Next Article in Journal
Functional Impairments of Amyloidosis Patients: Physical Therapy Assessment
Previous Article in Journal
miR-494 in Extracellular Vesicles as a Potent Biomarker of Chronic Myeloid Leukemia Treatment with Tyrosine Kinase Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Measurable Residual Disease Assessment in Multiple Myeloma: How Deep Is Enough?

1
Hemato-Oncology Unit, Champalimaud Foundation, 1400-038 Lisbon, Portugal
2
Myeloma Lymphoma Research Group, Champalimaud Foundation, 1400-038 Lisbon, Portugal
3
NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
*
Author to whom correspondence should be addressed.
Hemato 2022, 3(3), 385-413; https://doi.org/10.3390/hemato3030027
Submission received: 18 March 2022 / Revised: 9 June 2022 / Accepted: 21 June 2022 / Published: 23 June 2022
(This article belongs to the Section Plasma Cell Disorders)

Abstract

:
The introduction of new and more effective therapeutic options for Multiple Myeloma (MM) has significantly deepened and prolonged patients’ remission. As currently used treatment protocols induce high rates of complete responses, Measurable Residual Disease (MRD) assessment has become essential to enhance the evaluation of treatment efficacy. Detection of MRD has improved with the development of highly sensitive and standardized techniques such as Next Generation Flow or Next Generation Sequencing, complemented by functional imaging techniques. These advances offer a valuable opportunity to further optimize criteria of response to treatment. Currently, extensive data demonstrate that MRD status is a valuable prognostic factor of survival. Since MRD represents a real measurement of disease burden, its incorporation in clinical trials to guide treatment decisions will certainly translate into clinical benefits. Sustained MRD negativity can be used to consider optimal candidates for treatment discontinuation, whereas MRD positive high-risk patients may have access to novel immunotherapeutic strategies such as bispecific drugs or CAR T cell therapy. In this review, we describe the available techniques to detect MRD, address the current data regarding MRD as a surrogate endpoint within clinical trials, examine how MRD can be introduced into the clinical management of MM patients, and discuss the future of MRD monitoring.

1. Introduction

Multiple Myeloma (MM) therapy has improved considerably in the past years, with the development of multi-drug regiments and the inclusion of novel agents such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and monoclonal antibodies (mAbs) [1,2,3,4,5]. These new treatment approaches have led to high complete remission (CR) rates, associated with longer progression free survival (PFS) and prolonged overall survival (OS) [6,7]. Furthermore, increased knowledge regarding the biology of this disease, the mutational landscape, and the interplay between myeloma cells and the bone marrow (BM) microenvironment have introduced new insights to tackle this hematological malignancy in a more effective way [8,9,10]. The prospect of eliminating the malignant plasma cells and of converting MM into a fully curable disease seems within reach. Alongside innovations regarding new treatments, the development of highly sensitive techniques to evaluate its efficacy is critical. Criteria for response to therapy can no longer solely rely on traditional methods such as serum immunofixation and determination of plasma cells by morphology, which lack the sensitivity and discrimination to fully define disease status and accurately predict patient response. Therefore, the inclusion of measurable residual disease (MRD) detection to monitor patients and assess the true depth of response, opens the field towards a more personalized treatment management in MM.
This review will focus on the current available methods to detect MRD, the relevance of MRD assessment in the treatment of MM, the benefits MRD evaluation can bring to clinical research, the questions MRD can help answer in the clinical setting, and the future directions for MRD testing.

2. Methods for BM MRD Detection

The development and validation of methods for MRD detection have evolved greatly in the last years. Technological advances, with the introduction of more accurate and sensitive approaches, have paved the way towards a more effective monitoring of MRD. Currently, BM MRD evaluation and imaging-based techniques are recommended by the International Myeloma Working Group (IMWG) as the most appropriate tools to assess low levels of disease [11].

2.1. Flow Cytometry-Based MRD

Multiparameter flow cytometry (MFC) immunophenotyping is one of the most frequently used methods to study plasma cell (PC) disorders. This technique is available worldwide, allowing for the fast evaluation of a high number of cells, discriminating, characterizing, and quantifying both neoplastic and normal PCs at low frequency, by using a combination of well-defined surface and cytoplasmatic markers and modern flow cytometers. The combination of anti-CD38 and anti-CD138 mAbs are used to specifically identify PCs, and the unequivocal presence of an aberrant phenotype is commonly evaluated with discriminatory markers such as CD19, CD56, CD45, CD27, CD117, CD81, and cytoplasmic Igκ and Igλ.
Identification of the minimum number of aberrant PCs remaining after treatment is of great utility to monitor therapy efficacy and predict outcome in MM patients, and MFC is a sensitive method for analyzing the PC compartment [12]. In 2008, a study by the Spanish group showed that MRD negative patients at day 100 after Autologous Stem Cell Transplant (ASCT) had longer PFS (median 71 vs. 37 months; p < 0.001) and OS (median not reached vs. 89 months; p = 0.002). An absence of residual clonal PCs by MFC evaluation was the most relevant independent prognostic factor [13]. Similarly, MM patients that maintained MRD positivity by MFC 3 months after ASCT had shorter PFS. Being MRD positive, despite a negative immunofixation result, was associated with a worse outcome [14]. In the French IFM 2009, MRD negativity evaluated by MFC was likewise associated with higher PFS and OS at a sensitivity level of 10−4 [15]. Taken together, these results reflect the higher prognostic value and clinical relevance of MRD assessment by MFC.
The ability to quantify residual disease using MFC assays and the level of sensitivity reached have evolved over the years. First generation assays using 4 to 6 colors were progressively replaced by 2nd generation MFC using 8 colors, and now it is standard to use panels of two tubes with 8 colors or even one tube with 10 colors. The EuroFlow Consortium has developed and validated a standardized Next Generation Flow (NGF) methodology [16] included in the International Myeloma Foundation consensus criteria [17]. By using an 8-color two-tube panel, BM processing applying a bulk-lysis procedure, and a minimum recommended number of 107 cells/tube, has allowed this methodology to reach an increased level of sensitivity between 10−5 and 10−6, when enough cells are evaluated (≥5 × 106 per tube). Additionally, new software tools have been developed for automatic analysis with normal and malignant reference databases, speeding the process and avoiding biases potentially introduced by individual operator assessment [18]. Both the limit of detection and the limit of quantification can be determined based on the identification of at least 20 and 50 neoplastic PCs among 107 nucleated cells, respectively, discriminating between positive and negative MRD samples. A direct comparison between the 8-color two-tube and 10-color one-tube assays was performed, showing concordance of 98% between the two approaches. The first assay has the advantage of acquiring a higher number of cells and the possibility to have a confirmatory tube; the second assay is less time consuming and affordable [19]. The possibility of new improvements in this field leading to an increase of the level of sensitivity is not to be excluded and alternative single-tube MFC approaches have also been presented [20,21].
Normal PCs display a heterogeneous immunophenotype according to their maturation process. Knowing the expression pattern in different conditions, from reactive to regenerating BM samples, enables a clear distinction from aberrant PCs [22]. Likewise, it renders possible to identify neoplastic PCs in follow-up samples, even upon phenotypic shifts [23]. As such, one major advantage of NGF is that a diagnostic sample is unrequired. Although the introduction of new anti-CD38 Ab treatments for MM, such as daratumumab or isatuximab, might hamper the use of this marker to specifically detect PCs, this can be overcome with the use of multi-epitope CD38 Abs [24].
Additionally, a broad detection of the different cell populations present in a sample can be done, including those that permit the identification of potential hemodiluted samples (mast cells, nucleated red blood cells, myeloid, and B-cell precursors). This intra-assay quality control is especially important as hemodiluted BM aspirates may induce a false negative result. The risk can be minimized if the first BM aspirate is sent to MRD evaluation by NGF, as recommended by the recently published work on consensus on performing and reporting MRD evaluation [25].
Determining the depth of MRD negativity and the level of sensitivity that is clinically relevant is especially important. The PETHEMA/GEM2012MENOS65 trial published in 2020 showed that patients with undetectable MRD after being treated with lenalidomide–bortezomib–dexamethasone (RVd) induction, followed by consolidation with ASCT and RVd, had an 82% reduced risk of progression or death (Hazard Ratio (HR) = 0.18; 95% Confidence Interval (CI) 0.11 to 0.30; p < 0.001). An MRD negative response defined by NGF identified a group of patients with a lower risk of progression, confirming a cut-off of 10−6 as being clinically relevant [26]. In the EMN02/HO95 MM Phase 3 trial, newly diagnosed MM (NDMM) patients, who had reached CR prior to maintenance therapy and retained it during the maintenance period, had a significant reduced risk of progression or death when MRD was negative by NGF evaluation every 6 months. After a median follow-up of 75 months, the 5-year PFS was 66% in MRD negative versus 31% in MRD positive patients (HR = 0.39; p < 0.001), and the 5-year OS was 86% versus 69%, respectively (HR = 0.41; p < 0.001) [27]. Despite using lesser sensitive MFC assays, several studies have reported a better outcome for patients reaching MRD negativity [13]. Rawstron et al., demonstrated a benefit in OS per log reduction of MRD levels as evaluated by MFC (5.9 years–10−2–10−3; 6.8 years–10−3–10−4; >7.5 years-10−4) [28]. More recent trials confirmed that achieving undetectable MRD at a sensitivity of 10−5–10−6 was able to overcome the worse outcome in high-risk patients at baseline, though these patients require more intense regimens to reach these levels of MRD [26,27,29,30]. Deep response to treatment may therefore shift patients with an adverse prognosis to a more favorable one, defining a more accurate risk stratification.
Despite the increased sensitivity of NGF approaches, patients with undetectable MRD still progress, particularly those who present extramedullary disease or single focal lesions. This emphasizes the importance of combining it with complementary techniques such as high-precision imaging; a subject that will be discussed further in this review.

2.2. Molecular-Based MRD

Over the last decade, we have witnessed an improvement in molecular methods for MRD monitoring, particularly with the introduction of Next Generation Sequencing (NGS) techniques. Indeed, this molecular-based MRD assessment has replaced other laborious and low-applicable techniques, such as allele-specific quantitative polymerase chain reaction (ASO-qPCR) [31,32,33].
NGS is based on the use of consensual primers that amplify and sequence all rearranged immunoglobulin genes (IgH (VDJ), IgH (DJ), IgK, and IgL receptor gene sequences, translocated BCL1/IgH (J) and BCL2/IgH (J) sequences) on aberrant BM PCs. A baseline sample is required to identify the dominant sequence(s) used to monitor MRD after treatment. This sequencing method has shown a sensitivity up to 10−6 and is applicable in more than 90% of myeloma cases [34]. This technique has also been applied in other hematological neoplasms such as acute lymphoblastic leukemia, mantle cell lymphoma, and chronic lymphocytic leukemia [35,36,37].
In the context of MM, several groups have shown the efficacy of MRD assessment to predict patient outcome in CR and independently of disease risk. Martinez-Lopez et al., in 2014 demonstrated that deep sequencing of immunoglobulin rearranged genes can effectively identify and quantify neoplastic PCs in the BM of MM patients. More importantly, this method was shown to be a prognostic biomarker of time-to-tumor progression (TTP) and OS. In this study, three groups of patients were identified based on MRD levels: high (<10−3), intermediate (10−3 to 10−5), and low (>10−5), with significantly different TTPs of 27, 48, and 80 months (p = 0.003 to 0.0001), respectively [38]. Another study by Avet-Louiseau et al., published in 2015, evaluated NGS sensitivity using BM aspirates from the IFM/DFCI2009 clinical trial (NCT01191060). This trial included NDMM patients receiving RVd induction therapy followed by consolidation therapy with ASCT plus RVd or RVd alone and had MRD evaluation of gene rearrangement in PC performed before and after maintenance. Researchers found that, by establishing a cut-off of 10−6, patients below this value presented a 3-year PFS of 83% pre-maintenance and 90% post-maintenance. Notably, these values were similar when restricted to MM patients in CR, demonstrating that deep sequencing through NGS can predict PFS, even in patients treated with state-of-the-art strategies [15]. These results were further confirmed by Perrot et al., who demonstrated that an MRD negative status is indeed a powerful prognostic biomarker of both PFS (adjusted HR = 0.22; 95% CI, 0.15–0.34; p < 0.001) and OS (adjusted HR = 0.24; 95% CI, 0.11–0.54; p = 0.001) [39]. Overall, these studies suggest that deeper responses are associated with better patient outcome.
Furthermore, Takamatsu et al., showed that NSG-based MRD detection in autograft and BM samples from MM patients who received high-dose melphalan plus ASCT had significant prognostic value. Briefly, MRD negative patients (<10−6) by NGS showed significant better PFS (96%, p < 0.001) and OS (100%, p = 0.04) compared with MRD positive patients. Patients who underwent ASCT treatment using novel therapeutic agents and whose autografts were MRD negative (<10−7) by NGS had significant better PFS compared with untreated patients (p = 0.001). Overall, these results demonstrate a strong correlation between NGS-based MRD negativity and PFS. Conversely, ASO-qPCR showed no significant benefit compared to NGS. However, this study lacked a standardized therapy and sample size, therefore these results should be further confirmed in other prospective studies to draw stronger conclusions [40]. Nevertheless, several other randomized trials such as ALCYONE, MAIA, CASTOR, or POLLUX have demonstrated that, independently of the treatment scheme, patients who achieved MRD negativity through NGS techniques had significantly higher PFS [1,41,42]. Although BM-based MRD evaluation using NGS has shown a strong prognostic value that correlates with the majority of patients’ outcomes, this is not witnessed in a cohort of patients who, despite being MRD negative, eventually relapse [43,44]. This reflects the need to further improve currently available techniques for MRD assessment in terms of sensitivity and specificity to better stratify this group of patients.
Despite NGS being an extremely sensitive MRD detection method with the advantage that samples do not require immediate processing, contrary to NGF, some pitfalls are starting to arise. The fact that it is labor intensive, time consuming, more expensive, and less available, combined with the presence of somatic mutations which are genetic hallmarks of MM and may confound NGS results, giving rise to false-negative results due to clonal evolution [45,46]. A major disadvantage is the lack of standardization and harmonization across laboratories, including analysis of results, characteristics of tested patients, or time points assessed [47]. Therefore, it is of utmost importance that MRD testing procedures are uniformed so that data can be compared between institutions, allowing large-scale analysis and introduction into routine clinical practice [48]. An international group of 21 laboratories with expertise in NGS—EuroClonality-NGS consortium—is currently working on its standardization and has already validated assays for MRD assessment in Acute Lymphoblastic Leukemia [49].
So far, most of the published data relies on the use of the clonoSEQ platform (Adaptive Biotechnologies) [50] as the only assay cleared by the FDA for MRD assessment. The clonoSEQ Watch Registry (NCT04545333)—a prospective multicenter observational study of adult patients—is currently collecting data regarding the use of this assay in the management of lymphoid malignancies, including MM. Tests to develop non-commercial alternatives are ongoing. Recently, Martinez-Lopez et al., successfully described and validated a novel in-house NGS method to assess MRD in MM patients. This technology demonstrated high reproducibility with a sensibility of 10−5 and can be fully automated, minimizing variations across laboratories. Results showed a global concordance of 89% between in-house NGS data and flow cytometry (Spearman correlation r = 0.8; p < 0.001) [51].
Despite the data available on the performance of each BM-based methodology, information is still lacking regarding concordance between NGF and NGS. Results from the FORTE trial showed a concordance between the two methods of 86% at 10−5 (n= 335; r = 0.61) and 78% at 10−6 (n = 56; r = 0.77). In pre-maintenance samples, however, MRD assessed by MFC revealed a higher percentage of positive cases, with a discordance of 1–10% at the same level of sensitivity [50]. In the CASSIOPEIA trial, concordance was also high (92.9%) [52]. Additional clinical trials with extended follow-up periods will be needed to tackle this issue.
Taken together, NGS results are very promising, although more studies are required before widespread implementation in the routine clinical practice.

