor for PFS with an HR of 2.05 (95% CI: 1.13-3.69, p = 0.015). Conclusion [18F]FDG PET parameters are valuable prognostic tools for predicting outcome of lymphoma patients undergoing CAR T-cell therapy.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Ahmed Saad Abdlkadir
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Dhuha Ali Al-Adhami
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Henry Hee-Seung Bom
- Department of Nuclear Medicine, Chonnam National University Medical School (CNUMS) and Hospital, Gwangju, Republic of Korea
| | - Mohammad Ma’koseh
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Asem Mansour
- Department of Diagnostic Radiology, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Kamal Al-Rabi
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Enrique Estrada-Lobato
- Nuclear Medicine and Diagnostic Section, Division of Human Health, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Maysaa Al-Hussaini
- Department of Pathology, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Ismail Matalka
- Department of Pathology and Microbiology, King Abdullah University Hospital- Jordan University of Science and Technology, Irbid, Jordan
- Department of Pathology, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Zaid Abdel Rahman
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Stephano Fanti
- Nuclear Medicine Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
5
|
Wielenberg CF, Fostitsch JC, Volz C, Marks R, Michalski K, Wäsch R, Zeiser R, Ruf J, Meyer PT, Klein C. FDG-PET/CT is a powerful tool to predict and evaluate response to chimeric antigen receptor (CAR) T-cell therapy in Non-Hodgkin-Lymphoma (NHL). Nuklearmedizin 2024; 63:252-258. [PMID: 38593856 DOI: 10.1055/a-2283-8417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has dramatically shifted the landscape of treatment especially for Non-Hodgkin-Lymphoma (NHL). This study evaluates the role of fluorodeoxyglucose (FDG)-positron emission tomography/computed tomography (PET/CT) in NHL treated with CAR T-cell therapy concerning response assessment and prognosis.We evaluated 34 patients with NHL who received a CAR T-cell therapy between August 2019 and July 2022. All patients underwent a pre-therapeutic FDG-PET/CT (PET-0) 6 days prior and a post-therapeutic FDG-PET/CT (PET-1) 34 days after CAR T-cell therapy. Deauville score (DS) was used for evaluation of response to therapy and compared to a minimum follow-up of 5 months.19/34 (55.9%) patients achieved DS ≤ 3 on PET-1, the remaining 15 (44.1%) patients had DS > 3 on PET-1. 14/19 patients with DS ≤ 3 on PET-1 had no relapsed or refractory (r/r)-disease and were still alive at last follow-up. The other 5 patients had r/r-disease and 4 of these died. Except for two patients who had no r/r-disease, all other patients (13/15) with DS > 3 on PET-1 had r/r-disease and 12 of these subsequently died. Patients with DS ≤ 3 on PET-1 had significantly better progression free survival (PFS; HR: 5.7; p < 0.01) and overall survival (OS; HR: 5.0; p < 0.01) compared to patients with DS > 3 on PET-1. In addition, we demonstrated that patients with DS ≤ 4 on PET-0 tended to have longer PFS (HR: 3.6; p = 0.05).Early FDG-PET/CT using the established DS after CAR T-cell therapy is a powerful tool to evaluate response to therapy.
Collapse
Affiliation(s)
| | | | - Christian Volz
- Department of Nuclear Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Reinhard Marks
- Department of Internal Medicine I, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Ralph Wäsch
- Department of Internal Medicine I, University of Freiburg, Freiburg im Breisgau, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, University of Freiburg, Freiburg im Breisgau, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Claudius Klein
- Department of Nuclear Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
6
|
Eigendorff F, Filimonova I, Scholl S, Sayer-Klink A, Rummler S, Kunert C, Pietschmann K, Wittig A, Hochhaus A, Schnetzke U. Effective bridging strategies prior to infusion with tisagenlecleucel results in high response rates and long-term remission in relapsed/refractory large B-cell lymphoma: findings from a German monocentric study. J Cancer Res Clin Oncol 2024; 150:224. [PMID: 38693452 PMCID: PMC11062962 DOI: 10.1007/s00432-024-05765-8] [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: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Incorporating chimeric antigen receptor (CAR)-T cell therapy into relapsed or refractory large B-cell lymphoma (rr LBCL) treatment algorithms has yielded remarkable response rates and durable remissions, yet a substantial portion of patients experience progression or relapse. Variations in outcomes across treatment centers may be attributed to different bridging strategies and remission statuses preceding CAR-T cell therapy. PATIENTS Twenty-nine consecutive adult patients receiving tisagenlecleucel (tisa-cel) for rr LBCL from December 2019 to February 2023 at Jena University Hospital were analyzed. RESULTS The median age was 63, with a median of 3 prior treatments. Twenty patients (69%) were refractory to any systemic therapy before CAR-T cell treatment. Following leukapheresis, 25 patients (86%) received bridging therapy with the majority undergoing chemotherapy (52%) or combined modality therapy (32%). Radiotherapy (RT) was part of the bridging strategy in 44%, with moderately hypofractionated involved site RT (30.0 Gy/2.5 Gy) being applied most frequently (64%). Post-CAR-T infusion, the objective response rate at 30 days was 83%, with 55% achieving complete response. Twelve-month progression-free (PFS) and overall survival (OS) were 60% and 74%, respectively, with a median follow up of 11.1 months for PFS and 17.9 months for OS. Factors significantly associated with PFS were chemotherapy sensitivity pre-leukapheresis and response to bridging. CONCLUSION The study underscores the importance of minimal tumor burden at CAR-T initiation, emphasizing the need for suitable bridging regimens. The findings advocate for clinical trials and further real-world analyses to optimize CAR-T cell therapy outcomes by identifying the most effective bridging strategies.
