Skip to main content
Log in

MRD Assessment in Multiple Myeloma: Progress and Challenges

  • Multiple Myeloma (P Kapoor, Section Editor)
  • Published:
Current Hematologic Malignancy Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Over the last decade, the development of effective treatment approaches for multiple myeloma (MM) has been associated with higher response rates and longer survival. In patients who achieve complete response, several high sensitivity techniques have been studied to assess minimal residual disease (MRD) and detect residual neoplastic cells within the bone marrow (by flow cytometry or molecular biology techniques) or outside the bone marrow (by imaging or circulating disease markers in the peripheral blood). This is of utmost importance, since residual disease can drive clinical relapse. This review focuses on the progress made in the assessment of MRD in MM.

Recent Findings

The achievement of MRD negativity after therapy is considered prognostically important for MM patients, and data from clinical trials and meta-analyses have confirmed that it is strongly associated with better survival. Along with well-known techniques, such as next-generation sequencing (NGS), next-generation flow (NGF), and positron emission tomography/computed tomography (PET/CT), other methods such as mass spectrometry (MS) and circulating tumor cells are under study. Intensive treatment regimens at diagnosis can lead up to 70% of MRD negativity in MM patients, although the current proportion of curable patients is still unknown.

Summary

Today, clinicians who treat MM deal with MRD assessment in routine clinical practice. Its appropriate use in therapeutic decision making may be the most fascinating and challenging issue to be addressed over the next few years.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. •• Munshi NC, Avet-Loiseau H, Anderson KC, et al. A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma. Blood Adv. 2020;4:5988–99 A recent meta-analysis on around 8000 patients from different MRD studies, confirming the important prognostic impact of the achievement of MRD negativity, regardless of the adopted method (next-generation sequencing [NGS] or multiparameter flow cytometry [MFC]), time point, and setting (diagnosis or relapse).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Oliva S, D’Agostino M, Boccadoro M, Larocca A. Clinical applications and future directions of minimal residual disease testing in multiple myeloma. Front Oncol. 2020;10. https://doi.org/10.3389/fonc.2020.00001.

  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.

    Article  PubMed  Google Scholar 

  4. •• Costa LJ, Derman BA, Bal S, et al. International harmonization in performing and reporting minimal residual disease assessment in multiple myeloma trials. Leukemia. 2021;35:18–30 An international expert consensus on how to report MRD analysis, to foster the standardization and comparison among different studies.

    Article  PubMed  Google Scholar 

  5. Flores-Montero J, de Tute R, Paiva B, Perez JJ, Böttcher S, Wind H, et al. Immunophenotype of normal vs. myeloma plasma cells: toward antibody panel specifications for MRD detection in multiple myeloma. Cytometry B Clin Cytom. 2016;90:61–72.

    Article  CAS  PubMed  Google Scholar 

  6. Stetler-Stevenson M, Paiva B, Stoolman L, Lin P, Jorgensen JL, Orfao A, et al. Consensus guidelines for myeloma minimal residual disease sample staining and data acquisition. Cytometry B Clin Cytom. 2016;90:26–30.

    Article  CAS  PubMed  Google Scholar 

  7. Flores-Montero J, Sanoja-Flores L, Paiva B, Puig N, García-Sánchez O, Böttcher S, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 2017;31:2094–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Paiva B, Puig N, Cedena MT, Rosiñol L, Cordón L, Vidriales MB, et al. Measurable residual disease by next-generation flow cytometry in multiple myeloma. J Clin Oncol. 2020;38:784–92.

    Article  CAS  PubMed  Google Scholar 

  9. Ladetto M, Pagliano G, Ferrero S, Cavallo F, Drandi D, Santo L, et al. Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma. J Clin Oncol. 2010;28:2077–84.

    Article  CAS  PubMed  Google Scholar 

  10. Gambella M, Omedé P, Spada S, Muccio VE, Gilestro M, Saraci E, et al. Minimal residual disease by flow cytometry and allelic-specific oligonucleotide real-time quantitative polymerase chain reaction in patients with myeloma receiving lenalidomide maintenance: a pooled analysis. Cancer. 2019;125:750–60.

