1
|
Paiva B, Shi Q, Puig N, Cedena MT, Orfao A, Durie BGM, Munshi NC, San-Miguel J. Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma. Nat Rev Clin Oncol 2025; 22:424-438. [PMID: 40195455 DOI: 10.1038/s41571-025-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
Measurable residual disease (MRD) assessment is, from the methodological point of view, ready for prime time in multiple myeloma (MM). Abundant evidence underscores the value of MRD status determined using highly sensitive next-generation flow cytometry and next-generation sequencing tests in evaluating response to treatment and, therefore, prognosis in patients with this disease. MRD response assessment and monitoring might present a range of opportunities for individualized patient management. Moreover, the considerable amounts of high-quality and standardized MRD data generated in clinical trials have led to the acceptance of MRD negativity as an early end point for accelerated regulatory approval of treatments for MM. The data leave no doubt that the efficacy of new regimens in inducing deeper and durable MRD-negative responses is connected with prolonged survival. Yet, several evidential, technical and practical challenges continue to limit the implementation of MRD-guided treatment strategies in routine practice, and the use of MRD as a surrogate end point remains controversial to some. In this Review, we draw on past and present research to propose opportunities for overcoming some of these challenges, and to accelerate the use of MRD assessment for improved clinical management of patients with MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain.
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Noemi Puig
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00233, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Maria-Teresa Cedena
- Instituto de Investigación imas12, CIBER-ONC number CB16/12/00369, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service, Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00400, University of Salamanca, Salamanca, Spain
| | - Brian G M Durie
- Division of Hematology/Oncology, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| | - Nikhil C Munshi
- Basic and Correlative Science, Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, VA Boston Healthcare System, Boston, MA, USA
| | - Jesús San-Miguel
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Aljama MA, Sidiqi HM, Gertz MA. Are we maintaining minimal residual disease in myeloma? Leuk Lymphoma 2025; 66:1001-1009. [PMID: 39835888 DOI: 10.1080/10428194.2025.2455485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Minimal residual disease (MRD) has emerged as an important prognostic maker in patients with multiple myeloma at different stages of their treatment. Moreover, it is being increasingly incorporated as an endpoint in various clinical trials. Since maintenance therapy is an integral part of myeloma treatment, especially in the upfront setting post autologous transplantation, it is imperative to understand the role of MRD testing in the maintenance stetting. This review aims to examine the utility and dynamics of MRD testing in order to elucidate its prognostic role and possible incorporation in clinical decision making processes.
Collapse
Affiliation(s)
| | - Hasib M Sidiqi
- Hematology Department, Fiona Stanley Hospital, Perth Western, Australia
- Curtin Medical School, Curtin University, Perth Western, Australia
| | - Morie A Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Kaur H, Kumar S, Watts A, Singh C, Sachdeva MUS, Sreedharanunni S, Kumar R, Malhotra P, Singh B. 68 Ga-Pentixafor PET/CT-Based Response Evaluation and its Prognostic Value in Multiple Myeloma: Comparison With IMWG and 18 F-FDG-Based Response. Clin Nucl Med 2025; 50:e331-e339. [PMID: 40051087 DOI: 10.1097/rlu.0000000000005731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/20/2024] [Indexed: 05/06/2025]
Abstract
PURPOSE 68 Ga-Pentixafor PET/CT targets CXCR4 receptors and provides superior diagnostic accuracy in multiple myeloma (MM) compared with 18 F-FDG PET/CT. However, its role in response evaluation remains unexplored. We propose a 68 Ga-Pentixafor PET/CT-based response evaluation criterion and evaluate its utility compared with International Myeloma Working Group (IMWG) criteria and 18 F-FDG PET/CT-based response. PATIENTS AND METHODS In this prospective single-center study, 40 treatment-naive myeloma patients were recruited between February 2021 and April 2023. Both 68 Ga-Pentixafor and 18 F-FDG PET/CT were performed at baseline and at follow-up (7.2 mo-median). Response to treatment was evaluated using the proposed 68 Ga-Pentixafor PET/CT criteria and compared with responses assessed by IMWG and 18 F-FDG PET/CT. Progression-free survival (PFS) and overall survival (OS) were analyzed and compared