Collapse
MESH Headings
- Humans
- Male
- Middle Aged
- Female
- Aged
- Immunotherapy, Adoptive/methods
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Adult
- Remission Induction
- Neoplasm Recurrence, Local/therapy
- Neoplasm Recurrence, Local/pathology
- Germany
- Receptors, Antigen, T-Cell/therapeutic use
- Retrospective Studies
- Combined Modality Therapy
Collapse
Affiliation(s)
- Farina Eigendorff
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Irina Filimonova
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Sebastian Scholl
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Anne Sayer-Klink
- Institut für Transfusionsmedizin, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Silke Rummler
- Institut für Transfusionsmedizin, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Christa Kunert
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Klaus Pietschmann
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Andrea Wittig
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Klinik und Poliklinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Andreas Hochhaus
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Ulf Schnetzke
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany.
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany.
| |
Collapse
|
7
|
Marcus C. Editorial Comment: FDG PET/CT Has Impact in Chimeric Antigen Receptor T-Cell Therapy Response Assessment and Outcome Prediction. AJR Am J Roentgenol 2024; 222:e2330703. [PMID: 38117102 DOI: 10.2214/ajr.23.30703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
|
8
|
Alderuccio JP, Kuker RA, Yang F, Moskowitz CH. Quantitative PET-based biomarkers in lymphoma: getting ready for primetime. Nat Rev Clin Oncol 2023; 20:640-657. [PMID: 37460635 DOI: 10.1038/s41571-023-00799-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 08/20/2023]
Abstract
The use of functional quantitative biomarkers extracted from routine PET-CT scans to characterize clinical responses in patients with lymphoma is gaining increased attention, and these biomarkers can outperform established clinical risk factors. Total metabolic tumour volume enables individualized estimation of survival outcomes in patients with lymphoma and has shown the potential to predict response to therapy suitable for risk-adapted treatment approaches in clinical trials. The deployment of machine learning tools in molecular imaging research can assist in recognizing complex patterns and, with image classification, in tumour identification and segmentation of data from PET-CT scans. Initial studies using fully automated approaches to calculate metabolic tumour volume and other PET-based biomarkers have demonstrated appropriate correlation with calculations from experts, warranting further testing in large-scale studies. The extraction of computer-based quantitative tumour characterization through radiomics can provide a comprehensive view of phenotypic heterogeneity that better captures the molecular and functional features of the disease. Additionally, radiomics can be integrated with genomic data to provide more accurate prognostic information. Further improvements in PET-based biomarkers are imminent, although their incorporation into clinical decision-making currently has methodological shortcomings that need to be addressed with confirmatory prospective validation in selected patient populations. In this Review, we discuss the current knowledge, challenges and opportunities in the integration of quantitative PET-based biomarkers in clinical trials and the routine management of patients with lymphoma.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Russ A Kuker
- Department of Radiology, Division of Nuclear Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fei Yang
- Department of Radiation Oncology, Division of Medical Physics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Craig H Moskowitz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Jiménez-Ubieto A, Martín-Muñoz A, Poza M, Dorado S, García-Ortiz A, Revilla E, Sarandeses P, Ruiz-Heredia Y, Baumann T, Rodríguez A, Calbacho M, Sánchez PM, Pina JMS, García-Sancho AM, Figaredo G, Gil-Alós D, Rufián L, Rodríguez M, Carneros L, Martínez-Laperche C, Bastos-Oreiro M, Wang C, Cedena MT, Rapado I, de Toledo P, Gallardo M, Valeri A, Ayala R, Martínez-López J, Barrio S. Personalized monitoring of circulating tumor DNA with a specific signature of trackable mutations after chimeric antigen receptor T-cell therapy in follicular lymphoma patients. Front Immunol 2023; 14:1188818. [PMID: 37342332 PMCID: PMC10277746 DOI: 10.3389/fimmu.2023.1188818] [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: 03/17/2023] [Accepted: 04/26/2023] [Indexed: 06/22/2023] Open
Abstract
Background CART therapy has produced a paradigm shift in the treatment of relapsing FL patients. Strategies to optimize disease surveillance after these therapies are increasingly necessary. This study explores the potential value of ctDNA monitoring with an innovative signature of personalized trackable mutations. Method Eleven FL patients treated with anti-CD19 CAR T-cell therapy were included. One did not respond and was excluded. Genomic profiling was performed before starting lymphodepleting chemotherapy to identify somatic mutations suitable for LiqBio-MRD monitoring. The dynamics of the baseline mutations (4.5 per patient) were further analyzed on 59 cfDNA follow-up samples. PET/CT examinations were performed on days +90, +180, +365, and every six months until disease progression or death. Results After a median follow-up of 36 months, all patients achieved a CR as the best response. Two patients progressed. The most frequently mutated genes were CREBBP, KMT2D and EP300. Simultaneous analysis of ctDNA and PET/CT was available for 18 time-points. When PET/CT was positive, two out of four ctDNA samples were LiqBio-MRD negative. These two negative samples corresponded to women with a unique mesenteric mass in two evaluations and never relapsed. Meanwhile, 14 PET/CT negative images were mutation-free based on our LiqBio-MRD analysis (100%). None of the patients had a negative LiqBio-MRD test by day +7. Interestingly, all durably responding patients had undetectable ctDNA at or around three months after infusion. Two patients presented discordant results by PET/CT and ctDNA levels. No progression was confirmed in these cases. All the progressing patients were LiqBio-MRD positive before progression. Conclusion This is a proof-of-principle for using ctDNA to monitor response to CAR T-cell therapy in FL. Our results confirm that a non-invasive liquid biopsy MRD analysis may correlate with response and could be used to monitor response. Harmonized definitions of ctDNA molecular response and pinpointing the optimal timing for assessing ctDNA responses are necessary for this setting. If using ctDNA analysis, we suggest restricting follow-up PET/CT in CR patients to a clinical suspicion of relapse, to avoid false-positive results.