    Article  CAS  PubMed  Google Scholar 

  11. Puig N, Sarasquete ME, Balanzategui A, Martínez J, Paiva B, García H, et al. Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myeloma. A comparative analysis with flow cytometry Leukemia. 2014;28:391–7.

    CAS  PubMed  Google Scholar 

  12. Avet-Loiseau H. Minimal residual disease by next-generation sequencing: pros and cons. Am Soc Clin Oncol Educ B. 2016;36:e425–30.

    Article  Google Scholar 

  13. Perrot A, Lauwers-Cances V, Corre J, Robillard N, Hulin C, Chretien ML, et al. Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma. Blood. 2018;132:2456–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hillengass J, Usmani S, Rajkumar SV, Durie BGM, Mateos MV, Lonial S, et al. International myeloma working group consensus recommendations on imaging in monoclonal plasma cell disorders. Lancet Oncol. 2019;20:e302–12.

    Article  PubMed  Google Scholar 

  15. Cavo M, Terpos E, Nanni C, Moreau P, Lentzsch S, Zweegman S, et al. Role of 18F-FDG PET/CT in the diagnosis and management of multiple myeloma and other plasma cell disorders: a consensus statement by the International Myeloma Working Group. Lancet Oncol. 2017;18:e206–17.

    Article  PubMed  Google Scholar 

  16. Bhutani M, Foureau DM, Atrash S, Voorhees PM, Usmani SZ. Extramedullary multiple myeloma. Leukemia. 2020;34:1–20. https://doi.org/10.1038/s41375-019-0660-0.

    Article  CAS  PubMed  Google Scholar 

  17. Rasche L, Chavan SS, Stephens OW, et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat Commun. 2017;8:1–10.

    Article  CAS  Google Scholar 

  18. Rasche L, Angtuaco E, McDonald JE, et al. Low expression of hexokinase-2 is associated with false-negative FDG–positron emission tomography in multiple myeloma. Blood. 2017;130:30–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lapa C, Garcia-Velloso MJ, Lückerath K, Samnick S, Schreder M, Otero PR, et al. 11C-Methionine-PET in multiple myeloma: a combined study from two different institutions. Theranostics. 2017;7:2956–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Oliva S, Genuardi E, Belotti A, et al (2020) Multiparameter flow cytometry (MFC) and next generation sequencing (NGS) for minimal residual disease (MRD) evaluation: Results of the FORTE trial in newly diagnosed multiple myeloma (MM). J Clin Oncol 38:Abstract #8533 [ASCO 2020 Annual Meeting].

  21. 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 Lymphoma, Myeloma Leuk. 2019;19:e3–4.

    Article  Google Scholar 

  22. Medina A, Puig N, Flores-Montero J, Jimenez C, Sarasquete ME, Garcia-Alvarez M, et al. Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma. Blood Cancer J. 2020;10:108.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Zamagni E, Nanni C, Gay F, et al (2020) MRD evaluation by PET/CT according to Deauville criteria combined with bone marrow techniques in newly diagnosed transplant eligible multiple myeloma patients enrolled in the phase II FORTE trial. HemaSphere 4:60 [Abstract #S207, EHA 2020 25th Congress].

  24. Bertamini L, Grasso M, D’Agostino M, et al (2020) Poor prognosis of multiple myeloma predicted by high levels of circulating plasma cells is independent from other high-risk features but is modulated by the achievement of minimal residual disease negativity. Blood 136:12-13 [Abstract #720, ASH 2020 62nd Meeting].

  25. • Sanoja-Flores L, Flores-Montero J, Puig N, et al. Blood monitoring of circulating tumor plasma cells by next generation flow in multiple myeloma after therapy. Blood. 2019;134:2218–22. This paper focuses on peripheral blood (PB) MRD assessment with next-generation flow (NGF), as compared with bone marrow (BM) MRD assessment. Up to now, these data have revealed a lower sensitivity and inferiority of PB assessment compared to BM assessment.

  26. • Mazzotti C, Buisson L, Maheo S, et al. Myeloma MRD by deep sequencing from circulating tumor DNA does not correlate with results obtained in the bone marrow. Blood Adv. 2018;2:2811–3 This paper focuses on peripheral blood (PB) MRD assessment with next-generation sequencing (NGS), as compared with bone marrow (BM) MRD assessment. Up to now, these data have revealed a lower sensitivity and inferiority of PB assessment compared to BM assessment.

