Skip to main content

EDITORIAL article

Front. Oncol., 22 May 2023
Sec. Hematologic Malignancies
This article is part of the Research Topic Minimal Residual Disease (MRD) Assessment in Multiple Myeloma Patients View all 9 articles

Editorial: Minimal residual disease (MRD) assessment in multiple myeloma patients

  • 1Department of Internal Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
  • 2Department of Hematology, Instituto Americas de Ensino, Pesquisa e Inovação, Rio de Janeiro, Brazil
  • 3Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) Institute for Biomedical Research of Salamanca (IBASAL), CIBERONC and Department of Medicine, University of Salamanca, Salamanca, Spain

Multiple myeloma (MM) is a neoplastic plasma cell disorder characterized by the expansion and accumulation of clonal plasma cells. The introduction of novel drugs and new therapeutic options has led to significantly higher complete response rates and prolonged progression-free and overall survival (1). Despite these advances, MM remains an incurable disease. Thus, there is a need to refine response criteria and methods for more sensitive identification of persistence of lower levels of minimal (measurable) residual disease (MRD) (2). The International Myeloma Working Group has defined the response criteria for patients with MM by including MRD (3). Moreover, depth response based on MRD has emerged as one of the most important independent prognostic factors in MM and has been tested as a dynamic tool for treatment/disease monitoring, prognostication, and as a new (potential) therapeutic endpoint in clinical trials and drug approval for MM patients (4).

The so-called Next Generation Flow (NGF) approach developed by the EuroFlow Consortium (www.euroflo.org) is a high sensitive method to evaluate MRD by flow cytometry, which has been validated now in many centers around the world (5). Turner R. et al. analyzed EuroFlow MRD results in a real-world practice from patients with MM patients eligible for autologous stem cell transplantation (ASCT) treated with VCD as induction. Patients who achieved MRD negativity after ASCT had a significantly longer PFS than those with persisting MRD (Turner et al.). This observation is true not only in the ASCT scenario, but also for older patients who are not eligible for ASCT, and treated with more intensive combination of drugs where MRD negativity is an important parameter for prognosis (6).

Another validated method to evaluate MRD relies on Next Generation Sequencing (NGS). This method was tested not only in clinical trials but also in a real-world practice (7). Also, for innovative treatment strategies like CAR-T therapy in the MM relapse setting, MRD evaluation becomes an important prognostic factor. (8) Wong et al. performed a retrospective analysis of 54 BCMA-CAR-T treated MM patients from five different clinical trials at the University of California San Francisco. Patients achieving MRD-NGS below the detectable limit at a sensitivity of <10−6 had a better PFS than those with detectable disease at one month and three months after starting treatment.

Continuous MRD monitoring based on bone marrow (BM) analysis remains challenging because BM aspiration is an invasive procedure that cannot be repeated frequently. BM-based assays do not allow for the detection of extramedullary disease, which is increasingly seen in the clinic. From this moment on, MRD monitoring during treatment, and particularly after therapy, currently relies mainly on other less-invasive (but less sensitive) techniques, such as serum-based assessment in addition to imaging. Therefore, MRD assays based on peripheral blood (PB) would be worthwhile (9). Mass spectrometry techniques have recently been used to detect M-protein in serum with higher sensitivity than the current electrophoretic methods (10). Many groups have also investigated the role of circulating plasma cells (CPCs) at diagnosis. CPCs are becoming recognized as an independent prognostic factor in MM (including smoldering MM as well) both at diagnosis and after starting therapy (9, 11, 12).

Dhakl B.et al compared the performance of a non-invasive, circulating tumor DNA (ctDNA)-based MRD assay with multiparameter flow cytometry (MFC) of marrow aspirate to predict relapse in ASCT recipients with MM. MRD assessment using ctDNA was retrospectively analyzed on 80 plasma samples from 28 patients collected at different time points, post-AHCT. The median PFS for ctDNA-positive patients was 31 months, and that for the ctDNA-negative patients was 84 months (HR: 5.6; 95%CI: 1.8-17; p=0.0003) (Dhakal et al.).

MRD evaluation in MM treatment scenario is nowadays an important tool not only as prognostic marker but also for a risk based guided therapeutic strategy (13). Soon, the MM treatment decision, probably will be guided by sequencing evaluation of MRD (14).

In this special issue of Frontiers in Oncology | Hematologic Malignancies, a total of 8 papers related to MRD in MM are presented, these include: i) a retrospective validation of Euroflow MRD in Real-World MM patients (Turner et al.); ii) an assessment of MRD using circulating tumor DNA from patients post-ASCT (Dhakal et al.); iii) a single institution analysis of MRD in patients treated with BCMA CAR-T Cells (Wong et al.); iv) a retrospective analysis of MRD significance in patients who received ASCT (Sun et al.); v) a study evaluating bone marrow and apheresis samples with a highly sensitive 10-color MFC panel (Riebl et al.); vi) a review of the possible role of quantifying measurable clonal plasma cells in stem cell grafts (Seval et al.); and vii/viii) two studies about the state of the art of MRD evaluation in MM. (Charalampous and Kourelis, Alonso and Lahuerta).

