MRD in Autografts Might Predict Clinical Outcome: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: ,

Proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies (triple class drugs), and autologous stem cell transplantation (ASCT) are promising myeloma treatments that have resulted in minimal residual disease (MRD) negativity and improvement in the bone marrow microenvironment.

  • multiple myeloma
  • ultra-high-risk chromosomal abnormalities
  • minimal residual disease
  • autograft

1. Introduction

Multiple myeloma (MM) is an incurable hematologic malignancy, although the development of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), autologous stem cell transplantation (ASCT), and monoclonal antibody (MoAb) drugs has prolonged survival in patients [1]. Previously, researchers reviewed the literature regarding the total therapy strategy, including PIs, IMiDs, anti-CD38 MoAb, and ASCT, which not only induce a therapeutic effect on the myeloma cells but also improve the bone marrow microenvironment, including the enhancement of anti-myeloma immunological activity and the suppression of inhibitory anti-myeloma immunological effects [2][3]. Furthermore, it has recently been suggested that minimal residual disease (MRD) negativity is a surrogate marker for prolonged survival [4][5]. In fact, several studies have shown that myeloma cells detected as MRD may develop drug resistance and affect the surrounding immune environment.
The Monoclonal Antibody-Based Sequential Therapy for Deep Remission in Multiple Myeloma (MASTER) trial, a phase II study, demonstrated that a four-drug combination of daratumumab (DARA), carfilzomib (CFZ), lenalidomide (LEN), and dexamethasone (DEX) (D-KRd) followed by ASCT and subsequent D-KRd consolidation therapy is effective for patients with newly diagnosed MM (NDMM). Once a patient entered an MRD surveillance (MRD-SURE) phase, after bone marrow samples tested MRD-negative twice consecutively, the treatment was discontinued [6]. In the MASTER study, MRD-SURE was achieved in most patients, and the survival outcome was excellent in patients with zero or one high-risk chromosomal aberration (HRCA). However, patients with two or more HRCAs, who were considered to have ultra-high-risk chromosomal aberrations (UHRCAs), not only had a low MRD negativity rate but also achieved MRD-SURE relapse in some cases. That is, the MASTER trial results suggest that other strategies are necessary for patients with UHRCAs, even if the total therapeutic strategy concerning the bone marrow microenvironment was performed and MRD negativity was achieved [6].