Collapse
Affiliation(s)
- Ana Jiménez-Ubieto
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Alejandro Martín-Muñoz
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - María Poza
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Sara Dorado
- Altum sequencing Co., Madrid, Spain
- Computational Science Department, Carlos III University, Madrid, Spain
| | - Almudena García-Ortiz
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Enrique Revilla
- Departamento de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pilar Sarandeses
- Departamento de Medicina Nuclear, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Yanira Ruiz-Heredia
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Tycho Baumann
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Antonia Rodríguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - María Calbacho
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Pilar Martínez Sánchez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - José María Sánchez Pina
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | | | - Gloria Figaredo
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Daniel Gil-Alós
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Laura Rufián
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Margarita Rodríguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Laura Carneros
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | | | | | - Chongwu Wang
- Hosea Precision Medical Technology Co., Ltd., Weihai, Shangdong, China
| | - María-Teresa Cedena
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Inmaculada Rapado
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Paula de Toledo
- Computational Science Department, Carlos III University, Madrid, Spain
| | - Miguel Gallardo
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Antonio Valeri
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Rosa Ayala
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Joaquín Martínez-López
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Santiago Barrio
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| |
Collapse
|
10
|
Zinzi A, Gaio M, Liguori V, Cagnotta C, Paolino D, Paolisso G, Castaldo G, Nicoletti G, Rossi F, Capuano A, Rafaniello C. Late relapse after CAR-T cell therapy for adult patients with hematologic malignancies: a definite evidence from Systematic Review and Meta-Analysis on individual data. Pharmacol Res 2023; 190:106742. [PMID: 36963592 DOI: 10.1016/j.phrs.2023.106742] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
Chimeric Antigen Receptor (CAR)-modified T lymphocytes represent one of the most innovative and promising approaches to treating hematologic malignancies. CAR-T cell therapy is currently being used for the treatment of relapsed/refractory (r/r) B-cell malignancies including Acute Lymphoblastic Leukemia, Large B-Cell Lymphoma, Follicular Lymphoma, Multiple Myeloma and Mantle Cell Lymphoma. Despite the unprecedented clinical success, one of the major issues of the approved CAR-T cell therapy - tisagenlecleucel, axicabtagene, lisocabtagene, idecabtagene, ciltacabtagene and brexucabtagene - is the uncertainty about its persistence which in turn could lead to weak or no response to therapy with malignancy recurrence. Here we show that the prognosis of patients who do not respond to CAR-T cell therapy is still an unmet medical need. We performed a systematic review and meta-analysis collecting individual data on Duration of Response from at least 12-month follow-up studies. We found that the pooled prevalence of relapse within the first 12 months after CAR-T infusion was 61% (95% CI, 43%-78%); moreover, one year after the infusion, the analysis highlighted a pooled prevalence of relapse of 24% (95% CI, 11%-42%). Our results suggest that identifying potential predictive biomarkers of response to CAR-T therapy, especially for patients affected by the advanced stage of blood malignancies, could lead to stratification of the eligible population to that therapy, recognizing which patients will benefit and which will not, helping regulators to make decision in that way.
Collapse
Affiliation(s)
- Alessia Zinzi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Mario Gaio
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Valerio Liguori
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Cecilia Cagnotta
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Viale Europa s.n.c., I-88100 Catanzaro, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138 Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Gianfranco Nicoletti
- Department of Imaging, University of Campania "Luigi Vanvitelli", Breast Unit, Multidisciplinary Department of Medical-Surgical and Dental Specialties, Naples, Italy
| | - Francesco Rossi
- Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| |
Collapse
|