using Kaplan-Meier survival curves. RESULTS Among the 40 newly diagnosed MM patients [median age: 56.5 years (IQR 45.25 to 63.75); 24 men], 68 Ga-Pentixafor PET/CT was positive in a greater proportion of patients than 18 F-FDG PET/CT [90% (36/40) vs. 67.5% (27/40); P =0.02] thus, adequately evaluated response in additional 27.5% (11/40) of cases. Using the proposed criteria for 68 Ga-Pentixafor PET/CT, significant differences in PFS were observed across response categories [complete response (CR)-not reached, partial response (PR)-26.2 mo, progressive disease (PD)-15.3 mo; P =0.001]. Among patients achieving ≥very good partial response (VGPR) as per IMWG, those with positive 68 Ga-Pentixafor PET/CT had shorter PFS compared with those with negative findings (median PFS: 34.2 mo vs. not reached; P =0.056), whereas no significant difference was noted with 18 F-FDG PET/CT ( P =0.68). In addition, on follow-up of patients with negative 18 F-FDG at the response, those with discordant 68 Ga-Pentixafor findings had significantly shorter PFS (17.73 mo vs. not reached; P =0.010) compared with those with concordant negative findings. CONCLUSIONS 68 Ga-Pentixafor PET/CT offers a more accurate assessment of treatment response and prognosis in MM patients, adding valuable information beyond the IMWG and 18 F-FDG PET/CT-based criteria.
Collapse
Affiliation(s)
| | | | | | | | | | - Sreejesh Sreedharanunni
- Hematology, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | | | | |
Collapse
|
4
|
Sachpekidis C, Goldschmidt H, Edenbrandt L, Dimitrakopoulou-Strauss A. Radiomics and Artificial Intelligence Landscape for [ 18F]FDG PET/CT in Multiple Myeloma. Semin Nucl Med 2025; 55:387-395. [PMID: 39674756 DOI: 10.1053/j.semnuclmed.2024.11.005] [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: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024]
Abstract
[18F]FDG PET/CT is a powerful imaging modality of high performance in multiple myeloma (MM) and is considered the appropriate method for assessing treatment response in this disease. On the other hand, due to the heterogeneous and sometimes complex patterns of bone marrow infiltration in MM, the interpretation of PET/CT can be particularly challenging, hampering interobserver reproducibility and limiting the diagnostic and prognostic ability of the modality. Although many approaches have been developed to address the issue of standardization, none can yet be considered a standard method for interpretation or objective quantification of PET/CT. Therefore, advanced diagnostic quantification approaches are needed to support and potentially guide the management of MM. In recent years, radiomics has emerged as an innovative method for high-throughput mining of image-derived features for clinical decision making, which may be particularly helpful in oncology. In addition, machine learning and deep learning, both subfields of artificial intelligence (AI) closely related to the radiomics process, have been increasingly applied to automated image analysis, offering new possibilities for a standardized evaluation of imaging modalities such as CT, PET/CT and MRI in oncology. In line with this, the initial but steadily growing literature on the application of radiomics and AI-based methods in the field of [18F]FDG PET/CT in MM has already yielded encouraging results, offering a potentially reliable tool towards optimization and standardization of interpretation in this disease. The main results of these studies are presented in this review.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Hartmut Goldschmidt
- Internal Medicine V, Hematology, Oncology and Rheumatology, German-Speaking Myeloma Multicenter Group (GMMG), Heidelberg University Hospital, Heidelberg, Germany
| | - Lars Edenbrandt
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
5
|
Freeman CL, Noble J, Menges M, Villanueva R, Nakashima JY, Figura NB, Tonseth RP, Werner Idiaquez D, Skelson L, Smith E, Abraham-Miranda J, Corallo S, De Avila G, Castaneda Puglianini OA, Liu H, Alsina M, Nishihori T, Shain KH, Baz R, Blue B, Grajales-Cruz A, Koomen JM, Atkins RM, Hansen DK, S Silva A, Kim J, Balagurunathan Y, Locke FL. Tumor burden quantified by soluble B-cell maturation antigen and metabolic tumor volume determines myeloma CAR-T outcomes. Blood 2025; 145:1645-1657. [PMID: 39652773 DOI: 10.1182/blood.2024024965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/24/2024] [Indexed: 04/11/2025] Open
Abstract