2. MRD in Autografts Might Predict Clinical Outcome

In the MASTER trial, the MRD negativity rate after D-KRd induction therapy was lower in patients with UHRCAs than in the other CA groups, indicating that the myeloma cells are more frequently contaminated in the former than in the latter [6]. In two clinical studies, the MRD status of the autograft correlated with the survival time after ASCT [7][8]. Therefore, researchers discuss the role of MRD eradication in autografts in achieving persistent MRD negativity, particularly in patients with UHRCAs. In the MASTER trial, consecutive MRD assessments after D-KRd induction, ASCT, and four or eight cycles of D-KRd consolidation were performed using bone marrow samples. Before PIs and IMiDs were available, the presence of MRD in autografts was not associated with subsequent survival [9]. However, researchers considered that the contamination of myeloma cells not affecting the clinical outcomes in several previous trials because the therapeutic efficacy of conventional cytotoxic induction treatments, such as vincristine, doxorubicin, and dexamethasone, was inferior to the current induction therapy using novel agents [9][10][11][12].
In contrast, a single-center, retrospective analysis from Japan reported that the presence of MRD in autografts detected by next-generation sequencing (NGS) or real-time PCR predicts shorter OS and PFS after ASCT, and the lower levels of MRD in autografts are associated with longer OS and PFS [7]. In a retrospective study of patients with NDMM who underwent ASCT, those who achieved MRD negativity before ASCT had prolonged PFS compared with those who achieved MRD negativity after ASCT, suggesting that the achievement of earlier MRD negativity might be associated with a good response to induction therapy and that the incidence of MM cell contamination in the autograft might be lower in patients with MRD negativity before ASCT, leading to improved prognosis after ASCT [13]. Recently, two retrospective analyses employed next-generation flow cytometry (NGF) to reveal that MRD-negativity in autografts could predict long PFS and OS after ASCT [14][15]. Additionally, according to a retrospective analysis of MDACC, MRD-negativity was identified in patients treated with VRD induction therapy and without del17p and 1q21gain. MRD-negativity in autografts could predict long PFS and OS independent of induction therapy regimens [14]. While MRD-negativity in autografts could predict long PFS, the PFS in HRCA was short compared with non-HRCA, even in the patients with MRD-negativity in autografts [15]. Notably, the autograft MRD status could not be determined based on bone marrow samples, and the association of MRD status between autograft and peripheral blood samples was not analyzed in these two studies. Thus, the achievement of MRD negativity in autografts might be essential for longer OS and PFS, although the associated clinical significance has not yet been elucidated in large-scale prospective clinical trials in the era of novel agents.
Myeloma is generally distributed throughout the bone marrow. Therefore, residual myeloma cells may be present in untested bone marrow sites or extramedullary lesions, even when the tested bone marrow samples are MRD-negative [2][16][17][18][19]. Thus, a negative MRD status in bone marrow samples after induction therapy does not necessarily correspond to negative autografts, as MRD-positive autografts indicate the presence of circulating tumor cells (CTCs) in the peripheral blood [7][20]. Therefore, treatment strategies to eliminate MRD in the autograft include in vivo purging, which involves attacking CTCs using chemotherapy before mobilization, or ex vivo purging, which involves the positive selection of CD34-positive cells in the autograft. In several trials, ex vivo purging suppressed the contamination of myeloma cells in the autograft but did not improve survival time [11][21]. Hence, it is unlikely that a survival benefit from ex vivo purging will be achieved in the era of novel agents, considering that novel methods of ex vivo purging have not been studied on a large scale. Moreover, there is no evidence that a MoAb has been approved for in vivo purging following PI and IMiD approval.
The incidence of myeloma cell contamination in autografts is higher in patients with HRCAs, such as del(13q), even before PIs and IMiDs were available [22]. This was demonstrated by the FORTE trial, in which the MRD-positive rate before maintenance therapy in double-hit patients was lower than that in patients with a single HRCA regardless of the treatment group [23]. Thus, patients with UHRCAs may be more likely to have myeloma cells in their autografts than patients without UHRCAs. Accordingly, researchers consider that it is essential to reduce the tumor burden with intensive induction therapy to obtain MRD-negative autografts, especially in patients with UHRCAs. Moreover, given that a significant association has been reported between MRD in autografts and peripheral blood, following induction therapy, MRD assessment of the peripheral blood should be performed to monitor for CTCs and prevent myeloma cell contamination in autografts [8]. Indeed, some patients with negative MRD status in autografts test positive in the bone marrow [24]. As a result, MRD assessment of autograft specimens may be the most reliable method to confirm the MRD negativity of an autograft because the MRD positivity rate in autografts might be low even in patients with positive bone marrow samples [25].

3. Analyzing MRD Status: Optimal Sample and Device for UHRCA

According to the International Myeloma Working Group (IMWG) criteria, the CR criteria are focused on three biomarkers: levels of monoclonal (M) protein (products from myeloma cells), distribution of myeloma, and presence of myeloma cells in the bone marrow [4]. Although MRD assessment is currently focused primarily on myeloma cells in the bone marrow, it may be more useful to consider their presence throughout the entire body, particularly after achieving MRD negativity in the bone marrow [4][19][26]. This is important because the site of bone marrow aspiration is not indicative of myeloma in the body, given that myeloma has a partial, not diffuse, distribution [27][28]. Moreover, EMD can be observed at relapse after MRD negativity [29], suggesting that myeloma cells can be independent of the microenvironment or escape harmful microenvironments even if the myeloma cell burden is reduced below the cutoff level of MRD negativity as detected by NGS or NGF. Although the technology for MRD detection has developed over time, MRD negativity cannot be considered a sign of eradication of all myeloma cells [30][31][32]. Thus, intensive treatment should be continued for myeloma cells in patients with UHRCAs as they tend to relapse aggressively owing to changes in the beneficial microenvironment for myeloma cells with 1q21 CA and the incidence of EMD in patients with 1q21 amp and del(17p) [33][34][35][36].
In the MASTER trial, the MRD-negativity rate as the best response was similar among SRCA, HRCA, and UHRCA; meanwhile, the PFS in the UHRCA group was shorter than those in the other groups even when MRD-SURE was achieved [32]. However, MRD assessment was performed in bone marrow samples using NGF, and patients were neither tested for myeloma disease distribution using positron emission tomography/computerized tomography (PET/CT) or magnetic resonance imaging (MRI) [27][37][38] nor for M-protein levels using mass spectrometry [39][40][41][42]. The IMWG criteria suggest combining MRD measurements using bone marrow samples with imaging MRD measurements, indicating the importance of MRD imaging to confirm the presence of extramedullary lesions [4]. Considering that myeloma cells in patients with UHRCAs frequently exhibit genomic instability and are prone to EMD complications, MRD should be analyzed using various strategies to confirm the achievement of true MRD negativity.