2.3. Imaging Techniques-Based MRD

Whole body imaging techniques such as 18F-Fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) and Magnetic Resonance Imaging (MRI) provide important complementary information regarding the presence of residual disease. MM is a heterogenous disease with patchy infiltration [53,54], and most patients present focal lesions detectable by imaging techniques [55,56] that constitute a challenge for intramedullary MRD methods that use a single BM aspirate.
At present, recommendations for imaging during follow-up and response evaluation of MM patients include whole-body CT or PET/CT, depending on availability of initial results at baseline for comparison. PET/CT is currently the standard of care to evaluate and determine metabolic response after treatment [57,58]. This technique combines the imaging of a molecular process (usually FDG tracer uptake) with morphologic images obtained by CT and is especially useful when addressing extramedullary disease [59,60]. MRI can detect bone involvement and the presence of extramedullary disease and provides details on soft tissue disease and patterns of BM infiltration [61]. Both MRI and PET/CT techniques showed similar results at identifying bone lesions at diagnosis, but only normalization before maintenance therapy, detected by PET/CT, showed a predictive role for PFS and OS [62]. Evaluation of treatment efficacy by PET/CT has been shown to correlate with prognosis in MM [63]. Conventional MRI might not be the optimal approach to evaluate MRD, as focal lesions remain positive for several months after therapy, even in patients who respond to treatment [64]. However, Diffusion-Weighted MRI (DWI), a functional MRI technique that measures water molecule movement in tissues, could provide useful information on residual focal lesions. This technique still requires standardization, and further studies are needed to compare DWI with 18F-FDG PET/CT and to evaluate DWI results in the context of MM after therapy [65,66]. Optimization of PET/CT is also needed. Efforts were made to standardize this technique, particularly with the Deauville scores (standardized uptake value cut-offs using the liver background as reference to identify a complete metabolic response) [67]. Erroneous results might arise due to a low expression of the hexokinase enzyme responsible for the glycolysis of the FDG tracer used [68]. Alternatives to assess disease activity in MM are currently being evaluated, such as 11C-Methionine [67]. Other PET tracers under investigation include peptide tracers such as 68Ga-Pentixafor targeting CXCR4, presently being evaluated in a Phase 2 ongoing clinical trial in NDMM patients (NCT04561492), and lipid tracers such as choline or acetate. These new tracers and molecular imaging biomarkers are currently being included in clinical trials before they can be fully adopted in clinical practice [69,70].
Recent studies have demonstrated the importance of combining PET/CT with MRD BM techniques such as NGF or NGS to evaluate treatment response. A low agreement between BM MRD and imaging techniques was previously reported in the IFM/DFCI 2009 trial, with 26% of MRD negative patients by MFC (sensitivity 10−4) and positive by PET/CT, and by the CASSIOPET sub-study of CASSIOPEIA, with 10.5% of MRD-negative patients by NGF (sensitivity 10−5) being PET/CT positive [62,71]. A study by Rasche et al., revealed that a proportion of MRD negative patients by NGF eventually relapsed with extramedullary disease, and patients who were MRD negative by both NGF and PET/CT had better PFS than those only MRD negative by NGF. There are also more discrepant cases among the relapsed/refractory MM (RRMM) group (50% versus 12% in the NDMM patients) [72]. Patients with persistent 18F-FDG PET/CT lesions after induction and subsequent ASCT had shorter 4-year estimates of PFS (32% versus 47%; p = 0.02) and OS (66% versus 79%; p = 0.02) [73]. BM involvement may not be uniform and BM examination, usually done in the easily accessible pelvic bone, may not account for residual focal lesions in other sites that can be detected by imaging techniques. The clonal heterogeneity that characterizes MM, with the existence of multiple clones and distinct molecular and biological traits, further accentuates the need to combine both approaches. Recent data regarding genetic spatial tumor heterogeneity represented by focal lesions highlight the existence of sub-clones with gene expression signatures associated with disease aggressiveness and drug resistance, strengthening the hypothesis that these focal lesion sub-clones are at the origin of MM relapse [74].
Present recommendations by the IMWG on the use of MRD in clinical trials state that both BM MRD assessment and functional imaging by PET/CT should be evaluated simultaneously [25]. A comparison of the currently recommended methods to measure MRD is summarized in Table 1.
The combination of multiple approaches will undoubtedly provide solid and substantial information to support clinical decisions, namely the safe and effective discontinuation of treatment in a specific cohort of patients. Currently, there are ongoing clinical studies (NCT04108624) to assess MRD in MM patients and to determine whether patients who are MRD negative by multiple modalities (novel imaging and laboratory techniques) can safely and effectively discontinue post-transplant maintenance therapy after receiving at least one year of maintenance therapy.
Overall, neither NGF- nor NGS-based methods, in combination with imaging techniques, associate undetectable MRD with better outcomes, revealing that the depth of response and level of sensitivity achieved is more important than the technique itself.

3. Future of MRD Testing: Beyond BM Assessment

The most sensitive methods currently available to assess MRD in MM require an invasive BM aspiration. To overcome this concern, as well as the spatial heterogeneity of MM and the frequency of MRD assessment, alternative less invasive approaches are being investigated. The use of liquid biopsies using peripheral blood as a source for circulating tumor-derived components is a promising option to tackle these issues, including the high sensitivity detection of circulating tumor plasma cells (CTPC), circulating cell-free tumor DNA (cfDNA), and serum monoclonal immunoglobulins [75,76,77,78].
The same strategy used for BM PC analysis can be applied to the detection of neoplastic PC present in PB. CTPC have been detected in a high percentage of MM cases at diagnosis [79] and the release of CTPC from the BM to PB has been associated with decreased survival [80]. Furthermore, at different treatment timepoints, a higher CTPC frequency seems to have clinical utility as an unfavorable prognostic factor in MM patients [81,82,83] and is also associated with a greater risk of progression in pre-clinical stages such as monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM (SMM) [84,85]. Recently, Gonsalves et al., revealed that the presence of ≥5 CTPC/µL at diagnosis in patients with Revised International Staging System (R-ISS) Stage I and II that were re-classified as R-ISS IIB had worse prognosis than those with R-ISS Stage III (time to next treatment: R-ISS I, 40 months; R-ISS II, 30 months; R-ISS IIB, 21 months; R-ISS III, 20 months; OS: R-ISS I, not reached; R-ISS II, 72 months; R-ISS IIB, 45 months; R-ISS III, 47 months) [86]. Another study addressing the prognostic value of CTPC in NDMM patients revealed that the combination of R-ISS Stage III and a level of CTPC ≥ 0.105% was able to stratify a subgroup of patients associated with aggressive disease (OS = 11.5 months versus 33 months; p < 0.001; PFS = 10 months versus not reached; p < 0.001) [87]. These results emphasize the utility of CTPC as a complementary prognostic tool to improve MM patient risk stratification. It is well established that CTPC can be detected with a high throughput NGF strategy [88,89]. In the study by Sanoja-Flores et al., the NGF assay was able to detect and quantify CTPC in 17% of MM patients in CR, identifying a subgroup with lower PFS (HR = 7.4; 95% CI, 3.0–18.2; p < 0.0001). Although all patients with detectable CTPC were MRD positive in the BM, sensitivity is still suboptimal and 40% of patients who were positive in the BM had undetectable neoplastic PC in PB [90]. However, these results already hint that CTPC may reflect the ability for disease dissemination, providing an early indication of progression. In fact, CTPC analysis revealed specific features in terms of phenotype and subclonal mutations, different from the PC normally present in the BM [91]. In fact, besides quantification, CTPC can be evaluated using molecular approaches and eventually used as a biomarker for response to treatment [78,92,93]. The detection of CTPC might be useful to provide a complete image of the genomic landscape of MM patients, including detection of mutations and copy number variations by whole exome sequencing. In a study by Garcés et al., the mutations present in BM myeloma cells and extramedullary plasmacytoma were also detected in CTPC (86% and 87%, respectively). A high concordance between the genetic profile of matched CTPC and BM myeloma cells was observed (82%), including mutations in genes frequently altered in MM (83%), as well as copy number alterations (95%) [94]. However, MRD evaluation relying solely on CTPC is far from a routine clinical application and needs further technical and standardization improvements.
cfDNA has been explored as a liquid biopsy in cancer [95], including MM [96], but its relevance in MRD assessment is still being discussed. Although cfDNA can be used to effectively reconstitute the genetic landscape and clonal heterogeneity of MM in the BM [97], there are dissonant results regarding its applicability as an MRD monitoring tool. Studies using NGS to compare MRD in paired cfDNA from PB and BM samples showed that 69% of patients who were MRD positive in the BM had undetectable MRD in the PB [92], and that it failed to detect IgH rearrangements in 66% of patients in CR [98]. Even with the commercially available NGS kit used for MRD monitoring, a low concordance (49%) with BM results was obtained [92]. These results could be mostly due to decreased sensibility to detect specific Ig gene rearrangements in PB when there is a low disease burden in the BM and to technical limitations of this methodology to target DNA fragments that are longer than cfDNA. More encouraging results from a long-term study using cfDNA detected by ASO-qPCR found a specificity of 83.3% and a sensitivity of 66.7%, with a significant correlation between no or low (positive non-quantifiable) cfDNA level and the number of patients reaching CR (p = 0.012) [99]. Recently, our group published a review that summarizes the current knowledge regarding the use of CTPC and cfDNA to evaluate MRD [100]. Thus far, these methodologies do not seem sufficiently mature or sensitive for widespread MRD assessment. Frequent MRD monitoring in blood is an attractive approach, and further characterization of CTPC and cfDNA can offer insights into the molecular evolution of MRD during tumor progression with potential therapeutic repercussions.
M-protein peptides detected by mass spectrometry (MS)-based techniques in the serum and urine are also emerging as a possibility to monitor the MM burden [101]. Alternative methods to the standard serum protein electrophoresis, such as matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) and liquid chromatography quadrupole time-of-flight mass spectrometry (LC-MS), have shown promising results in MRD detection [102,103]. MALDI-TOF-MS detects total mass distribution of denatured intact Ig light chains, whereas LC-MS is a clonotypic peptide method recognizing specific peptides of the M-protein complementarity determining region [104]. Both techniques are more sensitive than conventional assays, but are still time consuming, costly, and may not be valid for all patients. The role of MS in the group of non-secretory MM patients, for instance, is still to be determined. An alternative method, Quantitative Immunoprecipitation MS (QIP-MS), uses polyclonal antibody-based technology to detect and quantify intact Ig in serum based on their molecular mass. Each M-protein is specific to each PC clone and differentiated from mAbs drugs applied in the clinical setting, allowing their tracing over time [105]. This method is currently being evaluated for MRD assessment in the context of clinical trials and has shown good concordance with BM NGF MRD results [106,107]. Comparison between BM NGS MRD and PB MS MRD revealed that all discordant cases were negative by the first technique, which could be explained with the presence of long half-life residual paraprotein. In fact, clearance of the monoclonal component can be a later occurrence and patients might not meet the criteria for CR despite achieving MRD negativity in the BM, thus not accurately reflecting the tumor burden [108]. In this regard, performing MRD evaluation in patients with a very good partial response (VGPR) according to the IMWG criteria to determine any deeper level of remission would be a more practical approach [109].
Routine use of liquid biopsy approaches for MRD assessment is still a long way ahead and requires further technical improvement, optimization, and clinical validation before it can replace BM MRD assessment. Ongoing studies regarding the concordance between PB and BM MRD assessment are essential to determine whether there is a true representation of disease status.
Liquid biopsies are certainly appealing as they allow a more frequent evaluation of disease burden and could play a complementary role to determine the precise timing for MRD monitoring, paving the way to a more personalized management of MM patients.

4. Clinical Relevance of MRD Evaluation in MM

Determining the response to treatment is crucial in monitoring MM patients. As data on MRD evaluation from different clinical trials becomes available, the evidence supporting its relevance in the management of MM is ever increasing. MRD testing has been in the spotlight for its role in comparing the efficacy of different treatment strategies and supporting clinical decisions towards an individualized approach to therapy in MM.

4.1. MRD Improve Definition of Response

In 2016, the IMWG revised response criteria introduced the definition of MRD in CR patients as the persistence or re-appearance of one myeloma cell in at least 105 normal cells [11]. Response to treatment is widely accepted as one of the most important indicators of survival, since, over the years, several studies have clearly demonstrated a correlation between prolonged survival and deeper responses. In 2008, Lahuerta et al., showed that the quality of response after ASCT, specifically CR, was significantly associated with longer event-free survival and OS in NDMM patients [7]. Similarly, Kapoor et al., showed that achieving stringent CR (sCR) after ASCT improved long-term outcome [110]. However, the conventional definition of CR was not able to distinguish between patients with different progression risks [111] and patients in CR and sCR still relapsed [6,7,110]. Moreover, conventional criteria of CR failed to identify patients at risk of early relapse. This was demonstrated by Paiva et al., that patients with unsustained CR within 1 year after ASCT, despite presenting similar CR rates at day 100 after high-dose therapy followed by ASCT, had a dismal outcome (median OS of 39 months; p < 0.001) [112]. It was the introduction of sensitive methodologies able to detect minimal levels of residual MM cells that allowed the improvement of CR definition. Previous studies using MFC with a cut-off of 10−4 already suggested that even lower levels of MRD would provide a more accurate outcome prediction [28]. Using NGS, Martinez-Lopez et al., were able to identify patients with high (<10−3), intermediate (10−3–10−5), and low (>10−5) levels of MRD with distinct TTP (27 versus 48 versus 80 months; p = 0.003 to 0.0001) [38].
Indeed, patients in CR can be further discriminated into those with detectable and undetectable MRD, with clear association of undetectable MRD with superior PFS and OS. In a study by the PETHEMA/GEM, it was already clear that CR patients with persistent MRD and patients in near CR/VGPR presented the same outcome in terms of both PFS (27 versus 29 months) and OS (59 versus 65 months). MRD negativity was associated with higher PFS (median 63 months; p < 0.001) and OS (median not reached; p < 0.001), surpassing CR in reducing risk of progression (PFS HR = 0.42 for MRD negativity versus HR = 0.67 for CR) and/or death (OS HR = 0.33, for MRD negativity versus HR = 0.58, for CR) [113]. Two meta-analyses demonstrated a positive correlation between MRD negativity assessed by immunophenotypic and molecular methods and prolonged PFS and OS. Munshi et al., showed a significant association of undetectable MRD with significantly better PFS overall (HR = 0.41; 95% CI 0.36–0.48; p < 0.001) and in CR patients (HR = 0.44; 95% CI 0.34–0.56; p < 0.001). OS was also favorable in MRD negative patients overall (HR = 0.57; 95% CI 0.46–0.71; p < 0.001) and in CR patients (HR = 0.47; 95% CI 0.33–0.67; p < 0.001) (HR of 0.44 for PFS and 0.47 for OS) [114]. Landgren et al., found that MRD negativity (versus positivity) was associated with better PFS (HR = 0.35; 95% CI 0.27–0.46; p < 0.001) and OS (HR = 0.48; 95% CI 0.33–0.70; p < 0.001) [115].
These results demonstrate that MRD redefined response criteria in MM and undoubtedly establish the importance of achieving undetectable MRD for patient outcome.