ABSTRACT Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a breakthrough treatment for relapsed and refractory multiple myeloma (RRMM). However, these products are complex to deliver, and alternative options are now available. Identifying biomarkers that can predict therapeutic outcomes is crucial for optimizing patient selection. There is a paucity of data evaluating the utility of both serum soluble B-cell maturation antigen (sBCMA) levels and metabolic tumor volume (MTV) at baseline in patients with RRMM undergoing CAR-T therapy. We identified a cohort of 183 patients with available serum to measure sBCMA and/or pretreatment MTV, derived from positron emission tomography-computed tomography scans obtained per standard of care. Expectedly, high pretreatment levels of sBCMA correlated with other established markers of tumor burden (eg, bone marrow plasma cells and β2 microglobulin) and inflammation and were highly prognostic for CAR-T-related toxicities and inferior progression-free survival (PFS). High MTV values were also associated with shorter PFS and inferior overall survival. The poor correlation observed between these 2 measures prompted evaluation of those with discordant results, identifying that those with low sBCMA and high MTV frequently had low/absent BCMA expression on plasma cells and suboptimal response. Our findings highlight the potential utility of sBCMA and MTV to facilitate more personalized treatment strategies in the management of RRMM eligible for BCMA-directed CAR-T.
Collapse
Affiliation(s)
- Ciara L Freeman
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Jerald Noble
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Meghan Menges
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | | | - Justyn Y Nakashima
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Nicholas B Figura
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | | | | | - Lawrence Skelson
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Eric Smith
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Julieta Abraham-Miranda
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Salvatore Corallo
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Gabriel De Avila
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Omar A Castaneda Puglianini
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Hien Liu
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Melissa Alsina
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Kenneth H Shain
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Rachid Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Brandon Blue
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Ariel Grajales-Cruz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - John M Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Reginald M Atkins
- Department of Clinical Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Doris K Hansen
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Ariosto S Silva
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Yoganand Balagurunathan
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
6
|
Talarico M, Barbato S, Cattabriga A, Sacchetti I, Manzato E, Restuccia R, Masci S, Bigi F, Puppi M, Iezza M, Rizzello I, Mancuso K, Pantani L, Tacchetti P, Nanni C, Cavo M, Zamagni E. Diagnostic Innovations: Advances in imaging techniques for diagnosis and follow-up of multiple myeloma. J Bone Oncol 2025; 51:100669. [PMID: 40124904 PMCID: PMC11930372 DOI: 10.1016/j.jbo.2025.100669] [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: 12/21/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction The International Myeloma Working Group (IMWG) defines myeloma related bone disease (MBD) as a diagnostic criterion for symptomatic multiple myeloma (MM) as the presence of osteolytic lesions ≥ 5 mm or more than one focal lesion (FL) ≥ 5 mm by magnetic resonance imaging (MRI). Whole-body low-dose CT (WBLDCT) is recommended as the first-choice imaging technique for the diagnosis of MBD with 18F-fluorodeoxyglucose-positron emission tomography/CT (18F-FDG-PET/CT) being considered a possible alternative at staging, whereas use of MRI studies is recommended in cases without myeloma-defining events (MDEs) in order to exclude the presence of FLs. Furthermore, use of 18F-FDG-PET/CT is recommended in response assessment, to be integrated with hematologic response and bone marrow minimal residual disease (MRD). Areas covered In this paper, we review novel functional imaging techniques in MM, particularly focusing on their advantages, limits, applications and comparisons with 18F-FDG-PET/CT or other standardized imaging techniques. Conclusions Combining both morphological and functional imaging, 18F-FDG-PET/CT is currently considered a standard imaging technique in MM for staging (despite false positive or negative results) and response assessment. The introduction of novel functional imaging techniques, as whole-body diffusion-weighted magnetic resonance imaging (WB-DWI-MRI), or novel PET tracers might be useful in overcoming these limits. Future studies will give more information on the complementarity of these imaging techniques or whether one of them might become a new gold standard in MM.