  27. • Murray DL, Puig N, Kristinsson S, et al. Mass spectrometry for the evaluation of monoclonal proteins in multiple myeloma and related disorders : an International Myeloma Working Group Mass Spectrometry Committee Report. Blood Cancer J. 2021:4–9 A first report by the International Myeloma Working Group (IMWG) on the possible role of mass spectrometry (MS) in plasma cell disorders.

  28. Zajec M, Langerhorst P, Noori S, et al (2020) Minimal residual disease in multiple myeloma: targeted mass spectrometry in blood vs next generation sequencing in bone marrow. Blood 136:9 [Abstract #3156, ASH 2020 62nd Meeting].

  29. Puíg N, Contreras T, Paiva B, et al (2020) Analysis of treatment efficacy in the GEM-CESAR trial for high-risk smoldering multiple myeloma patients: comparison between the standard and IMWG MRD criteria and QIP-MS including FLC (QIP-FLC-MS). J Clin Oncol 38: Abstract #8512 [ASCO 2020 Annual Meeting].

  30. Nandakumar B, Murray DL, Dispenzieri A, Kapoor P, Buadi FK, Lacy MQ, et al. Sequential comparison of conventional serum immunofixation (IFE) to mass spectrometry-based assessment (MASS FIX) in patients with multiple myeloma (MM). Blood. 2020;136:12–3.

    Article  Google Scholar 

  31. Eveillard M, Rustad E, Roshal M, Zhang Y, Ciardiello A, Korde N, et al. Comparison of MALDI-TOF mass spectrometry analysis of peripheral blood and bone marrow-based flow cytometry for tracking measurable residual disease in patients with multiple myeloma. Br J Haematol. 2020;189:904–7.

  32. Abeykoon JP, Murray DL, Murray I, Jevremovic D, Otteson GE, Dispenzieri A, et al. Implications of detecting serum monoclonal protein by MASS-fix following stem cell transplantation in multiple myeloma. Br J Haematol. 2021;193:380–5.

  33. Walker R, Barlogie B, Haessler J, Tricot G, Anaissie E, Shaughnessy JD Jr, et al. Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications. J Clin Oncol. 2007;25:1121–8.

    Article  PubMed  Google Scholar 

  34. Latifoltojar A, Hall-Craggs M, Rabin N, Popat R, Bainbridge A, Dikaios N, et al. Whole body magnetic resonance imaging in newly diagnosed multiple myeloma: early changes in lesional signal fat fraction predict disease response. Br J Haematol. 2017;176:222–33.

    Article  PubMed  Google Scholar 

  35. Messiou C, Hillengass J, Delorme S, Lecouvet FE, Moulopoulos LA, Collins DJ, et al. Guidelines for acquisition, interpretation, and reporting of whole-body MRI in myeloma: myeloma response assessment and diagnosis system (MY-RADS). Radiology. 2019;291:5–13.

    Article  PubMed  Google Scholar 

  36. Brüggemann M, Kotrová M, Knecht H, et al. Standardized next-generation sequencing of immunoglobulin and T-cell receptor gene recombinations for MRD marker identification in acute lymphoblastic leukaemia; a EuroClonality-NGS validation study. Leukemia. 2019;33:2241–53.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Scheijen B, Meijers RWJ, Rijntjes J, et al. Next-generation sequencing of immunoglobulin gene rearrangements for clonality assessment: a technical feasibility study by EuroClonality-NGS. Leukemia. 2019;33:2227–40.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Knecht H, Reigl T, Kotrová M, et al. Quality control and quantification in IG/TR next-generation sequencing marker identification: protocols and bioinformatic functionalities by EuroClonality-NGS. Leukemia. 2019;33:2254–65.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Martinez-Lopez J, Sanchez-Vega B, Barrio S, Cuenca I, Ruiz-Heredia Y, Alonso R, et al. Analytical and clinical validation of a novel in-house deep-sequencing method for minimal residual disease monitoring in a phase II trial for multiple myeloma. Leukemia. 2017;31:1446–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Arcila ME, Yu W, Syed M, Kim H, Maciag L, Yao JJ, et al. Establishment of immunoglobulin heavy (IGH) chain clonality testing by next-generation sequencing for routine characterization of B-cell and plasma cell neoplasms. J Mol Diagnostics. 2019;21:330–42.