This entry is adapted from the peer-reviewed paper 10.3390/cancers15092418

References

  1. Palumbo, A.; Anderson, K. Multiple Myeloma. N. Engl. J. Med. 2011, 364, 1046–1060.
  2. Suzuki, K.; Nishiwaki, K.; Yano, S. Treatment Strategy for Multiple Myeloma to Improve Immunological Environment and Maintain MRD Negativity. Cancers 2021, 13, 4867.
  3. Suzuki, K.; Nishiwaki, K.; Yano, S. Treatment Strategies Considering Micro-Environment and Clonal Evolution in Multiple Myeloma. Cancers 2021, 13, 215.
  4. Kumar, S.; Paiva, B.; Anderson, K.C.; Durie, B.; Landgren, O.; Moreau, P.; Munshi, N.; Lonial, S.; Bladé, J.; Mateos, M.-V.; et al. International Myeloma Working Group Consensus Criteria for Response and Minimal Residual Disease Assessment in Multiple Myeloma. Lancet Oncol. 2016, 17, e328–e346.
  5. Munshi, N.C.; Avet-Loiseau, H.; Anderson, K.C.; Neri, P.; Paiva, B.; Samur, M.; Dimopoulos, M.; Kulakova, M.; Lam, A.; Hashim, 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, 5988–5999.
  6. Costa, L.J.; Chhabra, S.; Medvedova, E.; Dholaria, B.R.; Schmidt, T.M.; Godby, K.N.; Silbermann, R.; Dhakal, B.; Bal, S.; Giri, S.; et al. Daratumumab, Carfilzomib, Lenalidomide, and Dexamethasone with Minimal Residual Disease Response-Adapted Therapy in Newly Diagnosed Multiple Myeloma. J. Clin. Oncol. 2021, 40, 2901–2912.
  7. Takamatsu, H.; Takezako, N.; Zheng, J.; Moorhead, M.; Carlton, V.E.H.; Kong, K.A.; Murata, R.; Ito, S.; Miyamoto, T.; Yokoyama, K.; et al. Prognostic Value of Sequencing-Based Minimal Residual Disease Detection in Patients with Multiple Myeloma Who Underwent Autologous Stem-Cell Transplantation. Ann. Oncol. 2017, 28, 2503–2510.
  8. Kostopoulos, I.V.; Eleutherakis-Papaiakovou, E.; Rousakis, P.; Ntanasis-Stathopoulos, I.; Panteli, C.; Orologas-Stavrou, N.; Kanellias, N.; Malandrakis, P.; Liacos, C.I.; Papaioannou, N.E.; et al. Aberrant Plasma Cell Contamination of Peripheral Blood Stem Cell Autografts, Assessed by Next-Generation Flow Cytometry, Is a Negative Predictor for Deep Response Post Autologous Transplantation in Multiple Myeloma; A Prospective Study in 199 Patients. Cancers 2021, 13, 4047.
  9. Waszczuk-Gajda, A.; Feliksbrot-Bratosiewicz, M.; Król, M.; Snarski, E.; Drozd-Sokołowska, J.; Biecek, P.; Król, M.; Lewandowski, Z.; Peradzyńska, J.; Jędrzejczak, W.W.; et al. Influence of Clonal Plasma Cell Contamination of Peripheral Blood Stem Cell Autografts on Progression and Survival in Multiple Myeloma Patients after Autologous Peripheral Blood Stem Cell Transplantation in Long-Term Observation. Transplant. Proc. 2018, 50, 2202–2211.
  10. Ho, J.; Yang, L.; Banihashemi, B.; Martin, L.; Halpenny, M.; Atkins, H.; Sabloff, M.; McDiarmid, S.A.; Huebsch, L.B.; Bence-Bruckler, I.; et al. Contaminating Tumour Cells in Autologous PBSC Grafts Do Not Influence Survival or Relapse Following Transplant for Multiple Myeloma or B-Cell Non-Hodgkin’s Lymphoma. Bone Marrow Transplant. 2009, 43, 223–228.