Collapse
Affiliation(s)
- M. Talarico
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - S. Barbato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - A. Cattabriga
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna 40138 Bologna, Italy
| | - I. Sacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - E. Manzato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - R. Restuccia
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - S. Masci
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - F. Bigi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - M. Puppi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - M. Iezza
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - I. Rizzello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - K. Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - L. Pantani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - P. Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - C. Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - M. Cavo
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - E. Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| |
Collapse
|
7
|
Quezada-Ramírez SI, Del Carmen Tarín-Arzaga L, Gómez-De León A, Gómez-Almaguer D. Novel drug combinations for newly diagnosed multiple myeloma: how can we improve on current regimens? Expert Rev Hematol 2025; 18:359-372. [PMID: 40207893 DOI: 10.1080/17474086.2025.2490764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION Multiple myeloma (MM) therapy has greatly evolved over the last decade. Immunomodulators and anti-CD38 antibodies have reshaped the therapeutic landscape. Nevertheless, relapses occur with worsening prognosis each relapse. Achieving deep responses in first-line treatment is key to reducing future disease burden. AREAS COVERED To advance patient outcomes, current regimens must be continuously refined through personalization, incorporation of biomarkers to guide therapy, and novel drugs. This review aims to assess existing therapies and investigate how integrating novel drug combinations and biomarker-driven approaches can improve efficacy and tolerability for newly diagnosed multiple myeloma (NDMM) patients. Meta-analysis, systematic reviews, original articles, and real-world studies were included in this review. Databases searched included PubMed, Scopus, and Google Scholar from inception until February 2025. EXPERT OPINION Discerning the best therapy sequence is a challenge for NDMM individuals. Minimal residual disease assessment is becoming a pivotal tool for guiding therapeutic approaches to enhance outcomes and tolerability. Emerging evidence supports early use of potent therapies - such as next-generation anti-CD38 antibodies and CELMoDs - to achieve deeper, more durable responses and possibly delay relapses. Immunotherapies like bispecific antibodies and CAR-T cells are also being explored in front-line settings, though reducing their infectious complications is still under investigation.
Collapse
Affiliation(s)
- Sofía Isabel Quezada-Ramírez
- Hematology Service, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León (UANL), Monterrey, Nuevo León, México
| | - Luz Del Carmen Tarín-Arzaga
- Hematology Service, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León (UANL), Monterrey, Nuevo León, México
| | - Andrés Gómez-De León
- Hematology Service, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León (UANL), Monterrey, Nuevo León, México
| | - David Gómez-Almaguer
- Hematology Service, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León (UANL), Monterrey, Nuevo León, México
| |
Collapse
|
8
|
Nanni C, Deroose CM, Balogova S, Lapa C, Withofs N, Subesinghe M, Jamet B, Zamagni E, Ippolito D, Delforge M, Kraeber-Bodéré F. EANM guidelines on the use of [ 18F]FDG PET/CT in diagnosis, staging, prognostication, therapy assessment, and restaging of plasma cell disorders. Eur J Nucl Med Mol Imaging 2024; 52:171-192. [PMID: 39207486 PMCID: PMC11599630 DOI: 10.1007/s00259-024-06858-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
We provide updated guidance and standards for the indication, acquisition, and interpretation of [18F]FDG PET/CT for plasma cell disorders. Procedures and characteristics are reported and different scenarios for the clinical use of [18F]FDG PET/CT are discussed. This document provides clinicians and technicians with the best available evidence to support the implementation of [18F]FDG PET/CT imaging in routine practice and future research.
Collapse
Affiliation(s)
- Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals (UZ) Leuven, 3000, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sona Balogova
- Nuclear Medicine, Comenius University, Bratislava, Slovakia
- Médecine Nucléaire, Hôpital Tenon, GH AP.SU, Paris, France
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liege, Belgium
- GIGA-CRC in Vivo Imaging, University of Liege, Liege, Belgium
| | - Manil Subesinghe
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Bastien Jamet
- Médecine Nucléaire, CHU Nantes, F-44000, Nantes, France
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy.
