Longitudinal minimal residual disease assessment in multiple myeloma patients in complete remission - results from the NMSG flow-MRD substudy within the EMN02/HO95 MM trial

Alexander Schmitz, Rasmus Froberg Brøndum, Hans Erik Johnsen, Ulf-Henrik Mellqvist, Anders Waage, Peter Gimsing, Davine Hofste Op Bruinink, Vincent van der Velden, Bronno van der Holt, Markus Hansson, Niels Frost Andersen, Ulf Christian Frølund, Carsten Helleberg, Fredrik H Schjesvold, Lucia Ahlberg, Nina Gulbrandsen, Bjorn Andreasson, Birgitta Lauri, Einar Haukas, Julie Støve Bødker, Anne Stidsholt Roug, Martin Bøgsted, Marianne T Severinsen, Henrik Gregersen, Niels Abildgaard, Pieter Sonneveld, Karen Dybkær, Alexander Schmitz, Rasmus Froberg Brøndum, Hans Erik Johnsen, Ulf-Henrik Mellqvist, Anders Waage, Peter Gimsing, Davine Hofste Op Bruinink, Vincent van der Velden, Bronno van der Holt, Markus Hansson, Niels Frost Andersen, Ulf Christian Frølund, Carsten Helleberg, Fredrik H Schjesvold, Lucia Ahlberg, Nina Gulbrandsen, Bjorn Andreasson, Birgitta Lauri, Einar Haukas, Julie Støve Bødker, Anne Stidsholt Roug, Martin Bøgsted, Marianne T Severinsen, Henrik Gregersen, Niels Abildgaard, Pieter Sonneveld, Karen Dybkær

Abstract

Background: Multiple myeloma remains an incurable disease with multiple relapses due to residual myeloma cells in the bone marrow of patients after therapy. Presence of small number of cancer cells in the body after cancer treatment, called minimal residual disease, has been shown to be prognostic for progression-free and overall survival. However, for multiple myeloma, it is unclear whether patients attaining minimal residual disease negativity may be candidates for treatment discontinuation. We investigated, if longitudinal flow cytometry-based monitoring of minimal residual disease (flow-MRD) may predict disease progression earlier and with higher sensitivity compared to biochemical assessments.

Methods: Patients from the Nordic countries with newly diagnosed multiple myeloma enrolled in the European-Myeloma-Network-02/Hovon-95 (EMN02/HO95) trial and undergoing bone marrow aspiration confirmation of complete response, were eligible for this Nordic Myeloma Study Group (NMSG) substudy. Longitdudinal flow-MRD assessment of bone marrow samples was performed to identify and enumerate residual malignant plasma cells until observed clinical progression.

Results: Minimal residual disease dynamics were compared to biochemically assessed changes in serum free light chain and M-component. Among 20 patients, reaching complete response or stringent complete response during the observation period, and with ≥3 sequential flow-MRD assessments analysed over time, increasing levels of minimal residual disease in the bone marrow were observed in six cases, preceding biochemically assessed disease and clinical progression by 5.5 months and 12.6 months (mean values), respectively. Mean malignant plasma cells doubling time for the six patients was 1.8 months (95% CI, 1.4-2.3 months). Minimal malignant plasma cells detection limit was 4 × 10-5.

Conclusions: Flow-MRD is a sensitive method for longitudinal monitoring of minimal residual disease dynamics in multiple myeloma patients in complete response. Increasing minimal residual disease levels precedes biochemically assessed changes and is an early indicator of subsequent clinical progression.

Trial registration: NCT01208766.