  11. Bourhis, J.H.; Bouko, Y.; Koscielny, S.; Bakkus, M.; Greinix, H.; Derigs, G.; Salles, G.; Feremans, W.; Apperley, J.; Samson, D.; et al. Relapse Risk after Autologous Transplantation in Patients with Newly Diagnosed Myeloma Is Not Related with Infused Tumor Cell Load and the Outcome Is Not Improved by CD34+ Cell Selection: Long Term Follow-Up of an EBMT Phase III Randomized Study. Haematologica 2007, 92, 1083–1090.
  12. Narayanasami, U.; Kanteti, R.; Morelli, J.; Klekar, A.; Al-Olama, A.; Keating, C.; O’Connor, C.; Berkman, E.; Erban, J.K.; Sprague, K.A.; et al. Randomized Trial of Filgrastim versus Chemotherapy and Filgrastim Mobilization of Hematopoietic Progenitor Cells for Rescue in Autologous Transplantation. Blood 2001, 98, 2059–2064.
  13. Baumelou, M.; Payssot, A.; Row, C.; Racine, J.; Lafon, I.; Bastie, J.N.; Chevreux, S.; Chrétien, M.L.; Maynadié, M.; Caillot, D.; et al. Early Achievement of Measurable Residual Disease Negativity in the Treatment of Multiple Myeloma as Predictor of Outcome. Br. J. Haematol. 2022, 197, e82–e85.
  14. Pasvolsky, O.; Milton, D.R.; Rauf, M.; Ghanem, S.; Masood, A.; Mohamedi, A.H.; Tanner, M.R.; Bashir, Q.; Srour, S.A.; Saini, N.; et al. Impact of Presence and Amount of Clonal Plasma Cells in Autografts Affect Outcomes in High-Risk Multiple Myeloma Patients Undergoing Autologous Hematopoietic Stem Cell Transplant. Blood 2022, 140 (Suppl. S1), 284–286.
  15. Nishimura, N.; Brown, S.; Devlin, S.M.; Dahi, P.B.; Landau, H.; Lahoud, O.B.; Scordo, M.; Shah, G.L.; Hassoun, H.; Hultcrantz, M.; et al. Stem Cell Autograft Minimal Residual Disease Negativity Improves Outcomes after Autotransplant for Multiple Myeloma. Blood 2022, 140 (Suppl. S1), 620–622.
  16. Bertamini, L.; D’Agostino, M.; Gay, F. MRD Assessment in Multiple Myeloma: Progress and Challenges. Curr. Hematol. Malig. Rep. 2021, 16, 162–171.
  17. Sanoja-Flores, L.; Flores-Montero, J.; Puig, N.; Contreras-Sanfeliciano, T.; Pontes, R.; Corral-Mateos, A.; García-Sánchez, O.; Díez-Campelo, M.; Pessoa de Magalhães, R.J.; García-Martín, L.; et al. Blood Monitoring of Circulating Tumor Plasma Cells by Next Generation Flow in Multiple Myeloma after Therapy. Blood 2019, 134, 2218–2222.
  18. Mazzotti, C.; Buisson, L.; Maheo, S.; Perrot, A.; Chretien, M.L.; Leleu, X.; Hulin, C.; Manier, S.; Hébraud, B.; Roussel, M.; 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, 2811–2813.
  19. Costa, L.J.; Derman, B.A.; Bal, S.; Sidana, S.; Chhabra, S.; Silbermann, R.; Ye, J.C.; Cook, G.; Cornell, R.F.; Holstein, S.A.; et al. International Harmonization in Performing and Reporting Minimal Residual Disease Assessment in Multiple Myeloma Trials. Leukemia 2021, 35, 18–30.
  20. Moor, I.; Bacher, V.U.; Jeker, B.; Taleghani, B.M.; Mueller, B.U.; Keller, P.; Betticher, D.; Egger, T.; Novak, U.; Pabst, T. Peripheral Flow-MRD Status at the Time of Autologous Stem Cell Collection Predicts Outcome in Multiple Myeloma. Bone Marrow Transplant. 2018, 53, 1599–1602.
  21. Stewart, A.K.; Vescio, R.; Schiller, G.; Ballester, O.; Noga, S.; Rugo, H.; Freytes, C.; Stadtmauer, E.; Tarantolo, S.; Sahebi, F.; et al. Purging of Autologous Peripheral-Blood Stem Cells Using CD34 Selection Does Not Improve Overall or Progression-Free Survival after High-Dose Chemotherapy for Multiple Myeloma: Results of a Multicenter Randomized Controlled Trial. J. Clin. Oncol. 2001, 19, 3771–3779.