    Article  CAS  Google Scholar 

  41. Nanni C, Zamagni E, Versari A, Chauvie S, Bianchi A, Rensi M, et al. Image interpretation criteria for FDG PET/CT in multiple myeloma: a new proposal from an Italian expert panel. IMPeTUs (Italian Myeloma criteria for PET USe). Eur J Nucl Med Mol Imaging. 2016;43:414–21.

    Article  CAS  PubMed  Google Scholar 

  42. Nanni C, Versari A, Chauvie S, Bertone E, Bianchi A, Rensi M, et al. Interpretation criteria for FDG PET/CT in multiple myeloma (IMPeTUs): final results. IMPeTUs (Italian myeloma criteria for PET USe). Eur J Nucl Med Mol Imaging. 2018;45:712–9.

    Article  PubMed  Google Scholar 

  43. • Zamagni E, Nanni C, Dozza L, et al. Standardization of 18 F-FDG–PET/CT according to Deauville criteria for metabolic complete response definition in newly diagnosed multiple myeloma. J Clin Oncol. 2021;39:116–25 An important step towards the standardization of positron emission tomography/computed tomography (PET/CT) response assessment. In a pooled analysis of two phase III trials, Zamagni et al. pinpointed the prognostic impact of response by PET/CT following the Deauville criteria.

    Article  PubMed  Google Scholar 

  44. Avet-Loiseau H, San-Miguel J, Casneuf T, Iida S, Lonial S, Usmani SZ, et al. Evaluation of sustained minimal residual disease negativity with daratumumab-combination regimens in relapsed and/or refractory multiple myeloma: analysis of POLLUX and CASTOR. J Clin Oncol. 2021;39:1139–49.

    Article  PubMed  Google Scholar 

  45. Kaufman JL, Laubach JP, Sborov D, Reeves B, Rodriguez C, Chari A, et al. Daratumumab (DARA) Plus lenalidomide, bortezomib, and dexamethasone (RVd) in patients with transplant-eligible newly diagnosed multiple myeloma (NDMM): updated analysis of Griffin after 12 months of maintenance therapy. Blood. 2020;136:45–6.

    Article  Google Scholar 

  46. Lahuerta J-J, Paiva B, Vidriales M-B, Cordón L, Cedena MT, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. U.S. Department of Health and Human Services - Food and Drug Administration. Clinical trial endpoints for the approval of cancer drugs and biologics. Guidance for Industry. Silver Spring, US-MD: Guidance for Industry; 2018.

  48. Holstein SA, Al-Kadhimi Z, Costa LJ, et al. Summary of the third annual blood and marrow transplant clinical trials network myeloma intergroup workshop on minimal residual disease and immune profiling. Biol Blood Marrow Transplant. 2020;26:e7–e15.

    Article  PubMed  Google Scholar 

  49. D’Agostino M, Zaccaria GM, Ziccheddu B, et al. Early relapse risk in patients with newly diagnosed multiple myeloma characterized by next-generation sequencing. Clin Cancer Res. 2020;26:4832–41.

    Article  PubMed  Google Scholar 

  50. Larocca A, Dold SM, Zweegman S, Terpos E, Wäsch R, D’Agostino M, et al. Patient-centered practice in elderly myeloma patients: an overview and consensus from the European Myeloma Network (EMN). Leukemia. 2018;32:1697–712.

    Article  PubMed  Google Scholar 

  51. Salvini M, D’Agostino M, Bonello F, Boccadoro M, Bringhen S. Determining treatment intensity in elderly patients with multiple myeloma. Expert Rev Anticancer Ther. 2018;18:917–30.

    Article  CAS  PubMed  Google Scholar 

  52. Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2020;21:1630–42.

    Article  CAS  PubMed  Google Scholar 

  53. Gay F, Musto P, Rota-Scalabrini D, et al (2020) Survival analysis of newly diagnosed transplant-eligible multiple myeloma patients in the randomized forte trial. Blood 136:35-37 [Abstract #141, ASH 2020 62nd Meeting].