Keywords: Flow cytometry; Minimal residual disease; Multiple myeloma.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
1st Flow-RA of bone marrow sample received from patients at response evaluation. X-axis: Patients with response status of CR and sCR are aligned according to PID number and flow-MRD status. Y-axis: Cell Counts/sFLCr values (log scale). (*1): sFLCr was close to normal range (0.24) and became normal with the next sFLCr measurement (1.58). (*2): sFLCr was taken 27 days after MFC date, previous sFLCr measurement (> 27 days before MFC date) displayed normal ratio. Abbreviations: CR: Complete response. SCR: Stringent complete response. Patient ID / PID: Patient identification number. SFLCr: Serum free light chain ratio. MFC: Multiparamteric flow cytometry. mPC: Malignant plasma cells. nPC: normal polyclonal plasma cells
Fig. 2
Fig. 2
Flow-MRD monitoring of patients reaching CR/sCR during their course, identifies flow-MRD positive patients over time before progression is clinically observed, while flow-MRD negative maintain progression-free regardless their initial RS status. Data points for longitudinal flow-MRD assessment (circles) after inclusion from 20 patients with ≥3 flow-MRD BM samples (inclusive 1st RA), analysed until clinical progression or until date of last clinical contact are lined up over time. Inclusion, RS, progression and date of last contact (until censoring) are indicated. Abbreviations: PR: Partial response. VGPR: Very good PR. CR: Complete response. SCR: stringent CR. INCL: Date of inclusion into the MRD-study. PROG: Date of clinical progression. Flow-MRD: MRD assessment using MFC. RA = Response assessment. RS: Response status (clinical). Patient ID: Patient identification number. SFLCr: Serum free light chain ratio. MFC: Multiparamteric flow cytometry. mPC: Malignant plasma cells. DLC: Date of last contact or censoring date. (*): Patients progressed with extramedullary disease. n.d.: 1st RA BM sample not received or flow-MRD status not determined
Fig. 3
Fig. 3
Monitoring the quantity of flow-MRD until clinical progression across six patients with clinical progression reveals, that flow-MRD positivity precedes substantial changes in clinical monitored values for sFLCr, SPEP and IF, and subsequent clinical progression. Time from inclusion to measured flow-MRD positivity varied from 5.9–64.4 month (Mean 19.1 month). MRD positivity was detected with a mean of 12.6 month (range 9.6–26.6) before clinical progression (according to IMWG standard) was recorded. Median mPC doubling time was calculated to 1.8 month (1.4–2.3) across the six patients, fitting a log-linear model for mPC concentration (mPC/ × 106 events) versus time to clinical progression. X-axis: Timeline (months to clinical progression). Y-axis: mPC frequency. Achieved flow-MRD detection limit for the MRD+ patient group is shown as a dotted horizontal line. Abbreviations: MRD: Minimal residual disease. PID: Patient identification ID. SCR: Stringent complete response. SFLCr: Serum free light chain ratio (abnormal/normal). IF: Immunofixation (positive/negative) SPEP: Serum protein electrophoresis (positive/negative). Lin: Linear. Log: Logarithmic. nd: not determined. “0”: Flow-MRD negative