4.2. MRD Is a Relevant Prognostic Factor

Undetectable MRD is clinically relevant regardless of the patient risk group [116]. However, it is even more important in those high-risk patients associated with unfavorable cytogenetic alterations and biological characteristics according to the R-ISS. The IFM2009/EMN02 trials demonstrated that achievement of MRD negativity was less frequent in patients with high-risk cytogenetics, namely with del(17p). High-risk patients according to R-ISS and cytogenetics, with detectable MRD after treatment, had shorter PFS compared with MRD negative patients (7 versus 67 months for R-ISS Stage III patients, HR = 0.12, 95% CI 0.05–0.31; p < 0.001; 15 versus 53 months for patients with high-risk cytogenetics, HR = 0.18, 95% CI 0.09–0.35; p < 0.001) [27]. Perrot et al., also showed that patients with high-risk cytogenetics and undetectable MRD had longer PFS than those with standard-risk cytogenetics but detectable MRD. Furthermore, irrespective of the R-ISS disease stage at diagnosis, the median PFS was the same when MRD was undetectable, with comparable survival rates. [39]. The PETHEMA/GEM2010 trial demonstrated that MRD status was an independent prognostic factor for TTP (HR = 2.7; p = 0.007) and OS (HR = 3.1; p = 0.04), with significant positive impact of MRD negativity on TTP of high-risk cytogenetic patients (HR = 12.6; p = 0.01) [117]. Furthermore, the PETHEMA/GEM2012 trial revealed that the worse PFS and OS was attained in R-ISS Stage III patients with detectable MRD [26]. Munshi et al., recently published a large meta-analysis showing the importance of attaining MRD negative status in improving long-term survival regardless of disease stage, risk factors, treatment setting, and method and sensitivity thresholds for MRD measurement. MRD negativity improved PFS (HR = 0.33; 95% CI 0.29–0.37; p < 0.001) and OS (HR = 0.45; 95% CI 0.39–0.51; p < 0.001) compared with MRD positivity, especially when MRD negativity was reached 12 months post-maintenance rather than pre-maintenance [118]. A recent study by Goicoechea et al., showed 36-month PFS rates above 90% in patients with standard- and high-risk cytogenetic abnormalities when achieving undetectable MRD [116]. Li et al., also found no significant difference in PFS and OS between MRD negative high-risk patients and MRD positive standard-risk patients (median PFS 45 versus 34 months; p = 0.3; 4 year OS 100% versus 83.6%; p = 0.196) [119].
These findings highlight that achieving MRD negativity could overcome adverse risk factors identified at diagnosis, substantially altering patient prognosis during treatment.
So far, data from several clinical trials have shaped MRD evaluation to be performed in specific timepoints for established treatment protocols. That is the case of NDMM patients less than 70–75 years old, for which the standard intensification approach is still high-dose melphalan with ASCT [120]. In 2008, the study by the PETHEMA/GEM group in NDMM patients who underwent ASCT, MRD evaluation by MFC was performed at day 100 after ASCT, with longer PFS (median 71 versus 37 months; p < 0.001) and OS (median not reached versus 89 months; p < 0.002) in MRD negative patients [13]. Likewise, in the Myeloma IX study, MRD was determined both post-induction with RVd and post-ASCT, with higher PFS in MRD negative patients at both timepoints compared with those that only became MRD negative post-ASCT, howbeit with no benefit in OS [121]. In the Phase 2 study of the French group, including NDMM treated with RVd followed by ASCT and lenalidomide maintenance, MRD was measured by MFC after induction revealing that 68% of patients achieved MRD negativity with an estimated 3-year PFS of 77% (95% CI, 57% to 88%) and OS of 100% [122]. Improved PFS and OS with high-dose melphalan consolidation plus ASCT were obtained when compared with melphalan–prednisone–lenalidomide (median PFS 43 months versus 22.4 months, HR for progression or death = 0.44, 95% CI 0.32 to 0.61; p < 0.001; 4-year OS 81.6% versus 65.3%, HR for death = 0.55, 95% CI 0.32 to 0.93; p = 0.02) [123] and cyclophosphamide and dexamethasone plus lenalidomide (median 43.3 months (33.2–52.2) versus median 28.6 months (95% CI 20.6 to 36.7), HR for the first 24 months = 2.51, 95% CI 1.60 to 3.94; p < 0·0001) [124]. The EMN02/HO95 study revealed, at a median follow-up of 60.3 months, a significant improvement in median PFS with ASCT compared with bortezomib–melphalan–prednisolone (VMP) (56.7 months (49.3 to 64.5) versus 41.9 months (37–5 to 46.9), HR = 0.73, 95% CI 0.62 to 0.85; p = 0·0001) [125]. The results of the prospective multi-center PRIMeR study, that assessed MRD by MFC in the context of the STAMiNA trial, showed that MRD negative patients pre-maintenance and 1 year after ASCT had better PFS and longer OS, independent of the treatment arm. HR for progression or death in MRD negative versus MRD positive patients pre-maintenance and 1 year after ASCT were 0.48 and 0.22 (p < 0.001), and HR for overall mortality were 0.77 (p = 0.52) and 0.10 (p < 0.001), respectively [126].
The positive effect of attaining undetectable MRD even in NDMM patients ineligible for ASCT, particularly older and frail patients, is extremely relevant, as shown by the Spanish group [117]. Moreover, with the inclusion of daratumumab to the standard combination regiments in this group of patients, PFS and OS improved, and MRD negativity rates increased compared with standard therapy (27–24% versus 7%; p < 0.001) [1,3,42]. The MAIA Phase 3 trial comparing daratumumab–lenalidomide–dexamethasone (Dara-Rd) with lenalidomide–dexamethasone (Rd) [1], and the ALCYONE study comparing daratumumab–bortezomib–melphalan–prednisolone (Dara-VMP) with VMP [3] investigated outcomes using MRD negativity as a secondary endpoint, defined as the proportion of patients who were MRD negative at any timepoint using NGS with a sensitivity threshold of 10−5. The concept of sustained MRD negativity was established as the maintenance of MRD negative results in BM more than 6 or 12 months apart. With a median follow-up of 36.4 months in MAIA and 40.1 months in ALCYONE, patients with sustained MRD negativity for more than 6 months showed better PFS compared with patients who did not maintain MRD negativity, both separately and when results from the two studies were pooled. This was also observed with sustained MRD negativity for longer than 12 months. Improved PFS was seen in patients in CR and MRD negative compared with those who were MRD positive or who achieved VGPR or worse (HR = 0.19; 95% CI 0.14 to 0.26; p < 0.0001). Time to subsequent treatment was higher in patients who were MRD negative at any of the follow-up times analyzed versus MRD positive patients and in patients with sustained MRD negativity for more than 6 or 12 months versus those without sustained MRD negativity [127]. Even in transplant eligible patients, the addition of daratumumab to bortezomib–thalidomide–dexamethasone (Dara-VTd) in pre-transplant induction and post-transplant consolidation improved depth of response, with higher rates of MRD negativity (64% versus 44%; p < 0.0001; at 10–5 sensitivity by MFC). Achievement of undetectable MRD was associated with prolonged PFS and such improvement was consistent across patients’ baseline characteristics [5]. These studies demonstrate that regardless of treatment, MRD negativity is the major prognostic determinant, conferring a favorable prognosis.
A recent study by Ubieto et al., confirmed the prognostic relevance of MRD by showing that traditionally used parameters, such as myeloma cell count by morphology and serum-free light chain ratios, were irrelevant in patients with negative immunofixation. In addition, the current IMWG stratification had no prognostic value in transplant-eligible MM patients with persistent MRD [128]. The latest recommendations by the National Comprehensive Cancer Network include MRD assessment during follow-up as an indicator of prognosis [129]. Taken together, compelling data are being gathered regarding the importance of MRD negativity that may lead to the definition of new risk stratification tools.
Thus, attaining MRD negativity as a response to treatment, irrespective of the measurement technique or the therapy strategy, is indeed a robust predictor of PFS and OS.

5. MRD as a Surrogate Endpoint in Clinical Trials

With the continuous innovations introduced in MM treatment, the need to accelerate the drug approval process is increasingly important. Until now, PFS has been used as a surrogate endpoint for OS in clinical trials. However, as patients achieve prolonged responses, it is becoming a late endpoint, incompatible with the swiftness needed. In NDMM patients, achieving PFS is likely to take an average of seven years [130]. As the drug discovery landscape for MM changes, the introduction of validated surrogate endpoints instead of the traditional clinical trial endpoints is critically needed. MRD negativity is perhaps the strongest clinically significant prognostic factor associated with PFS, in both NDMM and RRMM patients, even more so than clinical staging or cytogenetics [40,115,131]. MRD is a specific measure of tumor burden, and knowledge regarding the role of MRD in the clinical course of MM has pushed its consideration as the best candidate to be used as a surrogate marker for drug approval. The use of MRD as a surrogate endpoint for PFS in MM is being evaluated and validated by the International Independent Team for Endpoint Approval of Myeloma MRD Initiative, a consortium that includes academic and pharmaceutical members [132]. Both the European Medicines Agency and the US Food and Drug Administration have issued draft guidelines [133,134]. MRD-guided therapy decisions require that key questions such as optimal sensitivity cut-off, definition of sustained MRD negativity, and best timing for MRD evaluation be specified in MM clinical trials. Very recently, recommendations on the harmonization in designing, performing, and interpreting MRD assessment in MM clinical trials were published to address these issues [25].
Treatment strategies based on MRD status should take into consideration the potential technical pitfalls that might hamper the correlation between MRD negativity and improvement in PFS, such as early evaluation after myeloablative conditioning treatments or sampling biases due to patchy infiltration. As previously mentioned, MM is characterized by spatial heterogeneity, and the random nature of BM aspirate sampling is a challenge for MRD analysis that could, in the future, be overcome through the inclusion of liquid biopsy approaches.

6. Open Questions That MRD Can Help Answer in the Clinical Setting

Advancements in the knowledge of residual disease in MM provide an important opportunity to determine how to better implement therapeutic changes in clinical practice. With the inclusion of increasingly effective therapies for MM, MRD represents an interesting means to guide treatment decisions.

6.1. Can MRD Redefine the Use of ASCT?

Thus far, current data suggest that attaining MRD negativity is more important than the actual means to achieve this profound depth of response. Consequently, the need for high-dose chemotherapy with ASCT is being questioned in a time when new and more effective treatments are being introduced. Results from the FORTE trial presented in 2019 showed similar MRD negativity rates, defined as <10−5 by MFC, in patients who underwent ASCT with four induction cycles and four consolidation cycles with carfilzomib-lenalidomide–dexamethasone (KRd) and patients who received 12 KRd cycles without ASCT (58% versus 54%) [29]. Responses assessed before maintenance were well matched between groups, showing that approximately 65% of patients were MRD negative. Results from a Phase 2 study evaluating weekly KRd in combination with daratumumab (Dara-KRd) showed that 74% of NDMM patients achieved MRD negativity, with no progressions at a median follow-up of 10 months [131]. A large randomized multi-center trial was initiated to compare Dara-KRd with standard care for NDMM patients (NCT03290950).
Attal et al., (NCT01191060) showed that MRD negativity was higher in the group of patients submitted to ASCT than in the non-ASCT group (79% versus 65%; p < 0.001) [135]. Using MRD data evaluated by NGS, Perrot et al., found lower levels of MRD negativity in patients treated only with RVd than in those who underwent ASCT (adjusted odds ratio for MRD negativity 1.65; 95% CI, 1.10–2.49; p = 0.02), but PFS was similar in both groups for patients with the same MRD status [39], raising the hypothesis that achieving MRD negativity after induction might inform decisions regarding the role of ASCT.
New quadruplet regimens are also being investigated in NDMM patients in the context of ASCT. In the CASSIOPEIA study (NCT02541383), the addition of daratumumab to VTd before and after ASCT was shown to improve depth of response and PFS or OS. Significant MRD negativity rates, at a 10−5 sensitivity threshold, were also detected in the Dara-VTd group (p < 0.0001), except in the high-risk patients [5]. In the GRIFFIN study (NCT02874742), MRD negativity after Dara-RVd was improved compared with RVd (51.0% versus 20.4%; p < 0.0001) [4]. A high rate of MRD negativity at a sensitivity of 10−5 by NGS was also achieved in the MASTER trial with Dara-KRd after consolidation, revealing that patients treated with this four-drug regimen without subsequent ASCT achieved similar MRD results as those with ASCT [136].
Thus, MRD status might be used to challenge the current paradigm regarding the role of high-dose chemotherapy and a transplant-based approach.

6.2. Can MRD Guide Maintenance Therapy?

The role of MRD in shaping maintenance treatment for MM is still an unanswered question. Two pooled analyses of two Phase 3 studies in patients receiving lenalidomide maintenance revealed an improvement in depth of MRD response, with 27% of MRD positive patients after consolidation becoming MRD negative during maintenance (sensitivity 10−4–10−5) [137,138]. In earlier trials, such as the Myeloma IX where MRD was assessed by MFC at a level of sensitivity of 10−4, the percentage of patients with durable MRD negativity was higher in the group that received maintenance therapy with thalidomide compared with those who did not receive any maintenance treatment (96% versus 68%; p = 0.026). In the subsequent Myeloma XI study, higher PFS was reported in MRD negative patients who received lenalidomide maintenance [139]. In a retrospective observational study, Alonso et al., reported that 34.3% of patients who were MRD positive after induction achieved MRD negativity during maintenance with lenalidomide. Patients who did not achieve MRD negativity, but showed declining MRD values during maintenance, still obtained a benefit in survival, with no difference in PFS between these patients and those who ultimately obtained MRD negativity [140]. In the abovementioned FORTE trial, significantly more patients who were MRD positive at maintenance randomization became MRD negative with carfilzomib and lenalidomide (KR) versus lenalidomide after a median follow up of 31 months and a median duration of maintenance of 27 months (46% versus 32%; p = 0.04) [29]. Other studies have also highlighted the beneficial effect of prolonging maintenance therapy in MRD positive patients [141,142]. Hahn et al., demonstrated that MRD negativity one year after ASCT was prognostic of better PFS and OS (p < 0.001) [126]. Long-lasting MRD negativity might have a good prognostic effect, but its optimal duration is still a matter of debate. This was explored by Gu et al., in a study of MRD evaluated in post-induction and during 3 until 24 months post-ASCT. A better PFS and a significantly longer OS was identified in patients that maintained MRD negativity since post-induction with no progression until 24 months post-ASCT compared with those with MRD negativity at post-induction but who became MRD positive 24 months post-ASCT (PFS not reached versus 15.4 ± 2.4 months; p = 0.000; OS not reached versus 35.2 ± 18.6 months; p = 0.000). Interestingly, patients who were MRD negative 24 months after ASCT regardless of their MRD status post-ASCT showed similar OS [143].
Prospective clinical trials with the inclusion of long-term serial MRD measurements would help determine the clinical significance of MRD evolution and lead toward a tailored approach to maintenance treatment.

6.3. Can MRD Be Used to Decide Treatment Discontinuation?

Repeated MRD monitoring can be used as a tool to confirm sustained MRD negativity and to identify subgroups of patients with different prognosis. The study by Gu et al., already suggested that the pattern of MRD evolution is more informative than determining MRD in a single time point and that sustained MRD negativity throughout treatment is associated with better prognosis [143]. Whether patients with sustained MRD negative status during maintenance can discontinue or de-escalate treatment is another open question.
In the recently published results of the MASTER trial, MRD status was applied to inform about the efficacy and duration of maintenance with Dara-KRd after ASCT. In total, 80% of patients reached MRD < 10−5 and 66% reached MRD < 10−6. A total of 34% of patients had MRD < 10−5 post-induction, 70% post-transplant, and 80% at best response. A total of 71% of patients with confirmed MRD negative remission (two consecutive MRD results) received no further consolidation therapy, and cumulative incidence of MRD reappearance or progression after 12 months of maintenance cessation was between 0% and 27% for patients with one, two, or more high-risk cytogenetic abnormalities [136]. Currently, clinical trials are ongoing to address whether treatment can be safely reduced or even discontinued taking into consideration a sustained MRD status, without aggravating patient prognosis. In the EMN17 Perseus trial (NCT03710603), patients with at least two MRD negative samples 12 months apart during maintenance with daratumumab–lenalidomide (Dara-R) can hold back daratumumab maintenance after at least two years and continue with lenalidomide only. In the SWOG 1803 DRAMMATIC trial (NCT04071457) comparing Dara-R to lenalidomide alone as maintenance after ASCT, patients who are MRD negative after 2 years of maintenance are randomized to treatment discontinuation vs. continuation. In the “Monoclonal Antibody-Based Sequential Therapy for Deep Remission in Multiple Myeloma (MASTER)” study (NCT03224507) in NDMM patients who are eligible for transplant, MRD negative patients by NGS (10−5) after induction therapy with Dara-KRd, followed by ASCT discontinue treatment, whereas patients who are MRD positive undergo further consolidation with Dara-KRd for up to two cycles until MRD negativity [136].
Results of these ongoing trials have the potential to avoid intensive therapy, preventing associated toxicity, and to change the current paradigm of treating patients until progression.