  22. Vogel, W.; Kopp, H.G.; Kanz, L.; Einsele, H. Myeloma Cell Contamination of Peripheral Blood Stem-Cell Grafts Can Predict the Outcome in Multiple Myeloma Patients after High-Dose Chemotherapy and Autologous Stem-Cell Transplantation. J. Cancer Res. Clin. Oncol. 2005, 131, 214–218.
  23. Gay, F.; Mina, R.; Rota-Scalabrini, D.; Galli, M.; Belotti, A.; Zamagni, E.; Bertamini, L.; Zambello, R.; Gamberi, B.; De Sabbata, G.; et al. Carfilzomib-Based Induction/Consolidation with or without Autologous Transplant (ASCT) Followed by Lenalidomide (R) or Carfilzomib-Lenalidomide (KR) Maintenance: Efficacy in High-Risk Patients. J. Clin. Oncol. 2021, 39, 8002.
  24. Bal, S.; Landau, H.J.; Shah, G.L.; Scordo, M.; Dahi, P.; Lahoud, O.B.; Hassoun, H.; Hultcrantz, M.; Korde, N.; Lendvai, N.; et al. Stem Cell Mobilization and Autograft Minimal Residual Disease Negativity with Novel Induction Regimens in Multiple Myeloma. Biol. Blood Marrow Transplant. 2020, 26, 1394–1401.
  25. Tageja, N.; Korde, N.; Kazandjian, D.; Panch, S.; Manasanch, E.; Bhutani, M.; Kwok, M.; Mailankody, S.; Yuan, C.; Stetler-Stevenson, M.; et al. Combination Therapy with Carfilzomib, Lenalidomide and Dexamethasone (KRd) Results in an Unprecedented Purity of the Stem Cell Graft in Newly Diagnosed Patients with Myeloma. Bone Marrow Transplant. 2018, 53, 1445–1449.
  26. Moreau, P.; Attal, M.; Caillot, D.; Macro, M.; Karlin, L.; Garderet, L.; Facon, T.; Benboubker, L.; Escoffre-Barbe, M.; Stoppa, A.M.; et al. Prospective Evaluation of Magnetic Resonance Imaging and 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, 2911–2918.
  27. Hillengass, J.; Usmani, S.; Rajkumar, S.V.; Durie, B.G.M.; Mateos, M.-V.; Lonial, S.; Joao, C.; Anderson, K.C.; García-Sanz, R.; Riva, E.; et al. International Myeloma Working Group Consensus Recommendations on Imaging in Monoclonal Plasma Cell Disorders. Lancet Oncol. 2019, 20, e302–e312.
  28. Kyle, R.A.; Gertz, M.A.; Witzig, T.E.; Lust, J.A.; Lacy, M.Q.; Dispenzieri, A.; Fonseca, R.; Rajkumar, S.V.; Offord, J.R.; Larson, D.R.; et al. Review of 1027 Patients with Newly Diagnosed Multiple Myeloma. Mayo Clin. Proc. 2003, 78, 21–33.
  29. Rasche, L.; Alapat, D.; Kumar, M.; Gershner, G.; McDonald, J.; Wardell, C.P.; Samant, R.; Van Hemert, R.; Epstein, J.; Williams, A.F.; et al. Combination of Flow Cytometry and Functional Imaging for Monitoring of Residual Disease in Myeloma. Leukemia 2019, 33, 1713–1722.
  30. Costa, L.J.; Chhabra, S.; Medvedova, E.; Schmidt, T.M.; Dholaria, B.; Godby, K.N.; Silbermann, R.; Bal, S.; D’Souza, A.; Giri, S.; et al. Outcomes of MRD-Adapted Treatment Modulation in Patients with Newly Diagnosed Multiple Myeloma Receiving Daratumumab, Carfilzomib, Lenalidomide and Dexamethasone (Dara-KRd) and Autologous Transplantation: Extended Follow up of the Master Trial. Blood 2022, 140 (Suppl. S1), 7275–7277.
  31. Paiva, B.; van Dongen, J.J.M.; Orfao, A. New Criteria for Response Assessment: Role of Minimal Residual Disease in Multiple Myeloma. Blood 2015, 125, 3059–3068.