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Puertas B, González-Calle V, Sobejano-Fuertes E, Escalante F, Queizán JA, Bárez A, et al. Novel agents as main drivers for continued improvement in survival in multiple myeloma. Cancers (Basel) (2023) 15(5):1558. doi: 10.3390/cancers15051558

CrossRef Full Text | Google Scholar

2. Munshi NC, Avet-Loiseau H, Rawstron AC, Owen RG, Child JA, Thakurta A, et al. Association of minimal residual disease with superior survival outcomes in patients with multiple myeloma: a meta-analysis. JAMA Oncol (2017) 3:28–35. doi: 10.1001/jamaoncol.2016.3160

CrossRef Full Text | Google Scholar

3. Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, et al. International myeloma working group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol (2016) 17:e328–46. doi: 10.1016/S1470-2045(16)30206-6

CrossRef Full Text | Google Scholar

4. Lahuerta J-J, Paiva B, Vidriales M-B, Cordón L, Cedena M-T, Puig N, et al. Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol (2017) 35:2900–10. doi: 10.1200/JCO.2016.69.2517

CrossRef Full Text | Google Scholar

5. Flores-Montero J, Sanoja-Flores L, Paiva B, Puig N, Garcia-Sanchez O, Bottcher S, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia (2017) 31:2094–103. doi: 10.1038/leu.2017.29

CrossRef Full Text | Google Scholar

6. Facon T, Kumar S, Plesner T, Orlowski RZ, Moreau P, Bahlis N, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med (2019) 380:2104–15. doi: 10.1056/NEJMoa1817249

CrossRef Full Text | Google Scholar

7. Avet-Loiseau H, Bene MC, Wuilleme S, Corre J, Attal M, Arnulf B, et al. Concordance of post-consolidation minimal residual disease rates by multiparametric flow cytometry and next-generation sequencing in CASSIOPEIA. Clin Lymph Myeloma Leuk (2019) 19:e3–4. doi: 10.1016/j.clml.2019.09.005

CrossRef Full Text | Google Scholar

8. Munshi NC, Berdeja JG, Lin Y, Kochenderfer J, Raje NS, Liedtke M, et al. Early MRD negativity to predict deepening myeloma response in relapsed/refractory multiple myeloma (RRMM) patients treated with bb2121 anti-BCMA CAR T cells. J Clin Oncol (2018) 36:8024–4. doi: 10.1182/blood.2019002610

CrossRef Full Text | Google Scholar

9. Sanoja-Flores L, Flores-Montero J, Puig N, Contreras-Sanfeliciano T, Pontes R, Corral-Mateos A, et al. Blood monitoring of circulating tumor plasma cells by next generation flow in multiple myeloma after therapy. Blood (2019) 134:2218–22. doi: 10.1038/s41408-023-00803-z

CrossRef Full Text | Google Scholar

10. Noori S, Wijnands C, Langerhorst P, Bonifay V, Stingl C, Touzeau C, et al. MM. dynamic monitoring of myeloma minimal residual disease with targeted mass spectrometry. Blood Cancer J (2023) 13(1):30. doi: 10.1038/s41408-023-00803-z

CrossRef Full Text | Google Scholar

11. Termini R, Žihala D, Terpos E, Perez-Montaña A, Jelínek T, Raab M, et al. PETHEMA/GEM and iMMunocell cooperative groups. circulating tumor and immune cells for minimally invasive risk stratification of smoldering multiple myeloma. Clin Cancer Res (2022) 28(21):4771–81. doi: 10.1158/1078-0432.CCR-22-1594

CrossRef Full Text | Google Scholar

12. Garcés JJ, Cedena MT, Puig N, Burgos L, Perez JJ, Cordon L, et al. Circulating tumor cells for the staging of patients with newly diagnosed transplant-eligible multiple myeloma. J Clin Oncol (2022) 40(27):3151–61. doi: 10.1080/10428194.2022.2123231

CrossRef Full Text | Google Scholar

13. Bal S, Schmidt TM, Costa LJ, Callander NS. Clinical implications of measurable residual disease assessment in multiple myeloma in the era of quadruplet therapy. Leuk Lymphoma (2022) 63(14):3288–98. doi: 10.1200/JCO.21.01935

CrossRef Full Text | Google Scholar

14. Costa LJ, Chhabra S, Medvedova E, Dholaria BR, Schmidt TM, Godby KN, et al. Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma. J Clin Oncol (2022) 40(25):2901–12. doi: 10.1200/JCO.2018.36.15_suppl.8024

CrossRef Full Text | Google Scholar

Keywords: minimal residual disease, multiple myeloma, prognostic factor, disease monitoring, flow cytometry

Citation: Maiolino A, Costa ES and Orfao A (2023) Editorial: Minimal residual disease (MRD) assessment in multiple myeloma patients. Front. Oncol. 13:1211935. doi: 10.3389/fonc.2023.1211935

Received: 25 April 2023; Accepted: 27 April 2023;
Published: 22 May 2023.

Edited and Reviewed by:

Alessandro Isidori, AORMN Hospital, Italy

Copyright © 2023 Maiolino, Costa and Orfao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Angelo Maiolino, maiolino@hucff.ufrj.br

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.