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- University of Milano-Bicocca, School of Medicine, Via Cadore 33, 20090, Monza, Italy
| | | | | |
Collapse
|
9
|
Rossi A, Cattabriga A, Bezzi D. Symptomatic Myeloma: PET, Whole-Body MR Imaging with Diffusion-Weighted Imaging or Both. PET Clin 2024; 19:525-534. [PMID: 38969566 DOI: 10.1016/j.cpet.2024.05.004] [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
According to international guidelines, patients with suspected myeloma should primarily undergo low-dose whole-body computed tomography (CT) for diagnostic purposes. To optimize sensitivity and specificity and enable treatment response assessment, whole-body MR (WB-MR) imaging should include diffusion-weighted imaging with apparent diffusion coefficient maps and T1-weighted Dixon sequences with bone marrow Fat Fraction Quantification. At baseline WB-MR imaging shows greater sensitivity for the detecting focal lesions and diffuse bone marrow infiltration pattern than 18F-fluorodeoxyglucose PET-CT, which is considered of choice for evaluating response to treatment and minimal residual disease and imaging of extramedullary disease.
Collapse
Affiliation(s)
- Alice Rossi
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Arrigo Cattabriga
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna; Dipartimento di Scienze Mediche e Chirurgiche, Via Massarenti 9, 40138 Bologna, Italy
| | | |
Collapse
|
10
|
Shim KG, Fonseca R. Measurable Residual Disease Testing in Multiple Myeloma Following T-Cell Redirecting Therapies. Cancers (Basel) 2024; 16:3288. [PMID: 39409909 PMCID: PMC11476300 DOI: 10.3390/cancers16193288] [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: 08/14/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Several novel T-cell-based therapies have recently become available for multiple myeloma (MM). These T-cell redirecting therapies (TRTs) include chimeric antigen receptor T-cells (CAR-T) and bispecific antibodies (BiAbs). In both clinical trial and real-world data, these therapies have demonstrated high rates of deep clinical response, and some are now approved for second-line treatment for relapsed MM. The deep and sustained clinical responses these therapies are capable of inducing will require sophisticated response monitoring to provide meaningful information for patient care. Obtaining measurable residual disease (MRD) negativity has been validated as an independent positive prognostic marker for progression-free survival (PFS) and overall survival (OS) in both newly diagnosed and relapsed refractory patients with multiple myeloma. Assessment for MRD negativity was performed in all of the trials for FDA-approved TRT. Here, we summarize pertinent data for MRD assessment following TRT in MM and provide a rationale and structured framework for conducting MRD testing post TRT.
Collapse
Affiliation(s)
- Kevin Guanwen Shim
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ 85054, USA
| | | |
Collapse
|
11
|
Morè S, Corvatta L, Manieri VM, Morsia E, Offidani M. The Challenging Approach to Multiple Myeloma: From Disease Diagnosis and Monitoring to Complications Management. Cancers (Basel) 2024; 16:2263. [PMID: 38927968 PMCID: PMC11202048 DOI: 10.3390/cancers16122263] [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: 05/16/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
The outcome of multiple myeloma (MM) has significantly improved in the last few decades due to several factors such as new biological discoveries allowing to better stratify disease risk, development of more effective therapies and better management of side effects related to them. However, handling all these aspects requires an interdisciplinary approach involving multiple knowledge and collaboration of different specialists. The hematologist, faced with a patient with MM, must not only choose a treatment according to patient and disease characteristics but must also know when therapy needs to be started and how to monitor it during and after treatment. Moreover, he must deal not only with organ issues related to MM such as bone disease, renal failure or neurological disease but also with adverse events, often very serious, related to novel therapies, particularly new generation immunotherapies such as CAR T cell therapy and bispecific antibodies. In this review, we provide an overview on the newer MM diagnostic and monitoring strategies and on the main side effects of MM therapies, focusing on adverse events occurring during treatment with CAR T cells and bispecific antibodies.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy; (S.M.); (V.M.M.); (E.M.)