6.4. Can MRD Be Used to Intensify Treatment?

Another open issue is whether patients who are MRD positive would benefit from a treatment change or intensification with the purpose of achieving MRD negativity. In the IFM 2009 trial, no significant difference was observed in survival outcomes between patients who were MRD negative before maintenance and those who achieved MRD negativity during the first year of maintenance [39]. Response improvement over time under continuous therapy was observed in a retrospective study, where 34.3% of MRD positive patients after induction became MRD negative during maintenance [140]. The benefit of including new drugs in the therapeutic armamentarium against MM to attain deeper and more sustained responses is particularly important and is currently being examined in the AURIGA study (NCT03901963) with the association of daratumumab to lenalidomide during maintenance. The kinetics of MRD was also investigated in the GIMEMA trial, where patients receiving VTd after ASCT were monitored for MRD. This clinical trial revealed that the median duration of response was shorter in patients with MRD persistence (9 months); longer for those with MRD reappearance (38 months); and was not reached for patients in major MRD response (two consecutive MRD results < 10−4 by RQ-PCR) (p < 0.001). Moreover, results showed a median time lag between MRD reappearance and need for salvage treatment of 9 months [144]. Oliva et al., demonstrated that MRD progression, as measured by both MFC and ASO-qPCR every 6 months during maintenance, anticipated clinical relapse by a median of 9 months and biochemical relapse by a median of 4 months [145]. As reported in a randomized prospective study by Mina et al., early intervention with bortezomib and dexamethasone (Vd) induced disease stability or improvement in 82% of patients and delayed clinical progression by 11 months versus observation until clinical relapse (20.3 versus 9.5 months) [146].
Data regarding the approach to patients who lose MRD negativity is still scarce. The recently published study by Mohan et al., regarding the clinical implication of MRD loss revealed that MRD conversion occurred in 39% of the cohort of NDMM patients submitted to ASCT and at least 2 years of combined maintenance treatment at a median of 6.3 years. MRD conversion was associated with a higher risk of relapse, being able to predict clinical relapse in 70% of patients and preceding it by a median of 1 year (range 0-4.9). Noticeably, patients who converted from negative to positive MRD within the first 3 years after ASCT had lower PFS and OS compared with patients with sustained MRD negativity (PFS: HR = 4.5; 95% CI, 4.3–33.7; p < 0.0001; OS: HR = 5.7; 95% CI, 6.3–63.0; p < 0.0001) [147]. Treatment intervention upon signs of biochemical relapse rather than clinical symptoms can have a favorable impact on the outcome of these patients, as observed by the ENDEAVOR trial [148]. The hypothesis that outcome could be improved when considering an even lower level of disease, as determined by MRD assessment, is being addressed in the ongoing REMNANT trial (NCT04513639), where MRD negative patients after induction are randomized to start treatment if MRD becomes positive versus at the time of progressive disease [149].
These findings encourage the use of MRD as a marker to predict an imminent progression and to trigger an early therapeutic intervention to avoid the development of MM-related comorbidities.
The value of MRD in the RRMM setting is also being explored. Several studies have demonstrated an improved PFS in RRMM patients who achieve MRD negativity [2,150]. In the aforementioned meta-analysis by Munshi et al., MRD negativity improved PFS (HR = 0.34; 95% CI 0.24 to 0.47; p < 0.001) and OS (HR = 0.28; 95% CI 0.18 to 0.45; p < 0.001) in the group of relapsed heavily pre-treated patients [118]. The POLLUX [42] and CASTOR [151] trials assessed MRD by NGS (sensitivity threshold of 10−5) in patients at suspected CR, following confirmed CR (3 and 6 months in POLLUX; 6 and 12 months in CASTOR), and every 12 months after CR. With a median follow-up of 54.8 months in POLLUX and 50.2 months in CASTOR, the MRD negativity rate and the proportion of patients in CR achieving sustained MRD negativity for longer than 6 months were higher in daratumumab-based regimens compared with standard treatment (35.8 versus 9.2; p < 0.0001 for POLLUX; 36.1 versus 13; p = 0.404 for CASTOR). Similar results were observed for patients with sustained MRD negativity for longer than 12 months (28.4 versus 6.2; p = 0.0001 for POLLUX; 23.6 versus 0; p = 0.0098 for CASTOR). More than 80% of patients with sustained MRD negativity for more than 6 months had not progressed at 36 months [152]. The value of sustained MRD has been a matter of open discussion in recent years, and both POLLUX and CASTOR trials emphasized that the depth of response and the durability of MRD negativity is a favorable prognostic marker in RRMM patients.
In this particularly difficult-to-treat group of patients, how to approach the re-appearance of MRD is an additional unanswered clinical question.

7. MRD in the Context of New Therapies

Achieving deeper remissions is now frequently attainable. Since the first studies, new immunotherapeutic therapies such as chimeric antigen receptor T (CAR T) and T cell engagers have demonstrated the ability to achieve rapid MRD responses [153,154]. The emergence of new therapies against MM is particularly critical in the context of RRMM. Recent studies regarding mAbs and cellular products described exceptional rates of CR and MRD negativity among heavily pre-treated patients. Studies in the RRMM setting revealed a rate of MRD negativity between 5% and 30% of patients treated with an anti-CD38 mAb (daratumumab or isatuximab) in combination with IMiDs or PIs [155,156]. In the CARTITUDE-1 update presented at ASH 2021, with a median follow-up of 18 months, 91.8% MRD negativity was reported in patients with CR in a heavily pre-treated population after a single infusion of the anti-BCMA CAR T cell therapy ciltacabtagene–autoleucel (cilta-cel). Importantly, 44.3% of patients had sustained MRD negativity for ≥6 months and 18% for ≥12 months [157]. The Phase 2 CARTITUDE-2 study (NCT04133636) is currently ongoing and will evaluate the overall MRD negativity rate in various clinical settings. Additionally, in a first-in-human study of another anti-BCMA CAR T cell therapy LCAR-B38M, included in the LEGEND-2 study and identical to the construct used in the CARTITUDE-1 (NCT03548207) clinical study, CR was achieved in 74% of RRMM patients, with 92.8% of CR patients being MRD-negative with a PFS rate of 71% at 18 months [158]. In the KarMMa study, using a CAR T product targeting the BCMA idecabtagene–vicleucel (ide-cel, bb 2121), at a median follow-up of 13.3 months, the overall response rate (ORR) was 73% with 33% CR; 26% of the patients in CR and 39% of the patients in VGPR or better achieved MRD negativity by NGS (sensitivity < 10−5) [159]. Despite these impressive results, patients still relapse, with loss of BCMA expression as a potential underlying factor. A recent report by Da Via et al., provided an in-depth analysis of the molecular mechanisms behind alterations in the TNFRSF17 BCMA-encoding gene in a patient with irreversible loss of BCMA expression [160]. Results of the EVOLVE study using an investigational BCMA-directed CAR T cell product (Orva-cel) reported a median follow-up of 6.9 months with ORR of 92% and VGPR or better in 68% of the patients, with MRD negativity by NGS (sensitivity < 10−5) in 84% at 3 months. Of note, MRD assessment was performed in patients achieving partial response (PR) or better and not in CR [161]. A systemic review and meta-analysis of 30 studies using anti-BCMA CAR T cell in MM confirmed an estimated response rate of 78.3% (95% CI 72.4 to 84.3%) [162]. Based on the information provided, Bravo-Perez et al., estimated the pooled proportion of patients achieving MRD negativity to be 58.7% (95% CI 44.4 to 73.1%) in patients treated with CAR T cell constructs [163]. The first-in-human study of the anti-B-cell maturation antigen BiTE molecule AMG420 for RRMM patients revealed a response rate of 70%, including 50% MRD negative complete responses at the maximum tolerated dose [164].
In the study using a new combination of BCL-2 inhibitor venetoclax (Ven) associated with daratumumab (Dara) and dexamethasone (d) (Dara-Vd) with or without bortezomib (B) for RRMM, an increase in ORR and VGPR or better in the arm without bortezomib was reported (ORR of 92% and 96% with 79% and 96% VGPR or better in the Dara-Vd + B and Dara-Vd arms, respectively). Patients achieving CR/sCR who also reached MRD negativity were significantly higher in the arm without bortezomib [165].
As previously stated, deeper responses are associated with prolonged survival, and the new agents that are currently being evaluated in ongoing clinical trials convey a new hope to obtain sustained MRD negativity.

8. Sustained MRD Negative Response: Role of the Immune System

One of the aspects that should be considered when considering the meaning of durable MRD negativity is the role of the immune microenvironment and the patient immunological profile. The equilibrium between the BM microenvironment and a residual MM clone that remains stable with no clinical implication is an interesting concept in the field of MM. The latest data on the cellular patterns within the BM microenvironment of MM patients, particularly those that maintain long-term responses, revealed features of active immune surveillance [166]. A deeper understanding of the interaction between MM cells and the immune system will eventually allow us to define specific immune profiles associated with a stable disease state versus an early relapse state.
Immune suppression is responsible for increasing the risk of infection in MM patients and it has also been associated with MM relapse and progression. Data are emerging to suggest an association of specific immune profiles with MRD status and outcome. In ASCT ineligible patients, immune profile concomitant with MRD assessment was able to identify three groups with distinct outcomes (OS at 3 years: poor 25%, intermediate 61%, and favorable 100%; p = 0.01). An increase in mature B cells was observed in patients with a more favorable outcome [117]. Ho et al., recently showed that higher peripheral CD19+ B cell counts before ASCT was correlated with better 2-year PFS (83% (highest quartile) vs 53% (lowest quartile); p = 0.01) and OS (93% (highest quartile) vs 63% (lowest quartile); p = 0.0003), particularly in MRD positive patients who were more impacted by the level of B cells, implying a beneficial effect from an improved immune profile. At day 100 after ASCT, higher peripheral γδ and CD4+ central memory T cell counts correlated with improved 2-year OS (89% (highest quartile) vs 65% (lowest quartile); p = 0.01 and 95% (upper quartile) vs 47% (lowest quartile); p = 0.0003, respectively), only in MRD negative patients, suggesting that an improved immune profile is not sufficient to control higher tumor loads [167]. A study by Bhutani et al., addressing the role of the immune system in achieving MRD negativity, revealed distinct circulating NK, NK-T, and T cell phenotypes according to MRD status [168]. Papadimitriou et al., identified a specific peripheral blood immune profile in MRD positive patients with lower effector/effector memory CD4+ T cell and higher naïve CD4+ T cell subsets. These specific immune signatures were informative and predictive of BM MRD status (Area Under the Curve (AUC) = 0.8) [169].
Previous studies have addressed the immune profile in patients achieving long-term disease control. These patients seem to revert to a MGUS-like profile, showing a lower expression of regulatory T cells in the BM and recovery of BM and circulating B cell counts. Interestingly, patients who did not reach CR after HDT/ASCT but achieved a MGUS-like profile presented similar TTP (p = 0.81) and OS (p = 0.24) than those who reached CR [170,171]. Recently presented data on the cellular and immune microenvironment of MM patients treated with lenalidomide maintenance therapy, revealed that patients who reached sustained MRD negativity showed a normalization of the immune microenvironment. However, an immunosuppressive landscape was present before and remained after 1 year of maintenance therapy in patients with unsustainable MRD negativity [172]. The superior outcome of an MGUS-like profile was already confirmed by GEP70 [173,174]. Recently, MRD conversion from negative to positive was associated with different GEP subtypes, with short-time conversion (less than 5 years from diagnosis) being mostly HY subtype and longer-time conversion (10 years from diagnosis) by LB and CD-2 subtypes [147]. In fact, subtype CD-2, despite having lower rates of CR and MRD negativity, showed a comparable clinical outcome to the superior performing CD-1 subtype [175]. In such cases, clones remaining after therapy may not be able to drive disease relapse, being composed of immature clonotypic cells without most somatic mutations and copy number variations found in MM cells [166]. Thus, despite failure to achieve CR and MRD negative response, there is a subgroup of patients who might remain in stable disease without further treatment or compromise in outcome.
Incorporating immune profiling in clinical trials as an exploratory endpoint, along with MRD to assess depth of response, will help to accurately determine prognosis and ultimately help guide the management of MM patients.

9. Ideal Threshold for MRD: How Deep Should We Go?

Continuous advancement of the treatment options for MM has dramatically increased patient outcome. The sensitivity limit of the current available techniques is, in fact, determinant, as MRD-negative strictly means that disease is undetectable below a certain level. Therefore, “minimal” is now replaced with “measurable” as there is no guarantee that the disease was eliminated. Along with improvements in the techniques to evaluate MRD, we are now closer to achieving an unprecedented level of response. What is the depth for MRD detection needed to enhance patient management?
One simple answer could be that the assay with the highest sensitivity should be considered as standard. As previously stated, several studies using both NGF and NGS showed an association between the level of sensitivity and the rate of PFS, clearly defining subsets of patients with different risks of relapse [26,38,176].
There is now ample consensus regarding MRD as the most powerful prognostic tool in MM, clearly surpassing the traditional definition of CR. The recent large meta-analysis by Munshi et al., showed that undetectable MRD is associated with favorable outcomes in various disease and treatment settings [118]. These data confirm the advantage of using MRD as a surrogate for PFS and OS, setting the stage to use this knowledge for determining treatment strategies.
Currently, MRD-guided treatment decisions are taken in other hematological diseases such as acute lymphoblastic leukemia and chronic myeloid leukemia [177,178]. Thus far, this is not the case in MM, where treatment strategies are not adapted according to MRD results. An increasing number of clinical trials currently being opened include MRD negativity as a primary endpoint or use MRD assessment to guide treatment decisions, as summarized in Table 2. Results from these clinical trials will eventually lead to changes in this treatment paradigm.

10. Conclusions

The results arising from multiple clinical trials have shown that a deeper and durable response is associated with prolonged survival and that undetectable MRD has a prognostic impact in NDMM and RRMM patients. However, MRD assessment, despite providing valuable information with high accuracy regarding the quality of response, is yet to attain the deserved spotlight in clinical practice. With the development of more innovative and effective therapies for MM, and the simultaneous efforts to standardize testing and reporting of MRD data, results of prospective, ongoing, and future MRD-based clinical trials will help to overcome the apparent reluctancy in implementing MRD to drive treatment decisions. We anticipate that MRD will prove to have a pivotal role in correctly defining and ultimately improving MM patient outcome.
The impact of MRD should also take into consideration the new therapy regimens available for both NDMM and RRMM patients. It is expectable that different therapies, with diverse mechanisms of actions and applied in different disease settings, could require distinct levels of MRD for optimal prognosis and treatment decisions. Supported by results from clinical trials, clinical practice can take advantage of the knowledge apported by sustained MRD to define monitoring guidelines and to drive treatment decisions. We believe that establishing MRD monitoring as a surrogate for OS and a biomarker to assess therapy efficacy, combined with baseline risk factors, will certainly help to establish an individualized and tailored approach for MM treatment.

