392
Views
2
CrossRef citations to date
0
Altmetric
Reviews

Measurable residual disease in multiple myeloma and light chain amyloidosis: more than meets the eye

ORCID Icon & ORCID Icon
Pages 1544-1553 | Received 22 Oct 2020, Accepted 04 Jan 2021, Published online: 28 Jan 2021

References

  • Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med. 2019;380(22):2104–2115.
  • Mateos MV, Cavo M, Blade J, et al. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial. Lancet (London, England). 2020;395(10218):132–141.
  • Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood. 2020;136(8):936–945.
  • Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet (London, England). 2019;394(10192):29–38.
  • Paquin A, Visram A, Kumar SK, et al. Characteristics of exceptional responders to autologous stem cell transplantation in multiple myeloma. Blood Cancer J. 2020;10(8):87.
  • Cedena MT, Martin-Clavero E, Wong S, et al. The clinical significance of stringent complete response in multiple myeloma is surpassed by minimal residual disease measurements. PloS One. 2020;15(8):e0237155.
  • Rasche L, Alapat D, Kumar M, et al. Combination of flow cytometry and functional imaging for monitoring of residual disease in myeloma. Leukemia. 2019;33(7):1713–1722.
  • 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(1):18–30.
  • Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17(8):e328–e46.
  • Kapoor P, Kumar SK, Dispenzieri A, et al. Importance of achieving stringent complete response after autologous stem-cell transplantation in multiple myeloma. J Clin Oncol. 2013;31(36):4529–4535.
  • Perrot A, Lauwers-Cances V, Corre J, et al. Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma. Blood. 2018;132(23):2456–2464.
  • Rawstron AC, Child JA, de Tute RM, 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.
  • Paiva B, Cedena MT, Puig N, et al. Minimal residual disease monitoring and immune profiling in multiple myeloma in elderly patients. Blood. 2016;127(25):3165–3174.
  • Martinez-Lopez J, Wong SW, Shah N, et al. Clinical value of measurable residual disease testing for assessing depth, duration, and direction of response in multiple myeloma. Blood Adv. 2020;4(14):3295–3301.
  • Paiva B, Puig N, Cedena MT, on behalf of the GEM (Grupo Español de Mieloma)/PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group, et al. Measurable residual disease by next-generation flow cytometry in multiple myeloma. J Clin Oncol. 2020;38(8):784–792.
  • Munshi NC, Avet-Loiseau H, Rawstron AC, et al. Association of minimal residual disease with superior survival outcomes in patients with multiple myeloma: a meta-analysis. JAMA Oncol. 2017;3(1):28–35.
  • Lahuerta JJ, Paiva B, Vidriales MB, on behalf of the GEM (Grupo Español de Mieloma)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Group, et al. Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol. 2017;35(25):2900–2910.
  • Kunacheewa C, Lee HC, Patel K, et al. Minimal residual disease negativity does not overcome poor prognosis in high-risk multiple myeloma: a single-center retrospective study. Clin Lymphoma Myeloma Leuk. 2020;20(5):e221–e38.
  • Munshi MNC, Avet-Loiseau H, Anderson KC, et al. Expanded meta-analyses confirms the association between MRD and long-term survival outcomes in multiple myeloma (MM). Blood. 2019;134(Supplement_1):4742.
  • Flores-Montero J, Sanoja-Flores L, Paiva B, et al. Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia. 2017;31(10):2094–2103.
  • Roshal M, Flores-Montero JA, Gao Q, et al. MRD detection in multiple myeloma: comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Advances. 2017;1(12):728–732.
  • Terpos E, Kostopoulos IV, Kastritis E, et al. Impact of minimal residual disease detection by next-generation flow cytometry in multiple myeloma patients with sustained complete remission after frontline therapy. Hemasphere. 2019;3(6):e300.
  • Martinez-Lopez J, Sanchez-Vega B, Barrio S, 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(6):1446–1449.
  • Medina A, Puig N, Flores-Montero J, 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(10):108.
  • Kriegsmann K, Hundemer M, Hofmeister-Mielke N, for the German-speaking Myeloma Multicenter Group (GMMG), et al. Comparison of NGS and MFC methods: key metrics in multiple myeloma MRD assessment. Cancers. 2020;12(8):2322.