| | - Laura Corvatta
- U.O.C. Medicina, Ospedale Profili, 60044 Fabriano, Italy;
| | - Valentina Maria Manieri
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy; (S.M.); (V.M.M.); (E.M.)
| | - Erika Morsia
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy; (S.M.); (V.M.M.); (E.M.)
| | - Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy; (S.M.); (V.M.M.); (E.M.)
| |
Collapse
|
12
|
Derman BA, Fonseca R. Measurable Residual Disease and Decision-Making in Multiple Myeloma. Hematol Oncol Clin North Am 2024; 38:477-495. [PMID: 38184470 DOI: 10.1016/j.hoc.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
Measurable (minimal) residual disease (MRD) has already proven to be one of the most important prognostic factors in multiple myeloma (MM). Each improvement in the depth of MRD testing has led to superior discrimination of outcomes, and sustained MRD negativity seems to be paramount to durable responses. Peripheral blood assays to assess for MRD are still under investigation but hold promise as complementary tools to bone marrow MRD assays such as next-generation sequencing and flow cytometry. Herein, the authors explore the evidence and potential benefits and drawbacks of MRD-adapted clinical decision-making in MM.
Collapse
Affiliation(s)
- Benjamin A Derman
- Section of Hematology/Oncology, University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637, USA.
| | - Rafael Fonseca
- Division of Hematology and Medical Oncology, Mayo Clinic in Arizona, 13400 East Shea Boulevard, MCCRB 3-001, Phoenix, AZ 85259, USA
| |
Collapse
|
13
|
Gay F. A rational approach to functional high-risk myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:433-442. [PMID: 38066896 PMCID: PMC10727111 DOI: 10.1182/hematology.2023000443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Multiple myeloma is a clinically and biologically highly heterogeneous disease, as the overall survival can vary from more than a decade in patients with standard risk disease treated with intensive chemotherapy to 2-3 years in patients with high-risk features. The current staging systems, which rely on baseline biological risk factors to stratify patients into groups with differing risks of progression or death, are sometimes suboptimal tools for identifying high-risk patients. This is particularly evident when considering the so-called functional high-risk patients-patients who do not necessarily display baseline high-risk features but typically show a suboptimal response to induction therapy or relapse early after treatment initiation: the survival of these patients is particularly poor even in the context of newer therapies. The prompt identification, as well as a consistent definition, of this subset of patients, as well as their management, currently represents an unmet medical need. In this review we explore the main characteristics of functional high-risk patients, the available known risk factors and scoring systems, and the possible management.
Collapse
Affiliation(s)
- Francesca Gay
- Division of Hematology 1, Clinical Trial Unit, AOU Città della Salute e della Scienza, Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| |
Collapse
|
14
|
Pu Y, Li L, Peng H, Liu L, Heymann D, Robert C, Vallette F, Shen S. Drug-tolerant persister cells in cancer: the cutting edges and future directions. Nat Rev Clin Oncol 2023; 20:799-813. [PMID: 37749382 DOI: 10.1038/s41571-023-00815-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
Drug-tolerant persister (DTP) cell populations were originally discovered in antibiotic-resistant bacterial biofilms. Similar populations with comparable features have since been identified among cancer cells and have been linked with treatment resistance that lacks an underlying genomic alteration. Research over the past decade has improved our understanding of the biological roles of DTP cells in cancer, although clinical knowledge of the role of these cells in treatment resistance remains limited. Nonetheless, targeting this population is anticipated to provide new treatment opportunities. In this Perspective, we aim to provide a clear definition of the DTP phenotype, discuss the underlying characteristics of these cells, their biomarkers and vulnerabilities, and encourage further research on DTP cells that might improve our understanding and enable the development of more effective anticancer therapies.
Collapse
Affiliation(s)
- Yi Pu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Lung Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Haoning Peng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - François Vallette
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France.