  54. D’Agostino M, De Paoli L, Conticello C, et al. Continuous therapy in standard- and high-risk newly-diagnosed multiple myeloma: a pooled analysis of 2 phase III trials. Crit Rev Oncol Hematol. 2018;132:9–16.

    Article  PubMed  Google Scholar 

  55. Larocca A, Bonello F, Gaidano G, D'Agostino M, Offidani M, Cascavilla N, et al. Dose/schedule-adjusted Rd-R vs continuous Rd for elderly, intermediate-fit, newly diagnosed multiple myeloma patients. Blood. 2021; online ahead of print. https://doi.org/10.1182/blood.2020009507.

  56. D’Agostino M, Bertamini L, Oliva S, Boccadoro M, Gay F. Pursuing a curative approach in multiple myeloma: a review of new therapeutic strategies. Cancers (Basel). 2019;11:2015.

  57. Mateos M-V, Martinez-Lopez J, Rodriguez Otero P, et al (2019) Curative strategy (GEM-CESAR) for high-risk smoldering myeloma (SMM): carfilzomib, lenalidomide and dexamethasone (KRd) as induction followed by HDT-ASCT, consolidation with Krd and maintenance with Rd. Blood 134:Abstract #781 [ASH 2019 61st Meeting].

  58. Pandit-Taskar N. Functional imaging methods for assessment of minimal residual disease in multiple myeloma: current status and novel immunoPET based methods. Semin Hematol. 2018;55:22–32.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Martinez-Lopez J, Lahuerta JJ, Pepin F, González M, Barrio S, Ayala R, et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood. 2014;123:3073–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kriegsmann K, Hundemer M, Hofmeister-Mielke N, et al. Comparison of ngs and mfc methods: Key metrics in multiple myeloma mrd assessment. Cancers (Basel). 2020;12:1–15.

    Article  Google Scholar 

  61. Rodríguez S, Goicoechea I, Gemenetzi K, et al (2020) Discordances between immunofixation (IFx) and minimal residual disease (MRD) assessment with next-generation flow (NGF) and sequencing (NGS) in patients (Pts) with multiple myeloma (MM): clinical and pathogenic significance. Blood 136:5-6 [Abstract #62, ASH 2020 62nd Meeting].

  62. Sonneveld P, Broijl A, Gay F, et al (2019) Bortezomib, lenalidomide, and dexamethasone (VRd) ± daratumumab (DARA) in patients (pts) with transplant-eligible (TE) newly diagnosed multiple myeloma (NDMM): a multicenter, randomized, phase III study (PERSEUS). J Clin Oncol 37:Abstract #TPS8055 [ASCO 2019 Annual Meeting].

  63. Costa LJ, Chhabra S, Godby KN, et al (2019) Daratumumab, carfilzomib, lenalidomide and dexamethasone (Dara-KRd) induction, autologous transplantation and post-transplant, response-adapted, measurable residual disease (MRD)-based Dara-Krd consolidation in patients with newly diagnosed multiple myelo. Blood 134:Abstract #860 [ASH 2019 61st Meeting].

Download references

Author information

Authors and Affiliations

Authors

Contributions

All authors conceived and designed the work that led to the submission.

All authors analyzed the data and interpreted the results.

All authors drafted the first version of the manuscript.

All authors revised the manuscript and approved the final version.

All authors agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Corresponding author

Correspondence to Francesca Gay.

Ethics declarations

Conflict of Interest

LB declares no competing financial interests.

MD has received honoraria for lectures from Sanofi and GSK and has served on the advisory boards for GSK.

FG has received honoraria from Amgen, Celgene, Janssen, Takeda, Bristol-Myers Squibb, AbbVie, and GSK and has served on the advisory boards for Amgen, Celgene, Janssen, Takeda, Bristol-Myers Squibb, AbbVie, GSK, Roche, Adaptive Biotechnologies, Oncopeptides, Bluebird, and Secura Bio.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Multiple Myeloma

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bertamini, L., D’Agostino, M. & Gay, F. MRD Assessment in Multiple Myeloma: Progress and Challenges. Curr Hematol Malig Rep 16, 162–171 (2021). https://doi.org/10.1007/s11899-021-00633-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11899-021-00633-5

Keywords

Navigation