  • Avet-Loiseau H, Corre J, Lauwers-Cances V, et al. Evaluation of minimal residual disease (MRD) by next generation sequencing (NGS) is highly predictive of progression free survival in the IFM/DFCI 2009 trial. Blood. 2015;126(23):191.
  • 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. 2020;JCO2000386.
  • Moreau P, Attal M, Caillot D, et al. Prospective evaluation of magnetic resonance imaging and [18F]Fluorodeoxyglucose Positron emission tomography-computed tomography at diagnosis and before maintenance therapy in symptomatic patients with multiple myeloma included in the IFM/DFCI 2009 trial: results of the IMAJEM study. J Clin Oncol. 2017;35(25):2911–2918.
  • Moreau MP, Zweegman S, Perrot A, et al. Evaluation of the prognostic value of positron emission tomography-computed tomography (PET-CT) at diagnosis and follow-up in transplant-eligible newly diagnosed multiple myeloma (TE NDMM) Patients treated in the phase 3 cassiopeia study: results of the Cassiopet Companion Study. Blood. 2019;134(Supplement_1):692.
  • Pawlyn C, Fowkes L, Otero S, et al. Whole-body diffusion-weighted MRI: a new gold standard for assessing disease burden in patients with multiple myeloma? Leukemia. 2016;30(6):1446–1448.
  • Sachpekidis C, Mosebach J, Freitag MT, et al. Application of (18)F-FDG PET and diffusion weighted imaging (DWI) in multiple myeloma: comparison of functional imaging modalities. Am J Nucl Med Mol Imaging. 2015;5(5):479–492.
  • 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(1):30–34.
  • Ulaner GA, Sobol NB, O'Donoghue JA, et al. CD38-targeted immuno-PET of multiple myeloma: from xenograft models to first-in-human imaging. Radiology. 2020;295(3):606–615.
  • Derman B, McIver A, Jiang K, et al. Measurable residual disease (MRD) assessed by mass spectrometry (MS) in peripheral blood versus next generation sequencing (NGS) in bone marrow in multiple myeloma treated on phase II trial of KRd + ASCT. J Clin Oncol. 2020;38(supp 15).
  • Eveillard M, Rustad E, Roshal M, 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.
  • Noemi Puig MP, Mateos M-V, Contreras T, 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.
  • Kumar SK, Jacobus SJ, Cohen AD, et al. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020;21(10):1317–1330.
  • Gay F, Teresa Petrucci M, Zambello R, et al. Efficacy of carfilzomib lenalidomide dexamethasone (KRd) with or without transplantation in newly diagnosed myeloma according to risk status: results from the FORTE trial. J Clin Oncol. 2020;37(15).
  • Facon T, Lee JH, Moreau P, et al. Carfilzomib or bortezomib with melphalan-prednisone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood. 2019;133(18):1953–1963.
  • Costa LJ, Chhabra S, Godby KN, et al. 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 myeloma (NDMM). Blood. 2019;134(supp 1):860.
  • Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017;376(14):1311–1320.
  • Cavo M, Gay F, Beksac M, 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 multicentre, randomised, open-label, phase 3 study. Lancet Haematol. 2020;7(6):e456–e68.
  • Gay F, Oliva S, Petrucci MT, et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol. 2015;16(16):1617–1629.
  • Bahlis NJ, Dimopoulos MA, White DJ, et al. Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study. Leukemia. 2020;34(7):1875–1884.
  • Dimopoulos M, Quach H, Mateos MV, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. Lancet (London, England). 2020;396(10245):186–197.
  • Attal M, Richardson PG, Rajkumar SV, et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet (London, England). 2019;394(10214):2096–2107.
  • Moreau P, Cavo M, de La Rubia J, et al. Evaluation of minimal residual disease in relapsed/refractory multiple myeloma patients treated with venetoclax or placebo in combination with bortezomib and dexamethasone: BELLINI study analyses. J Clin Oncol. 2020;38(Supplement_15):8547.
  • Spencer A, Lentzsch S, Weisel K, et al. Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of CASTOR. Haematologica. 2018;103(12):2079–2087.
  • Berdeja JG, Usmani SZ, Singh I, et al. Update of CARTITUDE-1: a phase Ib/II study of JNJ-4528, a B-cell maturation antigen (BCMA)-directed CAR-T-cell therapy, in relapsed/refractory multiple myeloma. J Clin Oncol. 2020;38(Supplement_15):8505.