References

    1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011;364(11):1046–1060.
    1. Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK, et al. International myeloma working group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23(12):2210–2221.
    1. Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood. 2008;111(5):2516–2520.
    1. Ravi P, Kumar SK, Cerhan JR, Maurer MJ, Dingli D, Ansell SM, et al. Defining cure in multiple myeloma: a comparative study of outcomes of young individuals with myeloma and curable hematologic malignancies. Blood Cancer J. 2018;8(3):26. doi: 10.1038/s41408-018-0065-8.
    1. 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(8):e328–e346. doi: 10.1016/S1470-2045(16)30206-6.
    1. Dispenzieri A, Kyle R, Merlini G, Miguel JS, Ludwig H, Hajek R, et al. International myeloma working group guidelines for serum-free light chain analysis in multiple myeloma and related disorders. Leukemia. 2009;23(2):215–224.
    1. Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos M-VV, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538–e548. doi: 10.1016/S1470-2045(14)70442-5.
    1. Kapoor P, Kumar SK, Dispenzieri A, Lacy MQ, Buadi F, Dingli D, et al. Importance of achieving stringent complete response after autologous stem-cell transplantation in multiple myeloma. J Clin Oncol. 2013;31(36):4529–4535.
    1. Alhaj Moustafa M, Rajkumar SV, Dispenzieri A, Gertz MA, Lacy MQ, Buadi FK, et al. Utility of serum free light chain measurements in multiple myeloma patients not achieving complete response to therapy. Leukemia. 2015;29(10):2033–2038.
    1. Paiva B, Corchete LA, Vidriales M-B, Puig N, Maiso P, Rodriguez I, et al. Phenotypic and genomic analysis of multiple myeloma minimal residual disease tumor cells: a new model to understand chemoresistance. Blood. 2016;127(15):1896–1907.
    1. 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(10):2094–2103.
    1. Anderson KC, Auclair D, Kelloff GJ, Sigman CC, Avet-Loiseau H, Farrell AT, et al. The role of minimal residual disease testing in myeloma treatment selection and drug development: current value and future applications. Clin Cancer Res. 2017;23(15):3980–3993.
    1. Paiva B, Vidriales M-B, Pérez JJ, Mateo G, Montalbán MA, Mateos MV, et al. Multiparameter flow cytometry quantification of bone marrow plasma cells at diagnosis provides more prognostic information than morphological assessment in myeloma patients. Haematologica. 2009;94(11):1599–1602.
    1. Rawstron AC, Child JA, de Tute RM, Davies FE, Gregory WM, Bell SE, et al. Minimal residual disease assessed by multiparameter flow cytometry in multiple myeloma: impact on outcome in the Medical Research Council Myeloma IX Study. J Clin Oncol. 2013;31(20):2540–2547.
    1. Munshi NC, Avet-Loiseau H, Anderson KC, Neri P, Paiva B, Samur M, 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(23):5988–5999.
    1. Johnsen HE, Bøgsted M, Klausen TW, Gimsing P, Schmitz A, Kjaersgaard E, et al. Multiparametric flow cytometry profiling of neoplastic plasma cells in multiple myeloma. Cytometry B Clin Cytom. 2010;78(5):338–347.
    1. Soh KT, Tario JD, Wallace PK. Diagnosis of plasma cell Dyscrasias and monitoring of Minimal residual disease by multiparametric flow cytometry. Clin Lab Med. 2017;37(4):821–853.
    1. Buccisano F, Maurillo L, Schuurhuis GJ, Del Principe MI, Di Veroli A, Gurnari C, et al. The emerging role of measurable residual disease detection in AML in morphologic remission. Semin Hematol. 2019;56(2):125–130.
    1. Bassan R, Brüggemann M, Radcliffe H-S, Hartfield E, Kreuzbauer G, Wetten S. A systematic literature review and meta-analysis of minimal residual disease as a prognostic indicator in adult B-cell acute lymphoblastic leukemia. Haematologica. 2019;104(10):2028–2039.