Author Contributions

Conceptualization, J.C. and C.J.; writing—original draft preparation, J.C.; writing—review and editing, J.C., F.B., P.L. and C.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Facon, T.; Kumar, S.; Plesner, T.; Orlowski, R.Z.; Moreau, P.; Bahlis, N.; Basu, S.; Nahi, H.; Hulin, C.; Quach, H.; et al. Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. N. Engl. J. Med. 2019, 380, 2104–2115. [Google Scholar] [CrossRef] [PubMed]
  2. Spencer, A.; Lentzsch, S.; Weisel, K.; Avet-Loiseau, H.; Mark, T.M.; Spicka, I.; Masszi, T.; Lauri, B.; Levin, M.-D.; Bosi, A.; et al. Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: Updated analysis of CASTOR. Haematologica 2018, 103, 2079–2087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mateos, M.-V.; Cavo, M.; Blade, J.; Dimopoulos, M.A.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; et al. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): A randomised, open-label, phase 3 trial. Lancet 2020, 395, 132–141. [Google Scholar] [CrossRef]
  4. Voorhees, P.M.; Kaufman, J.L.; Laubach, J.; Sborov, D.W.; Reeves, B.; Rodriguez, C.; Chari, A.; Silbermann, R.; Costa, L.J.; Anderson, L.D.; et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: The GRIFFIN trial. Blood 2020, 136, 936–945. [Google Scholar] [CrossRef]
  5. Moreau, P.; Attal, M.; Hulin, C.; Arnulf, B.; Belhadj, K.; Benboubker, L.; Béné, M.C.; Broijl, A.; Caillon, H.; Caillot, D.; 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 2019, 394, 29–38. [Google Scholar] [CrossRef]
  6. Gay, F.; Larocca, A.; Wijermans, P.; Cavallo, F.; Rossi, D.; Schaafsma, R.; Genuardi, M.; Romano, A.; Liberati, A.M.; Siniscalchi, A.; 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, 3025–3031. [Google Scholar] [CrossRef]
  7. Lahuerta, J.J.; Mateos, M.V.; Martínez-López, J.; Rosiñol, L.; Sureda, A.; de la Rubia, J.; García-Laraña, J.; Martínez-Martínez, R.; Hernández-García, M.T.; Carrera, D.; et al. Influence of pre- and post-transplantation responses on outcome of patients with multiple myeloma: Sequential improvement of response and achievement of complete response are associated with longer survival. J. Clin. Oncol. 2008, 26, 5775–5782. [Google Scholar] [CrossRef]
  8. García-Ortiz, A.; Rodríguez-García, Y.; Encinas, J.; Maroto-Martín, E.; Castellano, E.; Teixidó, J.; Martínez-López, J. The Role of Tumor Microenvironment in Multiple Myeloma Development and Progression. Cancers 2021, 13, 217. [Google Scholar] [CrossRef]
  9. Lopes, R.; Caetano, J.; Ferreira, B.; Barahona, F.; Carneiro, E.A.; João, C. The Immune Microenvironment in Multiple Myeloma: Friend or Foe? Cancers 2021, 13, 625. [Google Scholar] [CrossRef]
  10. Ho, M.; Goh, C.Y.; Patel, A.; Staunton, S.; O’Connor, R.; Godeau, M.; Bianchi, G. Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. Clin. Lymphoma. Myeloma Leuk. 2020, 20, e752–e768. [Google Scholar] [CrossRef]
  11. Kumar, S.; Paiva, B.; Anderson, K.C.; Durie, B.; Landgren, O.; Moreau, P.; Munshi, N.; Lonial, S.; Bladé, J.; Mateos, M.-V.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. [Google Scholar] [CrossRef]
  12. San Miguel, J.F.; Almeida, J.; Mateo, G.; Bladé, J.; López-Berges, C.; Caballero, D.; Hernández, J.; Moro, M.J.; Fernández-Calvo, J.; Díaz-Mediavilla, J.; et al. Immunophenotypic evaluation of the plasma cell compartment in multiple myeloma: A tool for comparing the efficacy of different treatment strategies and predicting outcome. Blood 2002, 99, 1853–1856. [Google Scholar] [CrossRef]
  13. Paiva, B.; Vidriales, M.B.; Cerveró, J.; Mateo, G.; Pérez, J.J.; Montalbán, M.A.; Sureda, A.; Montejano, L.; Gutiérrez, N.C.; De Coca, A.G.; et al. Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation. Blood 2008, 112, 4017–4023. [Google Scholar] [CrossRef] [Green Version]
  14. Rawstron, A.C.; Davies, F.E.; DasGupta, R.; Ashcroft, A.J.; Patmore, R.; Drayson, M.T.; Owen, R.G.; Jack, A.S.; Child, J.A.; Morgan, G.J. Flow cytometric disease monitoring in multiple myeloma: The relationship between normal and neoplastic plasma cells predicts outcome after transplantation. Blood 2002, 100, 3095–3100. [Google Scholar] [CrossRef] [Green Version]
  15. Avet-Loiseau, H.; Corre, J.; Lauwers-Cances, V.; Chretien, M.-L.; Robillard, N.; Leleu, X.; Hulin, C.; Gentil, C.; Arnulf, B.; Belhadj, K.; 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, 191. [Google Scholar] [CrossRef]
  16. Flores-Montero, J.; Sanoja-Flores, L.; Paiva, B.; Puig, N.; García-Sánchez, O.; Böttcher, S.; van der Velden, V.H.J.; Pérez-Morán, J.-J.; Vidriales, M.-B.; García-Sanz, R.; et al. Next Generation Flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia 2017, 31, 2094–2103. [Google Scholar] [CrossRef] [Green Version]
  17. Stetler-Stevenson, M.; Paiva, B.; Stoolman, L.; Lin, P.; Jorgensen, J.L.; Orfao, A.; Van Dongen, J.; Rawstron, A.C. Consensus guidelines for myeloma minimal residual disease sample staining and data acquisition. Cytom. Part B-Clin. Cytom. 2016, 90, 26–30. [Google Scholar] [CrossRef]
  18. Costa, E.S.; Pedreira, C.E.; Barrena, S.; Lecrevisse, Q.; Flores, J.; Quijano, S.; Almeida, J.; del Carmen García-Macias, M.; Bottcher, S.; Van Dongen, J.J.M.M.; et al. Automated pattern-guided principal component analysis vs expert-based immunophenotypic classification of B-cell chronic lymphoproliferative disorders: A step forward in the standardization of clinical immunophenotyping. Leukemia 2010, 24, 1927–1933. [Google Scholar] [CrossRef] [Green Version]
  19. Roshal, M.; Flores-Montero, J.A.; Gao, Q.; Koeber, M.; Wardrope, J.; Durie, B.G.M.; Dogan, A.; Orfao, A.; Landgren, O. MRD detection in multiple myeloma: Comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Adv. 2017, 1, 728–732. [Google Scholar] [CrossRef] [Green Version]
  20. Bene, M.C.; Robillard, N.; Moreau, P.; Wuilleme, S. Comparison of the Performance of Surface Alone or Surface Plus Cytoplasmic Approaches for the Assessment of Minimal Residual Disease in Multiparameter Flow Cytometry in Multiple Myeloma. Blood 2019, 134, 1799. [Google Scholar] [CrossRef]
  21. Dold, S.M.; Riebl, V.; Wider, D.; Follo, M.; Pantic, M.; Ihorst, G.; Duyster, J.; Zeiser, R.; Wäsch, R.; Engelhardt, M. Validated single-tube multiparameter flow cytometry approach for the assessment of minimal residual disease in multiple myeloma. Haematologica 2020, 105, e523. [Google Scholar] [CrossRef] [Green Version]
  22. Van Dongen, J.J.M.; Lhermitte, L.; Böttcher, S.; Almeida, J.; van der Velden, V.H.J.; Flores-Montero, J.; Rawstron, A.; Asnafi, V.; Lécrevisse, Q.; Lucio, P.; et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia 2012, 26, 1908–1975. [Google Scholar] [CrossRef] [Green Version]
  23. Paíno, T.; Paiva, B.; Sayagués, J.M.; Mota, I.; Carvalheiro, T.; Corchete, L.A.; Aires-Mejía, I.; Pérez, J.J.; Sanchez, M.L.; Barcena, P.; et al. Phenotypic identification of subclones in multiple myeloma with different chemoresistant, cytogenetic and clonogenic potential. Leukemia 2015, 29, 1186–1194. [Google Scholar] [CrossRef] [Green Version]
  24. Zannetti, B.A.; Faini, A.C.; Massari, E.; Geuna, M.; Maffini, E.; Poletti, G.; Cerchione, C.; Martinelli, G.; Malavasi, F.; Lanza, F. Novel Insights in Anti-CD38 Therapy Based on CD38-Receptor Expression and Function: The Multiple Myeloma Model. Cells 2020, 9, 2666. [Google Scholar] [CrossRef]
  25. 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. [Google Scholar] [CrossRef]
  26. Paiva, B.; Puig, N.; Cedena, M.T.; Rosiñol, L.; Cordón, L.; Vidriales, M.B.; Burgos, L.; Flores-Montero, J.; Sanoja-Flores, L.; Lopez-Anglada, L.; et al. Measurable residual disease by next-generation flow cytometry in multiple myeloma. J. Clin. Oncol. 2020, 38, 784–792. [Google Scholar] [CrossRef]
  27. Oliva, S.; Hofste Op Bruinink, D.; Rihova, L.; D’agostino, M.; Pantani, L.; Capra, A.; Van Der Holt, B.; Troia, R.; Petrucci, M.T.; Villanova, T.; et al. Minimal residual disease assessment by multiparameter flow cytometry in transplant-eligible myeloma in the EMN02/HOVON 95 MM trial. Blood Cancer J. 2021, 11, 106. [Google Scholar] [CrossRef]
  28. Rawstron, A.C.; Gregory, W.M.; De Tute, R.M.; Davies, F.E.; Bell, S.E.; Drayson, M.T.; Cook, G.; Jackson, G.H.; Morgan, G.J.; Child, J.A.; et al. Minimal residual disease in myeloma by flow cytometry: Independent prediction of survival benefit per log reduction. Blood 2015, 125, 1932–1935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Gay, F.; Musto, P.; Rota-Scalabrini, D.; Bertamini, L.; Belotti, A.; Galli, M.; Offidani, M.; Zamagni, E.; Ledda, A.; Grasso, M.; et al. Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): A randomised, open-label, phase 2 trial. Lancet Oncol. 2021, 22, 1705–1720. [Google Scholar] [CrossRef]
  30. Jasielec, J.K.; Kubicki, T.; Raje, N.; Vij, R.; Reece, D.; Berdeja, J.; Derman, B.A.; Rosenbaum, C.A.; Richardson, P.; Gurbuxani, S.; et al. Carfilzomib, lenalidomide, and dexamethasone plus transplant in newly diagnosed multiple myeloma. Blood 2020, 136, 2513–2523. [Google Scholar] [CrossRef] [PubMed]
  31. Avet-Loiseau, H.A. Minimal Residual Disease by Next-Generation Sequencing: Pros and Cons. Am. Soc. Clin. Oncol. Educ. Book. 2016, 36, e425–e430. [Google Scholar] [CrossRef]
  32. Udd, K.A.; Spektor, T.M.; Berenson, J.R. Monitoring multiple myeloma. Clin. Adv. Hematol. Oncol. 2017, 15, 951–961. [Google Scholar]
  33. Gozzetti, A.; Nieto, Y.; Ramanathan, M.; Terragna, C.; Romano, A.; Palumbo, G.A.; Parrinello, N.L.; Conticello, C.; Martello, M.; Terragna, C. Minimal Residual Disease Assessment Within the Bone Marrow of Multiple Myeloma: A Review of Caveats, Clinical Significance and Future Perspectives. Front. Oncol. 2019, 9, 699. [Google Scholar] [CrossRef]
  34. Martinez-Lopez, J.; Blade, J.; Mateos, M.-V.; Grande, C.; Alegre, A.; García-Laraña, J.; Sureda, A.; de la Rubia, J.; Conde, E.; Martinez, R.; et al. Long-term prognostic significance of response in multiple myeloma after stem cell transplantation. Blood 2011, 118, 529–534. [Google Scholar] [CrossRef]
  35. Pulsipher, M.A.; Carlson, C.; Langholz, B.; Wall, D.A.; Schultz, K.R.; Bunin, N.; Kirsch, I.; Gastier-Foster, J.M.; Borowitz, M.; Desmarais, C.; et al. IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients. Blood 2015, 125, 3501–3508. [Google Scholar] [CrossRef] [Green Version]
  36. Pott, C.; Hoster, E.; Delfau-Larue, M.-H.; Beldjord, K.; Böttcher, S.; Asnafi, V.; Plonquet, A.; Siebert, R.; Callet-Bauchu, E.; Andersen, N.; et al. Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: A European MCL intergroup study. Blood 2010, 115, 3215–3223. [Google Scholar] [CrossRef]
  37. Bai, Y.; Orfao, A.; Chim, C.S. Molecular detection of minimal residual disease in multiple myeloma. Br. J. Haematol. 2018, 181, 11–26. [Google Scholar] [CrossRef] [Green Version]
  38. Martinez-Lopez, J.; Lahuerta, J.J.; Pepin, F.; González, M.; Barrio, S.; Ayala, R.; Puig, N.; Montalban, M.A.; Paiva, B.; Weng, L.; et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood 2014, 123, 3073–3079. [Google Scholar] [CrossRef]
  39. Perrot, A.; Lauwers-Cances, V.; Corre, J.; Robillard, N.; Hulin, C.; Chretien, M.L.; Dejoie, T.; Maheo, S.; Stoppa, A.M.; Pegourie, B.; et al. Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma. Blood 2018, 132, 2456–2464. [Google Scholar] [CrossRef] [Green Version]
  40. 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. [Google Scholar] [CrossRef]
  41. Mateos, M.-V.; Dimopoulos, M.A.; Cavo, M.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; Kaplan, P.; et al. Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma. N. Engl. J. Med. 2018, 378, 518–528. [Google Scholar] [CrossRef]
  42. Dimopoulos, M.A.; Oriol, A.; Nahi, H.; San-Miguel, J.; Bahlis, N.J.; Usmani, S.Z.; Rabin, N.; Orlowski, R.Z.; Komarnicki, M.; Suzuki, K.; et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2016, 375, 1319–1331. [Google Scholar] [CrossRef] [Green Version]
  43. Landgren, O.; Rustad, E.H. Meeting report: Advances in minimal residual disease testing in multiple myeloma 2018. Adv. Cell Gene Ther. 2019, 2, e26. [Google Scholar] [CrossRef] [Green Version]
  44. Takamatsu, H. Comparison of minimal residual disease detection by multiparameter flow cytometry, ASO-qPCR, droplet digital PCR, and deep sequencing in patients with multiple myeloma who underwent autologous stem cell transplantation. J. Clin. Med. 2017, 6, 91. [Google Scholar] [CrossRef] [Green Version]
  45. Ladetto, M.; Brüggemann, M.; Monitillo, L.; Ferrero, S.; Pepin, F.; Drandi, D.; Barbero, D.; Palumbo, A.; Passera, R.; Boccadoro, M.; et al. Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia 2014, 28, 1299–1307. [Google Scholar] [CrossRef]
  46. Paiva, B.; Puig, N.; García-Sanz, R.; San Miguel, J.F. Is this the time to introduce minimal residual disease in multiple myeloma clinical practice? Clin. Cancer Res. 2015, 21, 2001–2008. [Google Scholar] [CrossRef] [Green Version]
  47. Biran, N.; Ely, S.; Chari, A. Controversies in the Assessment of Minimal Residual Disease in Multiple Myeloma: Clinical Significance of Minimal Residual Disease Negativity Using Highly Sensitive Techniques. Curr. Hematol. Malig. Rep. 2014, 9, 368–378. [Google Scholar] [CrossRef]
  48. Gargis, A.S.; Kalman, L.; Berry, M.W.; Bick, D.P.; Dimmock, D.P.; Hambuch, T.; Lu, F.; Lyon, E.; Voelkerding, K.V.; Zehnbauer, B.A.; et al. Assuring the quality of next-generation sequencing in clinical laboratory practice. Nat. Biotechnol. 2012, 30, 1033–1036. [Google Scholar] [CrossRef]
  49. Brüggemann, M.; Kotrová, M.; Knecht, H.; Bartram, J.; Boudjogrha, M.; Bystry, V.; Fazio, G.; Froňková, E.; Giraud, M.; Grioni, A.; 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, 2241–2253. [Google Scholar] [CrossRef] [Green Version]
  50. Oliva, S.; Genuardi, E.; Belotti, A.; Frascione, P.M.M.; Galli, M.; Capra, A.; Offidani, M.; Vozella, F.; Zambello, R.; Auclair, D.; et al. 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. 2020, 38, 8533. [Google Scholar] [CrossRef]