  32. Rawstron, A.C.; Child, J.A.; de Tute, R.M.; Davies, F.E.; Gregory, W.M.; Bell, S.E.; Szubert, A.J.; Navarro-Coy, N.; Drayson, M.T.; Feyler, S.; 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, 2540–2547.
  33. Stork, M.; Sevcikova, S.; Minarik, J.; Krhovska, P.; Radocha, J.; Pospisilova, L.; Brozova, L.; Jarkovsky, J.; Spicka, I.; Straub, J.; et al. Identification of Patients at High Risk of Secondary Extramedullary Multiple Myeloma Development. Br. J. Haematol. 2022, 196, 954–962.
  34. Tirier, S.M.; Mallm, J.P.; Steiger, S.; Poos, A.M.; Awwad, M.H.S.; Giesen, N.; Casiraghi, N.; Susak, H.; Bauer, K.; Baumann, A.; et al. Subclone-Specific Microenvironmental Impact and Drug Response in Refractory Multiple Myeloma Revealed by Single-Cell Transcriptomics. Nat. Commun. 2021, 12, 6960.
  35. Billecke, L.; Murga Penas, E.M.; May, A.M.; Engelhardt, M.; Nagler, A.; Leiba, M.; Schiby, G.; Kröger, N.; Zustin, J.; Marx, A.; et al. Cytogenetics of Extramedullary Manifestations in Multiple Myeloma. Br. J. Haematol. 2013, 161, 87–94.
  36. Gozzetti, A.; Cerase, A.; Lotti, F.; Rossi, D.; Palumbo, A.; Petrucci, M.T.; Patriarca, F.; Nozzoli, C.; Cavo, M.; Offidani, M.; et al. Extramedullary Intracranial Localization of Multiple Myeloma and Treatment with Novel Agents: A Retrospective Survey of 50 Patients. Cancer 2012, 118, 1574–1584.
  37. Zamagni, E.; Nanni, C.; Dozza, L.; Carlier, T.; Bailly, C.; Tacchetti, P.; Versari, A.; Chauvie, S.; Gallamini, A.; Gamberi, B.; 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, 116–125.
  38. Belotti, A.; Ribolla, R.; Cancelli, V.; Villanacci, A.; Angelini, V.; Chiarini, M.; Giustini, V.; Facchetti, G.V.; Roccaro, A.M.; Ferrari, S.; et al. Predictive Role of Diffusion-Weighted Whole-Body MRI (DW-MRI) Imaging Response According to MY-RADS Criteria after Autologous Stem Cell Transplantation in Patients with Multiple Myeloma and Combined Evaluation with MRD Assessment by Flow Cytometry. Cancer Med. 2021, 10, 5859–5865.
  39. Murray, D.L.; Puig, N.; Kristinsson, S.; Usmani, S.Z.; Dispenzieri, A.; Bianchi, G.; Kumar, S.; Chng, W.J.; Hajek, R.; Paiva, B.; 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, 11, 24.
  40. Langerhorst, P.; Noori, S.; Zajec, M.; De Rijke, Y.B.; Gloerich, J.; van Gool, A.J.; Caillon, H.; Joosten, I.; Luider, T.M.; Corre, J.; et al. Multiple Myeloma Minimal Residual Disease Detection: Targeted Mass Spectrometry in Blood vs Next-Generation Sequencing in Bone Marrow. Clin. Chem. 2021, 67, 1689–1698.
  41. Liyasova, M.; McDonald, Z.; Taylor, P.; Gorospe, K.; Xu, X.; Yao, C.; Liu, Q.; Yang, L.; Atenafu, E.G.; Piza, G.; et al. A Personalized Mass Spectrometry–Based Assay to Monitor M-Protein in Patients with Multiple Myeloma (EasyM). Clin. Cancer Res. 2021, 27, 5028–5037.
  42. Abeykoon, J.P.; Murray, D.L.; Murray, I.; Jevremovic, D.; Otteson, G.E.; Dispenzieri, A.; Arendt, B.K.; Dasari, S.; Gertz, M.; Gonsalves, W.I.; et al. Implications of Detecting Serum Monoclonal Protein by MASS-Fix Following Stem Cell Transplantation in Multiple Myeloma. Br. J. Haematol. 2021, 193, 380–385.
More
This entry is offline, you can click here to edit this entry!
Video Production Service