    1. Del Giudice I, Raponi S, Della Starza I, De Propris MS, Cavalli M, De Novi LA, et al. Minimal residual disease in chronic lymphocytic leukemia: a new goal? Front Oncol. 2019;9(689):1–16.
    1. 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. JAMA Oncol. 2017;3(1):28.
    1. Oliva S, Gambella M, Gilestro M, Muccio VE, Gay F, Drandi D, et al. Minimal residual disease after transplantation or lenalidomide-based consolidation in myeloma patients: a prospective analysis. Oncotarget. 2017;8(4):5924–5935.
    1. Ferrero S, Ladetto M, Drandi D, Cavallo F, Genuardi E, Urbano M, et al. Long-term results of the GIMEMA VEL-03-096 trial in MM patients receiving VTD consolidation after ASCT: MRD kinetics’ impact on survival. Leukemia. 2015;29(3):689–695.
    1. Gu J, Liu J, Chen M, Huang B, Li J. Longitudinal flow cytometry identified “Minimal residual disease” (MRD) evolution patterns for predicting the prognosis of patients with transplant-eligible multiple myeloma. Biol Blood Marrow Transplant. 2018;000:1–7.
    1. Cavo M, Gay F, Beksac M, Pantani L, Petrucci MT, Dimopoulos MA, et al. Autologous haematopoietic stem-cell transplantation versus bortezomib–melphalan–prednisone, with or without bortezomib–lenalidomide–dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a mult. Lancet Haematol. 2020;7(6):e456–e468. doi: 10.1016/S2352-3026(20)30099-5.
    1. Rajkumar SV, Harousseau JL, Durie B, Anderson KC, Dimopoulos M, Kyle R, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the international myeloma workshop consensus panel 1. Blood. 2011;117(18):4691–4695.
    1. Durie BGM, Harousseau JL, Miguel JS, Bladé J, Barlogie B, Anderson K, et al. International uniform response criteria for multiple myeloma. Leukemia. 2006;20(9):1467–1473.
    1. Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia. 2009;23(1):3–9.
    1. Kalina T, Flores-Montero J, van der Velden VHJ, Martin-Ayuso M, Böttcher S, Ritgen M, et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia. 2012;26(9):1986–2010.
    1. Hofste op Bruinink D, Oliva S, Rihova L, van der Holt B, Gilestro M, te Marvelde JG, et al. Flowcytometric Minimal residual disease assessment in the EMN-02/HOVON-95 MM trial: used methods and a comparison of their sensitivity. Blood. 2016;128(22):2072.
    1. Almeida J, Oreao A, Ocqueteau M, Mateo G, Corral M, Caballero MD, et al. High-sensitive immunophenotyping and DNA ploidy studies for the investigation of minimal residual disease in multiple myeloma. Br J Haematol. 1999;107(1):121–131.
    1. Pérez-Persona E, Vidriales M-B, Mateo G, García-Sanz R, Mateos M-V, de Coca AG, et al. New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells. Blood. 2007;110(7):2586–2592.
    1. Arroz M, Came N, Lin P, Chen W, Yuan C, Lagoo A, et al. Consensus guidelines on plasma cell myeloma minimal residual disease analysis and reporting. Cytom Part B Clin Cytom. 2016;90(1):31–39.
    1. Jelinek T, Bezdekova R, Zatopkova M, Burgos L, Simicek M, Sevcikova T, et al. Current applications of multiparameter flow cytometry in plasma cell disorders. Blood Cancer J. 2017;7(10):e617.
    1. Op Bruinink DH, Oliva S, Rihova L, Schmitz A, Gilestro M, Te Marvelde J, et al. Standardization of flow cytometric minimal residual disease assessment in international clinical trials. A feasibility study from the European myeloma network. Haematologica. 2020;106(5):1496–1499.
    1. Rasche L, Alapat D, Kumar M, Gershner G, McDonald J, Wardell CP, et al. Combination of flow cytometry and functional imaging for monitoring of residual disease in myeloma. Leukemia. 2019;33(7):1713–1722.
    1. Pugh TJ. Circulating tumour DNA for detecting Minimal residual disease in multiple myeloma. Semin Hematol. 2018;55(1):38–40. doi: 10.1053/j.seminhematol.2018.03.002.
    1. 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(20):3073–3079.
    1. Munshi NC, Anderson KC. Minimal residual disease in multiple myeloma. J Clin Oncol. 2013;31(20):2523–2526.