  • Munshi NC, Jr, Shah N, Jagannath S, et al., on behalf of the KarMMa Study Investigators. Idecabtagene vicleucel (ide-cel; bb2121), a BCMA-targeted CAR T-cell therapy, in patients with relapsed and refractory multiple myeloma (RRMM): Initial KarMMa results. J Clin Oncol. 2020;38(Supplement_15):8503.
  • Mateos M-V, Otero PR, Gonzalez-Calle V, et al. 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. 2019;134(Supplement_1):781.
  • Mateos MV, Hernández MT, Giraldo P, et al. Lenalidomide plus dexamethasone versus observation in patients with high-risk smouldering multiple myeloma (QuiRedex): long-term follow-up of a randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17(8):1127–1136.
  • Lonial S, Jacobus S, Fonseca R, et al. Randomized trial of lenalidomide versus observation in smoldering multiple myeloma. J Clin Oncol. 2020;38(11):1126–1137.
  • Sidiqi MH, Aljama MA, Jevremovic D, et al. Prognostic significance of stringent complete response after stem cell transplantation in immunoglobulin light chain amyloidosis. Biol Blood Marrow Transplant. 2018;24(11):2360–2364.
  • Landau H, Lahoud O, Devlin S, et al. Pilot study of bortezomib and dexamethasone pre- and post-risk-adapted autologous stem cell transplantation in AL amyloidosis. Biol Blood Marrow Transplant. 2020;26(1):204–208.
  • Kastritis E, Dialoupi I, Gavriatopoulou M, et al. Primary treatment of light-chain amyloidosis with bortezomib, lenalidomide, and dexamethasone. Blood Adv. 2019;3(20):3002–3009.
  • Sidana S, Muchtar E, Sidiqi MH, et al. Impact of minimal residual negativity using next generation flow cytometry on outcomes in light chain amyloidosis. Am J Hematol. 2020;95(5):497–502.
  • Kastritis E, Kostopoulos IV, Theodorakakou F, et al. Next generation flow cytometry for MRD detection in patients with AL amyloidosis. Amyloid. 2020:1–5. DOI:10.1080/13506129.2020.1802713
  • Staron A, Burks EJ, Lee JC, et al. Assessment of minimal residual disease using multiparametric flow cytometry in patients with AL amyloidosis. Blood Adv. 2020;4(5):880–884.
  • Muchtar E, Dispenzieri A, Jevremovic D, et al. Survival impact of achieving minimal residual negativity by multi-parametric flow cytometry in AL amyloidosis. Amyloid. 2020;27(1):13–16.
  • Dispenzieri A, Arendt B, Dasari S, Kohlhagen M, et al. Blood mass spectrometry detects residual disease better than standard techniques in light-chain amyloidosis. Blood Cancer J. 2020;10(2):20.
  • Facon T, Dimopoulos MA, Dispenzieri A, et al. Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma. Blood. 2018;131(3):301–310.
  • Bonello F, Pulini S, Ballanti S, et al. Lenalidomide maintenance with or without prednisone in newly diagnosed myeloma patients: a pooled analysis. Cancers. 2019;11(11):1735.
  • Fonseca R, Abouzaid S, Bonafede M, et al. Trends in overall survival and costs of multiple myeloma, 2000-2014. Leukemia. 2017;31(9):1915–1921.
  • Korde N, Roschewski M, Zingone A, et al. Treatment with carfilzomib-lenalidomide-dexamethasone with lenalidomide extension in patients with smoldering or newly diagnosed multiple myeloma. JAMA Oncol. 2015;1(6):746–754.
  • Palumbo A, Gay F, Cavallo F, et al. Continuous therapy versus fixed duration of therapy in patients with newly diagnosed multiple myeloma. J Clin Oncol. 2015;33(30):3459–3466.
  • Goldschmidt H, Mai EK, Dürig J, German-speaking Myeloma Multicenter Group (GMMG), et al. Response-adapted lenalidomide maintenance in newly diagnosed myeloma: results from the phase III GMMG-MM5 trial. Leukemia. 2020;34(7):1853–1865.
  • 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(24):2218–2222.
  • Thakral D, Das N, Basnal A, et al. Cell-free DNA for genomic profiling and minimal residual disease monitoring in Myeloma- are we there yet? Am J Blood Res. 2020;10(3):26–45.
  • 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(21):2811–2813.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.