- Nantes Université, INSERM, U1307, CRCI2NA, Nantes, France.
| | - Shensi Shen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Oliva S, Genuardi E, Paris L, D'Agostino M, Rogers J, Rota-Scalabrini D, Jacob AP, Patriarca F, Luppi M, Bertazzoni P, Velluti C, Capra A, Saraci E, Rossi M, Allegra A, Mina R, Gentile M, Kirsch IR, Belotti A, Cavo M, Bruno B, Musto P, Boccadoro M, Zamagni E, Gay F. Prospective evaluation of minimal residual disease in the phase II FORTE trial: a head-to-head comparison between multiparameter flow cytometry and next-generation sequencing. EClinicalMedicine 2023; 60:102016. [PMID: 37396800 PMCID: PMC10314153 DOI: 10.1016/j.eclinm.2023.102016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 07/04/2023] Open
Abstract
Background Limited data are available on the concordance between multiparameter flow cytometry (MFC) and next-generation sequencing (NGS) for minimal residual disease (MRD) detection in a large trial for multiple myeloma (MM) patients. Methods MRD was explored in the FORTE trial for transplant-eligible MM patients randomised to three carfilzomib-based induction-intensification-consolidation treatments and carfilzomib-lenalidomide (KR) vs R maintenance. MRD was assessed by 8-colour 2nd-generation flow cytometry in patients with ≥very good partial response before maintenance. NGS was performed in case of suspected complete response (CR) in a correlative subanalysis. Biological/prognostic concordance between MFC and NGS, conversion to MRD negativity during maintenance, and 1-year/2-year sustained MRD negativity were explored. Findings Between September 28, 2015 and December 22, 2021, 2020 samples were available for MFC and 728 for the simultaneous MFC/NGS correlation in the "suspected CR population". Median follow-up was 62 months. Biological agreement was 87% at the 10-5 and 83% at the 10-6 cut-offs. A remarkable prognostic concordance was observed: hazard ratios in MFC-MRD and NGS-MRD-negative vs -positive patients were 0.29 and 0.27 for progression-free survival (PFS) and 0.35 and 0.31 for overall survival, respectively (p < 0.05). During maintenance, 4-year PFS was 91% and 97% in 1-year sustained MFC-MRD-negative and NGS-MRD-negative patients (10-5), respectively, and 99% and 97% in 2-year sustained MFC-MRD-negative and NGS-MRD-negative patients, regardless of treatment received. The conversion rate from pre-maintenance MRD positivity to negativity during maintenance was significantly higher with KR vs R both by MFC (46% vs 30%, p = 0.046) and NGS (56% vs 30%, p = 0.046). Interpretation The significant biological/clinical concordance between MFC and NGS at the same sensitivity suggests their possible use in the evaluation of one of the currently strongest predictors of outcome. Funding Amgen, Celgene/Bristol Myers Squibb, Multiple Myeloma Research Foundation.
Collapse
Affiliation(s)
- Stefania Oliva
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Elisa Genuardi
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Laura Paris
- Division of Hematology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Mattia D'Agostino
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Jennifer Rogers
- Multiple Myeloma Research Foundation (MMRF), Norwalk, CT, USA
| | - Delia Rota-Scalabrini
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO - IRCCS, Torino, Italy
| | | | - Francesca Patriarca
- Hematologic Clinic and Transplant Center, University Hospital of Central Friuli, DAME, University of Udine, Udine, Italy
| | - Mario Luppi
- Dipartimento di Scienze Mediche e Chirurgiche Materno Infantili e dell'Adulto, UNIMORE, UOC Ematologia, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | | | - Cristina Velluti
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Andrea Capra
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elona Saraci
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Marco Rossi
- SOC Ematologia, Department of Oncology/Hematology, Azienda Ospedaliera Pugliese-Ciaccio and Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Roberto Mina
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | | | - Angelo Belotti
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Benedetto Bruno
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Pellegrino Musto
- Department of Precision and Regenerative Medicine and Ionian Area, “Aldo Moro” University School of Medicine, Bari, Italy
- Hematology and Stem Cell Transplantation Unit, AOU Consorziale Policlinico, Bari, Italy
| | - Mario Boccadoro
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Francesca Gay
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|