  51. Martinez-Lopez, J.; Sanchez-Vega, B.; Barrio, S.; Cuenca, I.; Ruiz-Heredia, Y.; Alonso, R.; Rapado, I.; Marin, C.; Cedena, M.-T.; Paiva, B.; 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, 1446–1449. [Google Scholar] [CrossRef] [PubMed]
  52. Avet-Loiseau, H.; Bene, M.C.; Wuilleme, S.; Corre, J.; Attal, M.; Arnulf, B.; Garderet, L.; Macro, M.; Stoppa, A.-M.; Delforge, M.; 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–e4. [Google Scholar] [CrossRef]
  53. Lee, N.; Moon, S.Y.; Lee, J.-H.; Park, H.-K.; Kong, S.-Y.; Bang, S.-M.; Lee, J.H.; Yoon, S.-S.; Lee, D.S. Discrepancies between the percentage of plasma cells in bone marrow aspiration and BM biopsy: Impact on the revised IMWG diagnostic criteria of multiple myeloma. Blood Cancer J. 2017, 7, e530. [Google Scholar] [CrossRef] [PubMed]
  54. Rasche, L.; Chavan, S.S.; Stephens, O.W.; Patel, P.H.; Tytarenko, R.; Ashby, C.; Bauer, M.; Stein, C.; Deshpande, S.; Wardell, C.; et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat. Commun. 2017, 8, 268. [Google Scholar] [CrossRef]
  55. Hillengass, J.; Landgren, O. Challenges and opportunities of novel imaging techniques in monoclonal plasma cell disorders: Imaging “early myeloma”. Leuk. Lymphoma 2013, 54, 1355–1363. [Google Scholar] [CrossRef]
  56. Lu, Y.-Y.; Chen, J.-H.; Lin, W.-Y.; Liang, J.-A.; Wang, H.-Y.; Tsai, S.-C.; Kao, C.-H. FDG PET or PET/CT for detecting intramedullary and extramedullary lesions in multiple Myeloma: A systematic review and meta-analysis. Clin. Nucl. Med. 2012, 37, 833–837. [Google Scholar] [CrossRef]
  57. 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. [Google Scholar] [CrossRef]
  58. Cavo, M.; Terpos, E.; Nanni, C.; Moreau, P.; Lentzsch, S.; Zweegman, S.; Hillengass, J.; Engelhardt, M.; Usmani, S.Z.; Vesole, D.H.; 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–e217. [Google Scholar] [CrossRef]
  59. Moreau, P. PET-CT in MM: A new definition of CR. Blood 2011, 118, 5984–5985. [Google Scholar] [CrossRef] [Green Version]
  60. Bladé, J.; Fernández de Larrea, C.; Rosiñol, L.; Cibeira, M.T.; Jiménez, R.; Powles, R. Soft-tissue plasmacytomas in multiple myeloma: Incidence, mechanisms of extramedullary spread, and treatment approach. J. Clin. Oncol. 2011, 29, 3805–3812. [Google Scholar] [CrossRef]
  61. Zamagni, E.; Nanni, C.; Patriarca, F.; Englaro, E.; Castellucci, P.; Geatti, O.; Tosi, P.; Tacchetti, P.; Cangini, D.; Perrone, G.; et al. A prospective comparison of 18F-fluorodeoxyglucose positron emission tomography-computed tomography, magnetic resonance imaging and whole-body planar radiographs in the assessment of bone disease in newly diagnosed multiple myeloma. Haematologica 2007, 92, 50–55. [Google Scholar] [CrossRef]
  62. 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 [(18)F]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. [Google Scholar] [CrossRef]
  63. Zamagni, E.; Nanni, C.; Mancuso, K.; Tacchetti, P.; Pezzi, A.; Pantani, L.; Zannetti, B.; Rambaldi, I.; Brioli, A.; Rocchi, S.; et al. PET/CT Improves the Definition of Complete Response and Allows to Detect Otherwise Unidentifiable Skeletal Progression in Multiple Myeloma. Clin. Cancer Res. 2015, 21, 4384–4390. [Google Scholar] [CrossRef] [Green Version]
  64. Zamagni, E.; Cavo, M. The role of imaging techniques in the management of multiple myeloma. Br. J. Haematol. 2012, 159, 499–513. [Google Scholar] [CrossRef]
  65. Sachpekidis, C.; Mosebach, J.; Freitag, M.T.; Wilhelm, T.; Mai, E.K.; Goldschmidt, H.; Haberkorn, U.; Schlemmer, H.-P.; Delorme, S.; Dimitrakopoulou-Strauss, A. 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, 479–492. [Google Scholar]
  66. Pawlyn, C.; Fowkes, L.; Otero, S.; Jones, J.R.; Boyd, K.D.; Davies, F.E.; Morgan, G.J.; Collins, D.J.; Sharma, B.; Riddell, A.; et al. Whole-body diffusion-weighted MRI: A new gold standard for assessing disease burden in patients with multiple myeloma? Leukemia 2016, 30, 1446–1448. [Google Scholar] [CrossRef] [Green Version]
  67. Morales-Lozano, M.I.; Viering, O.; Samnick, S.; Rodriguez-Otero, P.; Buck, A.K.; Marcos-Jubilar, M.; Rasche, L.; Prieto, E.; Kortüm, K.M.; San-Miguel, J.; et al. 18F-FDG and 11C-Methionine PET/CT in Newly Diagnosed Multiple Myeloma Patients: Comparison of Volume-Based PET Biomarkers. Cancers 2020, 12, 1042. [Google Scholar] [CrossRef] [Green Version]
  68. Rasche, L.; Angtuaco, E.; McDonald, J.E.; Buros, A.; Stein, C.; Pawlyn, C.; Thanendrarajan, S.; Schinke, C.; Samant, R.; Yaccoby, S.; et al. Low expression of hexokinase-2 is associated with false-negative FDG-positron emission tomography in multiple myeloma. Blood 2017, 130, 30–34. [Google Scholar] [CrossRef] [Green Version]
  69. 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. [Google Scholar] [CrossRef]
  70. Ulaner, G.A.; Sobol, N.B.; O’Donoghue, J.A.; Kirov, A.S.; Riedl, C.C.; Min, R.; Smith, E.; Carter, L.M.; Lyashchenko, S.K.; Lewis, J.S.; et al. CD38-targeted Immuno-PET of Multiple Myeloma: From Xenograft Models to First-in-Human Imaging. Radiology 2020, 295, 606–615. [Google Scholar] [CrossRef]
  71. Moreau, P.; Zweegman, S.; Perrot, A.; Hulin, C.; Caillot, D.; Facon, T.; Leleu, X.; Belhadj, K.; Karlin, L.; Benboubker, L.; 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, 692. [Google Scholar] [CrossRef]
  72. 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. [Google Scholar] [CrossRef]
  73. Zamagni, E.; Patriarca, F.; Nanni, C.; Zannetti, B.; Englaro, E.; Pezzi, A.; Tacchetti, P.; Buttignol, S.; Perrone, G.; Brioli, A.; et al. Prognostic relevance of 18-F FDG PET/CT in newly diagnosed multiple myeloma patients treated with up-front autologous transplantation. Blood 2011, 118, 5989–5995. [Google Scholar] [CrossRef] [Green Version]
  74. John, L.; Poos, A.; Tirier, S.M.; Mallm, J.-P.; Prokoph, N.; Brobeil, A.; Lutz, R.; Schumacher, S.; Steiger, S.; Bauer, K.; et al. The Spatial Heterogeneity in Newly Diagnosed Multiple Myeloma Patients—From Sub-Clonal Architecture to the Immune Microenvironment. Blood 2021, 138, 729. [Google Scholar] [CrossRef]
  75. Thiele, J.-A.; Pitule, P.; Hicks, J.; Kuhn, P. Single-Cell Analysis of Circulating Tumor Cells. Methods Mol. Biol. 2019, 1908, 243–264. [Google Scholar] [CrossRef]
  76. Mishima, Y.; Paiva, B.; Shi, J.; Park, J.; Manier, S.; Takagi, S.; Massoud, M.; Perilla-Glen, A.; Aljawai, Y.; Huynh, D.; et al. The Mutational Landscape of Circulating Tumor Cells in Multiple Myeloma. Cell Rep. 2017, 19, 218–224. [Google Scholar] [CrossRef] [Green Version]
  77. Lohr, J.G.; Kim, S.; Gould, J.; Knoechel, B.; Drier, Y.; Cotton, M.J.; Gray, D.; Birrer, N.; Wong, B.; Ha, G.; et al. Genetic interrogation of circulating multiple myeloma cells at single-cell resolution. Sci. Transl. Med. 2016, 8, 363ra147. [Google Scholar] [CrossRef] [Green Version]
  78. Manier, S.; Park, J.; Capelletti, M.; Bustoros, M.; Freeman, S.S.; Ha, G.; Rhoades, J.; Liu, C.J.; Huynh, D.; Reed, S.C.; et al. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma. Nat. Commun. 2018, 9, 1691. [Google Scholar] [CrossRef]
  79. Rawstron, A.C.; Owen, R.G.; Davies, F.E.; Johnson, R.J.; Jones, R.A.; Richards, S.J.; Evans, P.A.; Child, J.A.; Smith, G.M.; Jack, A.S.; et al. Circulating plasma cells in multiple myeloma: Characterization and correlation with disease stage. Br. J. Haematol. 1997, 97, 46–55. [Google Scholar] [CrossRef] [Green Version]
  80. Nowakowski, G.S.; Witzig, T.E.; Dingli, D.; Tracz, M.J.; Gertz, M.A.; Lacy, M.Q.; Lust, J.A.; Dispenzieri, A.; Greipp, P.R.; Kyle, R.A.; et al. Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma. Blood 2005, 106, 2276–2279. [Google Scholar] [CrossRef] [Green Version]
  81. Gonsalves, W.I.; Rajkumar, S.V.; Gupta, V.; Morice, W.G.; Timm, M.M.; Singh, P.P.; Dispenzieri, A.; Buadi, F.K.; Lacy, M.Q.; Kapoor, P.; et al. Quantification of clonal circulating plasma cells in newly diagnosed multiple myeloma: Implications for redefining high-risk myeloma. Leukemia 2014, 28, 2060–2065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Gonsalves, W.I.; Morice, W.G.; Rajkumar, V.; Gupta, V.; Timm, M.M.; Dispenzieri, A.; Buadi, F.K.; Lacy, M.Q.; Singh, P.P.; Kapoor, P.; et al. Quantification of clonal circulating plasma cells in relapsed multiple myeloma. Br. J. Haematol. 2014, 167, 500–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chakraborty, R.; Muchtar, E.; Kumar, S.K.; Jevremovic, D.; Buadi, F.K.; Dingli, D.; Dispenzieri, A.; Hayman, S.R.; Hogan, W.J.; Kapoor, P.; et al. Risk stratification in myeloma by detection of circulating plasma cells prior to autologous stem cell transplantation in the novel agent era. Blood Cancer J. 2016, 6, e512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kumar, S.; Rajkumar, S.V.; Kyle, R.A.; Lacy, M.Q.; Dispenzieri, A.; Fonseca, R.; Lust, J.A.; Gertz, M.A.; Greipp, P.R.; Witzig, T.E. Prognostic value of circulating plasma cells in monoclonal gammopathy of undetermined significance. J. Clin. Oncol. 2005, 23, 5668–5674. [Google Scholar] [CrossRef] [PubMed]
  85. Gonsalves, W.I.; Rajkumar, S.V.; Dispenzieri, A.; Dingli, D.; Timm, M.M.; Morice, W.G.; Lacy, M.Q.; Buadi, F.K.; Go, R.S.; Leung, N.; et al. Quantification of circulating clonal plasma cells via multiparametric flow cytometry identifies patients with smoldering multiple myeloma at high risk of progression. Leukemia 2017, 31, 130–135. [Google Scholar] [CrossRef] [Green Version]
  86. Gonsalves, W.I.; Jevremovic, D.; Nandakumar, B.; Dispenzieri, A.; Buadi, F.K.; Dingli, D.; Lacy, M.Q.; Hayman, S.R.; Kapoor, P.; Leung, N.; et al. Enhancing the R-ISS classification of newly diagnosed multiple myeloma by quantifying circulating clonal plasma cells. Am. J. Hematol. 2020, 95, 310–315. [Google Scholar] [CrossRef]
  87. Han, W.; Jin, Y.; Xu, M.; Zhao, S.-S.; Shi, Q.; Qu, X.; Zhang, R.; Li, J.; Wu, Y.; Chen, L. Prognostic value of circulating clonal plasma cells in newly diagnosed multiple myeloma. Hematology 2021, 26, 510–517. [Google Scholar] [CrossRef]
  88. Sanoja-Flores, L.; Flores-Montero, J.; Garcés, J.J.; Paiva, B.; Puig, N.; García-Mateo, A.; García-Sánchez, O.; Corral-Mateos, A.; Burgos, L.; Blanco, E.; 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, 117. [Google Scholar] [CrossRef] [Green Version]
  89. Terpos, E.; Kostopoulos, I.V.; Papanota, A.-M.; Papadimitriou, K.; Malandrakis, P.; Micheli, P.; Ntanasis-Stathopoulos, I.; Fotiou, D.; Metousis, A.; Kanellias, N.; et al. Next Generation Flow Cytometry Provides a Standardized, Highly Sensitive and Informative Method for the Analysis of Circulating Plasma Cells in Newly Diagnosed Multiple Myeloma: A Single Center Study in 182 Patients. Blood 2019, 134, 4338. [Google Scholar] [CrossRef]
  90. 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. [Google Scholar] [CrossRef] [Green Version]
  91. Paiva, B.; Paino, T.; Sayagues, J.-M.M.; Garayoa, M.; San-Segundo, L.; Martín, M.; Mota, I.; Sanchez, M.-L.L.; Bárcena, P.; Aires-Mejia, I.; et al. Detailed characterization of multiple myeloma circulating tumor cells shows unique phenotypic, cytogenetic, functional, and circadian distribution profile. Blood 2013, 122, 3591–3598. [Google Scholar] [CrossRef] [Green Version]
  92. 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. [Google Scholar] [CrossRef] [Green Version]
  93. Huhn, S.; Weinhold, N.; Nickel, J.; Pritsch, M.; Hielscher, T.; Hummel, M.; Bertsch, U.; Huegle-Doerr, B.; Vogel, M.; Angermund, R.; et al. Circulating tumor cells as a biomarker for response to therapy in multiple myeloma patients treated within the GMMG-MM5 trial. Bone Marrow Transplant. 2017, 52, 1194–1198. [Google Scholar] [CrossRef]
  94. Garcés, J.-J.; Bretones, G.; Burgos, L.; Valdes-Mas, R.; Puig, N.; Cedena, M.-T.; Alignani, D.; Rodriguez, I.; Puente, D.Á.; Álvarez, M.-G.; et al. Circulating tumor cells for comprehensive and multiregional non-invasive genetic characterization of multiple myeloma. Leukemia 2020, 34, 3007–3018. [Google Scholar] [CrossRef]
  95. Mathai, R.A.; Vidya, R.V.S.; Reddy, B.S.; Thomas, L.; Udupa, K.; Kolesar, J.; Rao, M. Potential Utility of Liquid Biopsy as a Diagnostic and Prognostic Tool for the Assessment of Solid Tumors: Implications in the Precision Oncology. J. Clin. Med. 2019, 8, 373. [Google Scholar] [CrossRef] [Green Version]
  96. Biancon, G.; Gimondi, S.; Vendramin, A.; Carniti, C.; Corradini, P. Noninvasive Molecular Monitoring in Multiple Myeloma Patients Using Cell-Free Tumor DNA: A Pilot Study. J. Mol. Diagn. 2018, 20, 859–870. [Google Scholar] [CrossRef] [Green Version]
  97. Kis, O.; Kaedbey, R.; Chow, S.; Danesh, A.; Dowar, M.; Li, T.; Li, Z.; Liu, J.; Mansour, M.; Masih-Khan, E.; et al. Circulating tumour DNA sequence analysis as an alternative to multiple myeloma bone marrow aspirates. Nat. Commun. 2017, 8, 15086. [Google Scholar] [CrossRef]
  98. Oberle, A.; Brandt, A.; Voigtlaender, M.; Thiele, B.; Radloff, J.; Schulenkorf, A.; Alawi, M.; Akyüz, N.; März, M.; Ford, C.T.; et al. Monitoring multiple myeloma by next-generation sequencing of V(D)J rearrangements from circulating myeloma cells and cell-free myeloma DNA. Haematologica 2017, 102, 1105–1111. [Google Scholar] [CrossRef]
  99. Vrabel, D.; Sedlarikova, L.; Besse, L.; Rihova, L.; Bezdekova, R.; Almasi, M.; Kubaczkova, V.; Brožová, L.; Jarkovsky, J.; Plonkova, H.; et al. Dynamics of tumor-specific cfDNA in response to therapy in multiple myeloma patients. Eur. J. Haematol. 2020, 104, 190–197. [Google Scholar] [CrossRef]
  100. Arnault Carneiro, E.; Barahona, F.; Pestana, C.; João, C. Is Circulating DNA and Tumor Cells in Myeloma the Way Forward? Hemato 2022, 3, 63–81. [Google Scholar] [CrossRef]
  101. Thoren, K.L. Mass spectrometry methods for detecting monoclonal immunoglobulins in multiple myeloma minimal residual disease. Semin. Hematol. 2018, 55, 41–43. [Google Scholar] [CrossRef]
  102. Mills, J.R.; Barnidge, D.R.; Dispenzieri, A.; Murray, D.L. High sensitivity blood-based M-protein detection in sCR patients with multiple myeloma. Blood Cancer J. 2017, 7, e590. [Google Scholar] [CrossRef]