    1. Oliva S, Hofste op Bruinink D, Řĺhová L, Spada S, van der Holt B, Troia R, et al. Minimal residual disease (MRD) monitoring by multiparameter flow cytometry (MFC) in newly diagnosed transplant eligible multiple myeloma (MM) patients: results from the EMN02/HO95 phase 3 trial. J Clin Oncol. 2017;35(15_suppl):8011.
    1. Schinke C, Hoering A, Wang H, Carlton V, Thanandrarajan S, Deshpande S, et al. The prognostic value of the depth of response in multiple myeloma depends on the time of assessment, risk status and molecular subtype. Haematologica. 2017;102(8):e313–e316.
    1. Paiva B, Puig N, Cedena M-TT, Rosiñol L, Cordón L, Vidriales M-BB, et al. Measurable residual disease by next-generation flow cytometry in multiple myeloma. J Clin Oncol. 2019;38(8):JCO.19.01231.
    1. Scott SD, Fletcher M, Whitehouse H, Whitby L, Yuan C, Mazzucchelli S, et al. Assessment of plasma cell myeloma minimal residual disease testing by flow cytometry in an international inter-laboratory study: is it ready for primetime use? Cytom Part B Clin Cytom. 2019;96(3):201–208. doi: 10.1002/cyto.b.21754.
    1. Gay F, Larocca A, Wijermans P, Cavallo F, Rossi D, Schaafsma R, et al. Complete response correlates with long-term progression-free and overall survival in elderly myeloma treated with novel agents: analysis of 1175 patients. Blood. 2011;117(11):3025–3031.
    1. Martinez-Lopez J, Paiva B, Lopez-Anglada L, Mateos M-V, Cedena T, Vidriales M-B, et al. Critical analysis of the stringent complete response in multiple myeloma: contribution of sFLC and bone marrow clonality. Blood. 2015;126(7):858–862.
    1. Mainou M, Madenidou A-V, Liakos A, Paschos P, Karagiannis T, Bekiari E, et al. Association between response rates and survival outcomes in patients with newly diagnosed multiple myeloma. A systematic review and meta-regression analysis. Eur J Haematol. 2017;98(6):563–568.
    1. García de Veas Silva JL, Bermudo Guitarte C, Menéndez Valladares P, Rojas Noboa JC, Kestler K, Duro Millán R. Prognostic value of serum free light chains measurements in multiple myeloma patients. Lafrenie RM, PLoS One 2016;11(11):e0166841. Available from: 10.1371/journal.pone.0166841
    1. Brioli A, Giles H, Pawlyn C, Campbell JP, Kaiser MF, Melchor L, et al. Serum free immunoglobulin light chain evaluation as a marker of impact from intraclonal heterogeneity on myeloma outcome. Blood. 2014;123(22):3414–3419.
    1. Schaar CG, Kluin-Nelemans JC, Le Cessie S, Franck PFH, Te Marvelde MC, Wijermans PW. Early response to therapy and survival in multiple myeloma. Br J Haematol. 2004;125(2):162–166.
    1. Yan Y, Mao X, Liu J, Fan H, Du C, Li Z, et al. The impact of response kinetics for multiple myeloma in the era of novel agents. Blood Adv. 2019;3(19):2895–2904.
    1. Mishima Y, Paiva B, Shi J, Park J, Manier S, Takagi S, et al. The mutational landscape of circulating tumor cells in multiple myeloma Europe PMC funders group. Cell Rep. 2017;19(1):218–224.
    1. Gendron N, Chahabi SZ, Poenou G, Rivet N, Belleville-Rolland T, Lemaire P, et al. Pain assessment and factors influencing pain during bone marrow aspiration: a prospective study. PLoS One. 2019;14(8):1–13.
    1. Sanoja-Flores L, Flores-Montero J, Garcés JJ, Paiva B, Puig N, García-Mateo A, et al. Next generation flow for minimally-invasive blood characterization of MGUS and multiple myeloma at diagnosis based on circulating tumor plasma cells (CTPC) Blood Cancer J. 2018;8(12):117. doi: 10.1038/s41408-018-0153-9.
    1. Puig N, Mateos M-V, Contreras T, Paiva B, Cedena MT, Pérez JJ, et al. Qip-mass spectrometry in high risk smoldering multiple myeloma patients included in the GEM-CESAR trial: comparison with conventional and Minimal residual disease IMWG response assessment. Blood. 2019;134(Supplement_1):581.
    1. 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(5):904–907.

Source: PubMed

3
Sottoscrivi