  103. Bergen, H.R., 3rd; Dasari, S.; Dispenzieri, A.; Mills, J.R.; Ramirez-Alvarado, M.; Tschumper, R.C.; Jelinek, D.F.; Barnidge, D.R.; Murray, D.L. Clonotypic Light Chain Peptides Identified for Monitoring Minimal Residual Disease in Multiple Myeloma without Bone Marrow Aspiration. Clin. Chem. 2016, 62, 243–251. [Google Scholar] [CrossRef] [Green Version]
  104. Barnidge, D.R.; Tschumper, R.C.; Theis, J.D.; Snyder, M.R.; Jelinek, D.F.; Katzmann, J.A.; Dispenzieri, A.; Murray, D.L. Monitoring M-proteins in patients with multiple myeloma using heavy-chain variable region clonotypic peptides and LC-MS/MS. J. Proteome Res. 2014, 13, 1905–1910. [Google Scholar] [CrossRef]
  105. North, S.J.; Barnidge, D.R.; Brusseau, S.; Patel, R.I.; Haselton, M.; Du Chateau, B.K.; Wallis, G.L.F.; Harding, S.; Sakrikar, D.J.; Ashby, J.C. QIP-MS: A specific, sensitive, accurate, and quantitative alternative to electrophoresis that can identify endogenous m-proteins and distinguish them from therapeutic monoclonal antibodies in patients being treated for multiple myeloma. Clin. Chim. Acta 2019, 493, S433. [Google Scholar] [CrossRef]
  106. Dispenzieri, A.; Krishnan, A.Y.; Arendt, B.; Dasari, S.; Efebera, Y.A.; Geller, N.; Giralt, S.; Hahn, T.; Kohlhagen, M.C.; Landau, H.J.; et al. MASS-FIX versus standard methods to predict for PFS and OS among multiple myeloma patients participating on the STAMINA trial. J. Clin. Oncol. 2021, 39, 8009. [Google Scholar] [CrossRef]
  107. Puig, N.; Paiva, B.; Contreras, T.; Cedena, M.T.; Rosiñol, L.; Martínez, J.; Oriol, A.; Blanchard, M.J.; Rios, R.; Iñigo, M.B.; et al. Analysis of minimal residual disease in bone marrow by NGF and in peripheral blood by mass spectrometry in newly diagnosed multiple myeloma patients enrolled in the GEM2012MENOS65 clinical trial. J. Clin. Oncol. 2021, 39, 8010. [Google Scholar] [CrossRef]
  108. Jiménez Ubieto, A.; Martinez Lopez, J.; Rosinol, L.; Paiva, B.; Cedena, M.T.; Puig, N.; Calasanz, M.J.; Gonzalez Medina, J.; Martin-Ramos, M.L.; Fernandez, R.A.; et al. Absence of Contribution to a Differential Outcome of the Stringent Complete Response IMWG Category Respect to the Conventional CR in Multiple Myeloma. A Validation Analysis Based on the Pethema/GEM2012MENOS65 Phase III Clinical Trial. Blood 2018, 132, 1943. [Google Scholar] [CrossRef]
  109. Bal, S.; Weaver, A.; Cornell, R.F.; Costa, L.J. Challenges and opportunities in the assessment of measurable residual disease in multiple myeloma. Br. J. Haematol. 2019, 186, 807–819. [Google Scholar] [CrossRef] [PubMed]
  110. Kapoor, P.; Kumar, S.K.; Dispenzieri, A.; Lacy, M.Q.; Buadi, F.; Dingli, D.; Russell, S.J.; Hayman, S.R.; Witzig, T.E.; Lust, J.A.; et al. Importance of achieving stringent complete response after autologous stem-cell transplantation in multiple myeloma. J. Clin. Oncol. 2013, 31, 4529–4535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lopez-Anglada, L.; Cueto-Felgueroso, C.; Rosiñol, L.; Oriol, A.; Teruel, A.I.; Lopez de la Guia, A.; Bengoechea, E.; Palomera, L.; de Arriba, F.; Hernandez, J.M.; et al. Prognostic utility of serum free light chain ratios and heavy-light chain ratios in multiple myeloma in three PETHEMA/GEM phase III clinical trials. PLoS ONE 2018, 13, e0203392. [Google Scholar] [CrossRef] [Green Version]
  112. Paiva, B.; Gutiérrez, N.C.; Rosiñol, L.; Vídriales, M.-B.; Montalbán, M.-Á.; Martínez-López, J.; Mateos, M.-V.; Cibeira, M.-T.; Cordón, L.; Oriol, A.; et al. High-risk cytogenetics and persistent minimal residual disease by multiparameter flow cytometry predict unsustained complete response after autologous stem cell transplantation in multiple myeloma. Blood 2012, 119, 687–691. [Google Scholar] [CrossRef]
  113. Lahuerta, J.-J.J.; Paiva, B.; Vidriales, M.-B.B.; Cordón, L.; Cedena, M.-T.T.; Puig, N.; Martinez-Lopez, J.; Rosiñol, L.; Gutierrez, N.C.; Martín-Ramos, M.-L.L.; et al. Depth of response in multiple myeloma: A pooled analysis of three PETHEMA/GEM clinical trials. J. Clin. Oncol. 2017, 35, 2900–2910. [Google Scholar] [CrossRef] [Green Version]
  114. Munshi, N.C.; Avet-Loiseau, H.; Rawstron, A.C.; Owen, R.G.; Child, J.A.; Thakurta, A.; Sherrington, P.; Samur, M.K.; Georgieva, A.; Anderson, K.C.; 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. [Google Scholar] [CrossRef]
  115. Landgren, O.; Devlin, S.; Boulad, M.; Mailankody, S. Role of MRD status in relation to clinical outcomes in newly diagnosed multiple myeloma patients: A meta-analysis. Bone Marrow Transplant. 2016, 51, 1565–1568. [Google Scholar] [CrossRef] [Green Version]
  116. Goicoechea, I.; Puig, N.; Cedena, M.-T.; Burgos, L.; Cordón, L.; Vidriales, M.-B.; Flores-Montero, J.; Gutierrez, N.C.; Calasanz, M.-J.; Ramos, M.-L.M.; et al. Deep MRD profiling defines outcome and unveils different modes of treatment resistance in standard- and high-risk myeloma. Blood 2021, 137, 49–60. [Google Scholar] [CrossRef]
  117. Paiva, B.; Cedena, M.T.; Puig, N.; Arana, P.; Vidriales, M.B.; Cordon, L.; Flores-Montero, J.; Gutierrez, N.C.; Martín-Ramos, M.L.; Martinez-Lopez, J.; et al. Minimal residual disease monitoring and immune profiling in multiple myeloma in elderly patients. Blood 2016, 127, 3165–3174. [Google Scholar] [CrossRef]
  118. 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. [Google Scholar] [CrossRef]
  119. Li, H.; Li, F.; Zhou, X.; Mei, J.; Song, P.; An, Z.; Zhao, Q.; Guo, X.; Wang, X.; Zhai, Y. Achieving minimal residual disease-negative by multiparameter flow cytometry may ameliorate a poor prognosis in MM patients with high-risk cytogenetics: A retrospective single-center analysis. Ann. Hematol. 2019, 98, 1185–1195. [Google Scholar] [CrossRef]
  120. Dimopoulos, M.A.A.; Moreau, P.; Terpos, E.; Mateos, M.V.V.; Zweegman, S.; Cook, G.; Delforge, M.; Hájek, R.; Schjesvold, F.; Cavo, M.; et al. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2021, 32, 309–322. [Google Scholar] [CrossRef]
  121. 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. [Google Scholar] [CrossRef]
  122. Roussel, M.; Lauwers-Cances, V.; Robillard, N.; Hulin, C.; Leleu, X.; Benboubker, L.; Marit, G.; Moreau, P.; Pegourie, B.; Caillot, D.; et al. Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: A phase II study by the Intergroupe Francophone du Myélome. J. Clin. Oncol. 2014, 32, 2712–2717. [Google Scholar] [CrossRef]
  123. Palumbo, A.; Cavallo, F.; Gay, F.; Di Raimondo, F.; Ben Yehuda, D.; Petrucci, M.T.; Pezzatti, S.; Caravita, T.; Cerrato, C.; Ribakovsky, E.; et al. Autologous transplantation and maintenance therapy in multiple myeloma. N. Engl. J. Med. 2014, 371, 895–905. [Google Scholar] [CrossRef]
  124. Gay, F.; Oliva, S.; Petrucci, M.T.; Conticello, C.; Catalano, L.; Corradini, P.; Siniscalchi, A.; Magarotto, V.; Pour, L.; Carella, A.; 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, 1617–1629. [Google Scholar] [CrossRef]
  125. Cavo, M.; Gay, F.; Beksac, M.; Pantani, L.; Petrucci, M.T.; Dimopoulos, M.A.; Dozza, L.; van der Holt, B.; Zweegman, S.; Oliva, S.; 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, e456–e468. [Google Scholar] [CrossRef]
  126. Hahn, T.E.; Wallace, P.K.; Fraser, R.; Fei, M.; Tario, J.D.; Howard, A.; Zhang, Y.; Blackwell, B.; Brunstein, C.G.; Efebera, Y.A.; et al. Minimal Residual Disease (MRD) Assessment before and after Autologous Hematopoietic Cell Transplantation (AutoHCT) and Maintenance for Multiple Myeloma (MM): Results of the Prognostic Immunophenotyping for Myeloma Response (PRIMeR) Study. Biol. Blood Marrow Transplant. 2019, 25, S4–S6. [Google Scholar] [CrossRef]
  127. San-Miguel, J.; Avet-Loiseau, H.; Paiva, B.; Kumar, S.; Dimopoulos, M.A.; Facon, T.; Mateos, M.-V.; Touzeau, C.; Jakubowiak, A.; Usmani, S.Z.; et al. Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE. Blood 2022, 139, 492–501. [Google Scholar] [CrossRef]
  128. Jiménez-Ubieto, A.; Paiva, B.; Puig, N.; Cedena, M.-T.; Martínez-López, J.; Oriol, A.; Blanchard, M.-J.; Ríos, R.; Martin, J.; Martínez, R.; et al. Validation of the International Myeloma Working Group standard response criteria in the PETHEMA/GEM2012MENOS65 study: Are these times of change? Blood 2021, 138, 1901–1905. [Google Scholar] [CrossRef]
  129. Kumar, S.K.; Callander, N.S.; Adekola, K.; Anderson, L.; Baljevic, M.; Campagnaro, E.; Castillo, J.J.; Chandler, J.C.; Costello, C.; Efebera, Y.; et al. Multiple Myeloma, Version 3.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2020, 18, 1685–1717. [Google Scholar] [CrossRef]
  130. Landgren, O.; Iskander, K. Modern multiple myeloma therapy: Deep, sustained treatment response and good clinical outcomes. J. Intern. Med. 2017, 281, 365–382. [Google Scholar] [CrossRef]
  131. Landgren, O.; Hultcrantz, M.; Diamond, B.; Lesokhin, A.M.; Mailankody, S.; Hassoun, H.; Tan, C.; Shah, U.A.; Lu, S.X.; Salcedo, M.; et al. Safety and Effectiveness of Weekly Carfilzomib, Lenalidomide, Dexamethasone, and Daratumumab Combination Therapy for Patients With Newly Diagnosed Multiple Myeloma: The MANHATTAN Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, 862–868. [Google Scholar] [CrossRef] [PubMed]
  132. Holstein, S.A.; Al-Kadhimi, Z.; Costa, L.J.; Hahn, T.; Hari, P.; Hillengass, J.; Jacob, A.; Munshi, N.C.; Oliva, S.; Pasquini, M.C.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. FDA Guidance for Industry. Hematologic Malignancies: Regulatory Considerations for Use of Minimal Residual Disease in Development of Drug and Biological Products for Treatment. Available online: https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/U (accessed on 25 February 2022).
  134. EMA Guideline on the use of Minimal Residual Disease as a Clinical Endpoint in Multiple Myeloma Studies. Available online: https://www.ema.europa.eu/en/documents/scienti_cguideline/%0Adraft-guideline-use-minimal-residual-disease-clinical-endpoint-multiplemyeloma-%0Astudies_en.pdf%0A (accessed on 25 February 2022).
  135. Attal, M.; Lauwers-Cances, V.; Hulin, C.; Leleu, X.; Caillot, D.; Escoffre, M.; Arnulf, B.; Macro, M.; Belhadj, K.; Garderet, L.; et al. Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N. Engl. J. Med. 2017, 376, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
  136. 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, JCO2101935. [Google Scholar] [CrossRef]
  137. Gambella, M.; Omedé, P.; Spada, S.; Muccio, V.E.; Gilestro, M.; Saraci, E.; Grammatico, S.; Larocca, A.; Conticello, C.; Bernardini, A.; 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, 750–760. [Google Scholar] [CrossRef]
  138. Oliva, S.; Gambella, M.; Larocca, A.; Spada, S.; Marzanati, E.; Mantoan, B.; Grammatico, S.; Conticello, C.; Gamberi, B.; Offidani, M.; et al. Prognostic Impact of Minimal Residual Disease By ASO-RQ-PCR in Multiple Myeloma: A Pooled Analysis of 2 Phase III Studies in Patients Treated with Lenalidomide after Front-Line Therapy. Blood 2016, 128, 4409. [Google Scholar] [CrossRef]
  139. Jackson, G.H.; Davies, F.E.; Pawlyn, C.; Cairns, D.A.; Striha, A.; Collett, C.; Hockaday, A.; Jones, J.R.; Kishore, B.; Garg, M.; et al. Lenalidomide maintenance versus observation for patients with newly diagnosed multiple myeloma (Myeloma XI): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2019, 20, 57–73. [Google Scholar] [CrossRef] [Green Version]
  140. Alonso, R.; Cedena, M.-T.; Wong, S.; Shah, N.; Ríos-Tamayo, R.; Moraleda, J.M.; López-Jiménez, J.; García, C.; Bahri, N.; Valeri, A.; et al. Prolonged lenalidomide maintenance therapy improves the depth of response in multiple myeloma. Blood Adv. 2020, 4, 2163–2171. [Google Scholar] [CrossRef]
  141. Amsler, I.G.; Jeker, B.; Mansouri Taleghani, B.; Bacher, U.; Betticher, D.; Egger, T.; Zander, T.; Luethi, J.-M.; Novak, U.; Pabst, T. Prolonged survival with increasing duration of lenalidomide maintenance after autologous transplant for multiple myeloma. Leuk. Lymphoma 2019, 60, 511–514. [Google Scholar] [CrossRef]
  142. Mian, I.; Milton, D.R.; Shah, N.; Nieto, Y.; Popat, U.R.; Kebriaei, P.; Parmar, S.; Oran, B.; Shah, J.J.; Manasanch, E.E.; et al. Prolonged survival with a longer duration of maintenance lenalidomide after autologous hematopoietic stem cell transplantation for multiple myeloma. Cancer 2016, 122, 3831–3837. [Google Scholar] [CrossRef] [Green Version]
  143. 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, 24, 2568–2574. [Google Scholar] [CrossRef] [Green Version]
  144. Ferrero, S.; Ladetto, M.; Drandi, D.; Cavallo, F.; Genuardi, E.; Urbano, M.; Caltagirone, S.; Grasso, M.; Rossini, F.; Guglielmelli, T.; 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, 689–695. [Google Scholar] [CrossRef]
  145. Oliva, S.; Gambella, M.; Gilestro, M.; Muccio, V.E.; Gay, F.; Drandi, D.; Ferrero, S.; Passera, R.; Pautasso, C.; Bernardini, A.; et al. Minimal residual disease after transplantation or lenalidomide-based consolidation in myeloma patients: A prospective analysis. Oncotarget 2017, 8, 5924–5935. [Google Scholar] [CrossRef] [Green Version]
  146. Mina, R.; Belotti, A.; Petrucci, M.T.; Zambello, R.; Capra, A.; Di Lullo, G.; Ronconi, S.; Pescosta, N.; Grasso, M.; Monaco, F.; et al. Bortezomib-dexamethasone as maintenance therapy or early retreatment at biochemical relapse versus observation in relapsed/refractory multiple myeloma patients: A randomized phase II study. Blood Cancer J. 2020, 10, 58. [Google Scholar] [CrossRef]
  147. Mohan, M.; Kendrick, S.; Szabo, A.; Yarlagadda, N.; Atwal, D.; Pandey, Y.; Roy, A.; Parikh, R.; Lopez, J.; Thanendrarajan, S.; et al. Clinical implications of loss of bone marrow minimal residual disease negativity in multiple myeloma. Blood Adv. 2022, 6, 808–817. [Google Scholar] [CrossRef]
  148. Moreau, P.; Siegel, D.S.; Goldschmidt, H.; Niesvizky, R.; Bringhen, S.; Orlowski, R.Z.; Blaedel, J.; Yang, Z.; Dimopoulos, M.A. Subgroup Analysis of Patients with Biochemical or Symptomatic Relapse at the Time of Enrollment in the Endeavor Study. Blood 2018, 132, 3243. [Google Scholar] [CrossRef]
  149. Rasmussen, A.-M.; Askeland, F.B.; Schjesvold, F. The Next Step for MRD in Myeloma? Treating MRD Relapse after First Line Treatment in the REMNANT Study. Hemato 2020, 1, 36–48. [Google Scholar] [CrossRef]
  150. Dimopoulos, M.; Quach, H.; Mateos, M.-V.; Landgren, O.; Leleu, X.; Siegel, D.; Weisel, K.; Yang, H.; Klippel, Z.; Zahlten-Kumeli, A.; 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 2020, 396, 186–197. [Google Scholar] [CrossRef]
  151. Palumbo, A.; Chanan-Khan, A.; Weisel, K.; Nooka, A.K.; Masszi, T.; Beksac, M.; Spicka, I.; Hungria, V.; Munder, M.; Mateos, M.V.; et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2016, 375, 754–766. [Google Scholar] [CrossRef]
  152. Avet-Loiseau, H.; San-Miguel, J.; Casneuf, T.; Iida, S.; Lonial, S.; Usmani, S.Z.; Spencer, A.; Moreau, P.; Plesner, T.; Weisel, K.; 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. Med. 2021, 39, 1139–1149. [Google Scholar] [CrossRef]
  153. Munshi, N.C.; Berdeja, J.G.; Lin, Y.; Kochenderfer, J.; Raje, N.S.; Liedtke, M.; Jagannath, S.; Madduri, D.; Rosenblatt, J.; Maus, M.V.; 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. [Google Scholar] [CrossRef]
  154. Costa, L.J.; Wong, S.W.; Bermúdez, A.; de la Rubia, J.; Mateos, M.-V.; Ocio, E.M.; Rodríguez-Otero, P.; San-Miguel, J.; Li, S.; Sarmiento, R.; et al. First Clinical Study of the B-Cell Maturation Antigen (BCMA) 2+1 T Cell Engager (TCE) CC-93269 in Patients (Pts) with Relapsed/Refractory Multiple Myeloma (RRMM): Interim Results of a Phase 1 Multicenter Trial. Blood 2019, 134, 143. [Google Scholar] [CrossRef]
  155. Attal, M.; Richardson, P.G.; Rajkumar, S.V.; San-Miguel, J.; Beksac, M.; Spicka, I.; Leleu, X.; Schjesvold, F.; Moreau, P.; Dimopoulos, M.A.; 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 2019, 394, 2096–2107. [Google Scholar] [CrossRef]
  156. Bahlis, N.J.; Dimopoulos, M.A.; White, D.J.; Benboubker, L.; Cook, G.; Leiba, M.; Ho, P.J.; Kim, K.; Takezako, N.; Moreau, P.; 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, 1875–1884. [Google Scholar] [CrossRef] [Green Version]
  157. Martin, T.; Usmani, S.Z.; Berdeja, J.G.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Hari, P.; Avigan, D.; Deol, A.; Htut, M.; et al. Updated Results from CARTITUDE-1: Phase 1b/2Study of Ciltacabtagene Autoleucel, a B-Cell Maturation Antigen-Directed Chimeric Antigen Receptor T Cell Therapy, in Patients With Relapsed/Refractory Multiple Myeloma. Blood 2021, 138, 549. [Google Scholar] [CrossRef]
  158. Wang, B.-Y.; Zhao, W.-H.; Liu, J.; Chen, Y.-X.; Cao, X.-M.; Yang, Y.; Zhang, Y.-L.; Wang, F.-X.; Zhang, P.-Y.; Lei, B.; et al. Long-Term Follow-up of a Phase 1, First-in-Human Open-Label Study of LCAR-B38M, a Structurally Differentiated Chimeric Antigen Receptor T (CAR-T) Cell Therapy Targeting B-Cell Maturation Antigen (BCMA), in Patients (pts) with Relapsed/Refractory Multiple Myeloma (RRMM). Blood 2019, 134, 579. [Google Scholar] [CrossRef]
  159. Munshi, N.C.; Anderson, L.D.J.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef]
  160. Da Vià, M.C.; Dietrich, O.; Truger, M.; Arampatzi, P.; Duell, J.; Heidemeier, A.; Zhou, X.; Danhof, S.; Kraus, S.; Chatterjee, M.; et al. Homozygous BCMA gene deletion in response to anti-BCMA CAR T cells in a patient with multiple myeloma. Nat. Med. 2021, 27, 616–619. [Google Scholar] [CrossRef]
  161. Mailankody, S.; Jakubowiak, A.J.; Htut, M.; Costa, L.J.; Lee, K.; Ganguly, S.; Kaufman, J.L.; Siegel, D.S.D.; Bensinger, W.; Cota, M.; et al. Orvacabtagene autoleucel (orva-cel), a B-cell maturation antigen (BCMA)-directed CAR T cell therapy for patients (pts) with relapsed/refractory multiple myeloma (RRMM): Update of the phase 1/2 EVOLVE study (NCT03430011). J. Clin. Oncol. 2020, 38, 8504. [Google Scholar] [CrossRef]
  162. Mohyuddin, G.R.; Rooney, A.; Balmaceda, N.; Aziz, M.; Sborov, D.W.; McClune, B.; Kumar, S.K. Chimeric antigen receptor T-cell therapy in multiple myeloma: A systematic review and meta-analysis of 950 patients. Blood Adv. 2021, 5, 1097–1101. [Google Scholar] [CrossRef]
  163. Bravo-Pérez, C.; Sola, M.; Teruel-Montoya, R.; García-Malo, M.D.; Ortuño, F.J.; Vicente, V.; de Arriba, F.; Jerez, A. Minimal Residual Disease in Multiple Myeloma: Something Old, Something New. Cancers 2021, 13, 4332. [Google Scholar] [CrossRef] [PubMed]
  164. Topp, M.S.; Duell, J.; Zugmaier, G.; Attal, M.; Moreau, P.; Langer, C.; Krönke, J.; Facon, T.; Salnikov, A.V.; Lesley, R.; et al. Anti-B-Cell Maturation Antigen BiTE Molecule AMG 420 Induces Responses in Multiple Myeloma. J. Clin. Oncol. 2020, 38, 775–783. [Google Scholar] [CrossRef] [PubMed]
  165. Kaufman, J.L.; Baz, R.C.; Harrison, S.J.; Quach, H.; Ho, S.-J.; Vangsted, A.J.; Moreau, P.; Gibbs, S.D.J.; Salem, A.H.; Coppola, S.; et al. Updated analysis of a phase I/II study of venetoclax in combination with daratumumab and dexamethasone, +/- bortezomib, in patients with relapsed/refractory multiple myeloma. J. Clin. Oncol. 2020, 38, 8511. [Google Scholar] [CrossRef]
  166. Lutz, R.; Mahmoud, A.; Awwad, M.H.S.; Imbusch, C.D.; Boch, T.; Durie, B.G.M.; Weinhold, N.; Raab, M.S.; Müller-Tidow, C.; Haas, S.; et al. The Bone Marrow Microenvironment of Multiple Myeloma Long-Term Survivors at Single Cell Resolution. Blood 2020, 136, 32–33. [Google Scholar] [CrossRef]
  167. Ho, C.M.; McCarthy, P.L.; Wallace, P.K.; Zhang, Y.; Fora, A.; Mellors, P.; Tario, J.D.; McCarthy, B.L.S.; Chen, G.L.; Holstein, S.A.; et al. Immune signatures associated with improved progression-free and overall survival for myeloma patients treated with AHSCT. Blood Adv. 2017, 1, 1056–1066. [Google Scholar] [CrossRef] [Green Version]
  168. Bhutani, M.; Foureau, D.; Zhang, Q.; Robinson, M.; Wynn, A.S.; Steuerwald, N.M.; Druhan, L.J.; Guo, F.; Rigby, K.; Turner, M.; et al. Peripheral Immunotype Correlates with Minimal Residual Disease Status and Is Modulated by Immunomodulatory Drugs in Multiple Myeloma. Biol. Blood Marrow Transplant. 2019, 25, 459–465. [Google Scholar] [CrossRef] [Green Version]
  169. Papadimitriou, K.; Tsakirakis, N.; Malandrakis, P.; Vitsos, P.; Metousis, A.; Orologas-Stavrou, N.; Ntanasis-Stathopoulos, I.; Kanellias, N.; Eleutherakis-Papaiakovou, E.; Pothos, P.; et al. Deep Phenotyping Reveals Distinct Immune Signatures Correlating with Prognostication, Treatment Responses, and MRD Status in Multiple Myeloma. Cancers 2020, 12, 3245. [Google Scholar] [CrossRef]
  170. Paiva, B.; Vídriales, M.B.; Rosiñol, L.; Martínez-López, J.; Mateos, M.V.; Ocio, E.M.; Montalbán, M.A.; Cordón, L.; Gutiérrez, N.C.; Corchete, L.; et al. A multiparameter flow cytometry immunophenotypic algorithm for the identification of newly diagnosed symptomatic myeloma with an MGUS-like signature and long-term disease control. Leukemia 2013, 27, 2056–2061. [Google Scholar] [CrossRef] [Green Version]
  171. De Magalhães, R.J.P.; Vidriales, M.B.; Paiva, B.; Fernandez-Gimenez, C.; García-Sanz, R.; Mateos, M.V.; Gutierrez, N.C.; Lecrevisse, Q.; Blanco, J.F.; Hernández, J.; et al. Analysis of the immune system of multiple myeloma patients achieving long-term disease control by multidimensional flow cytometry. Haematologica 2013, 98, 79–86. [Google Scholar] [CrossRef] [Green Version]
  172. Coffey, D.G.; Maura, F.; Gonzalez-Kozlova, E.; Diaz-Mejia3, J.; Luo, P.; Zhang, Y.; Xu, Y.; Warren, E.H.; Smith, E.L.; Cho, H.J.; et al. Normalization of the Immune Microenvironment during Lenalidomide Maintenance Is Associated with Sustained MRD Negativity in Patients with Multiple Myeloma. Blood 2021, 138, 329. [Google Scholar] [CrossRef]
  173. Shaughnessy, J.D.J.; Zhan, F.; Burington, B.E.; Huang, Y.; Colla, S.; Hanamura, I.; Stewart, J.P.; Kordsmeier, B.; Randolph, C.; Williams, D.R.; et al. A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1. Blood 2007, 109, 2276–2284. [Google Scholar] [CrossRef] [Green Version]
  174. Zhan, F.; Barlogie, B.; Arzoumanian, V.; Huang, Y.; Williams, D.R.; Hollmig, K.; Pineda-Roman, M.; Tricot, G.; van Rhee, F.; Zangari, M.; et al. Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis. Blood 2007, 109, 1692–1700. [Google Scholar] [CrossRef] [Green Version]
  175. Schinke, C.; Hoering, A.; Wang, H.; Carlton, V.; Thanandrarajan, S.; Deshpande, S.; Patel, P.; Molnar, G.; Susanibar, S.; Mohan, M.; 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, e313–e316. [Google Scholar] [CrossRef] [Green Version]
  176. Palumbo, A.; Avet-Loiseau, H.; Oliva, S.; Lokhorst, H.M.; Goldschmidt, H.; Rosinol, L.; Richardson, P.; Caltagirone, S.; Lahuerta, J.J.; Facon, T.; et al. Revised international staging system for multiple myeloma: A report from international myeloma working group. J. Clin. Oncol. 2015, 33, 2863–2869. [Google Scholar] [CrossRef]
  177. Brown, P.A.; Shah, B.; Advani, A.; Aoun, P.; Boyer, M.W.; Burke, P.W.; DeAngelo, D.J.; Dinner, S.; Fathi, A.T.; Gauthier, J.; et al. Acute Lymphoblastic Leukemia, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2021, 19, 1079–1109. [Google Scholar] [CrossRef]
  178. Hochhaus, A.; Baccarani, M.; Silver, R.T.; Schiffer, C.; Apperley, J.F.; Cervantes, F.; Clark, R.E.; Cortes, J.E.; Deininger, M.W.; Guilhot, F.; et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia 2020, 34, 966–984. [Google Scholar] [CrossRef] [Green Version]
  179. Clinicaltrials.gov. Available online: https://clinicaltrials.gov/ct2/home (accessed on 25 February 2022).
Table 1. Characterization of currently available techniques for MRD evaluation in MM recommended by the IMWG.
Table 1. Characterization of currently available techniques for MRD evaluation in MM recommended by the IMWG.
Next Generation Flow
(NGF)
Next Generation Sequencing (NGS)PET/CT
Applicability~100%~90%~90% (specifically extramedullary disease)
AvailabilityHigh (≥8 colors needed)Low (commercial and academic platforms)Intermediate
Sample at
diagnosis
Not obligatoryObligatory (identification of dominant clone)Not obligatory (required for identification of focal lesions or extramedullary disease
Number of cells required10 × 106 cells1–3 × 106 cellsNA
Sample
processing
Fresh samples
Processing within 24–48 h
Fresh/frozen samplesNA
Patchy sampleImpactImpactNo impact
Whole sample characterizationYes (global cell characterization)NoNo
Clonal
evaluation
Not possiblePossible (identification of minor clones)Possible (requires focal lesion biopsy)
Quantitative methodYesYesYes
StandardizationYes
(EuroFlow Consortium)
Yes (Adaptive Biotechnologies; FDA approved)No (ongoing)
Sensitivity10−5–10−610−5–10−6High (4 mm)
Time to results3–4 h7 days2 h
ComplexityCytometry skills; automated analysis availableBioinformatic supportNuclear medicine support
ReproducibilityHighHighModerate
Abbreviations: NGF—Next Generation Flow; NGS—Next Generation Sequencing; PET/CT—Positron Emission Tomography/Computed Tomography; NA—Not Applicable; FDA—Food and Drug Administration.
Table 2. Examples of clinical trials that use MRD testing to guide treatment decisions [179].
Table 2. Examples of clinical trials that use MRD testing to guide treatment decisions [179].
TitlePhaseObjectiveStudy
Population
Time Point AssessmentDecisionTreatmentPrimary OutcomeMRD Method
MASTER trial (NCT03224507)2Decision to begin maintenanceNDMM after Dara-KRd and ASCTEnd of consolidationMRD+Maintenance Dara-KRdMRD negativity rateNGS (10−5)
MRD-Observation (2x MRD-)
DART4MM study (NCT03992170)2Effect of Dara on MRD positive patientsMRD+ patients >VGPRMaintenance (24 weeks)MRD+Dara (80 weeks)MRD negativity rateNGF (10−5)
MRD-Stop treatment
AURIGA study (NCT03901963)3Guide maintenanceMRD+ patients after ASCTMaintenanceMRD+Dara-RMRD negative statusNGS (10−5)
MRD-R
NCT041401622Guide consolidation after initial therapyNDMM after Dara-Rd inductionAfter inductionMRD+Consolidation Dara-RVdMRD negativity rateNGS (10−5)
MRD-Maintenance Dara-R/R
DRAMMATIC study (NCT04071457)3Guide maintenance after initial therapyNDMM after randomization to Dara-R versus R following ASCTMaintenance (2 years) (R versus Dara/rHuPH20)MRD+Continue maintenanceOSNGS (10−5)
MRD-Continue versus stopping maintenance
REMNANT study (NCT04513639)2–3Guide early treatment of relapseNDMM MRD- after VRd and ASCTAfter consolidation (MRD assessed every 4 months)MRD+Dara-KdMRD negativity rate; PFS; OSNGF (10−5)
MRD-Observation (until PD)
MRD2STOP (NCT04108624)NAGuide maintenance cessationPatients at CR after ASCT during maintenanceMaintenance (≥ 1 year)MRD+Continue maintenanceMRD conversion rate; PFS; OSNGS (exploring 10−7) or NGF and PET/CT
MRD-Stop maintenance
PERSEUS (NCT03710603)3Guide maintenance after initial therapyNDMM after Dara-VRd during maintenance (Dara-R)Maintenance (2 years)MRD+Dara-RPFSNGS (10−5)
MRD-R (sustained MRD- 12 months)
PREDATOR-MRD (NCT03697655)2Role of Dara in MRDRRMM (1–2 prior lines therapy)MaintenanceMRD+DaraEFSNGF (10−5)
MRD-Observation
NCT034903442Effect of Dara on MRD positive patientsPatients in ≤VGPR after induction with/without ASCTMaintenance (Dara-R)__MRD negativity rateNGF (10−5)
NCT04221178NAGuide maintenance cessationMRD- patients under maintenanceMaintenance (≥ 3 years)MRD+ContinueMRD negativity rate after 1 yearNGF (10−5)
MRD-Stop maintenance
NCT026592933Guide duration of maintenanceNDMM patients after ASCTMaintenance (end cycle 6)MRD+KRd (cycle 5–36)PFSNGS (10−5)
MRD-KRd (cycle 5–8)
NCT023895172Effect of Ixa on MRD positive patientsMRD+ patients after ASCTMaintenance
(Ixa-Rd versus R)
__MRD negativity rateNGF (10−5)
NCT029698372Guide duration of maintenanceNDMMMaintenanceMRD+E-KRd (6 cycles) + E-Rd (until PD)MRD negativity rateNGS (10−5)
MRD-E-Rd until PD
NCT040960663Guide maintenance therapyNDMM (≥ 65 years)Maintenance (KRd versus Rd)MRD+KRdMRD negativity rate; PFSNGF or NGS (10−5)
MRD-Rd (after 2 years)
NCT041401622Guide consolidation therapyNDMM after Dara-Rd inductionAfter InductionMRD+DRVd consolidationMRD negativity rateNGF or NGS (10−5)
MRD-Dara-Rd maintenance
NCT050913722Effect of Belantamab mafodotin on MRDNDMM MRD+ patients after ASCTMaintenance__MRD negativity rateNGF or NGS (10−5)
MILESTONE (NCT04991103)2Guide consolidation/maintenance therapyNDMM after Dara-VRdAfter InductionMRD+Proceed to ASCT and MaintenanceMRD negativity rateNGS (10−5)
MRD-Defer ASCT
MIDAS (NCT04934475)3Guide consolidation therapyNDMM after Isa-KRdAfter InductionMRD+ASCT+Isa-KRd versus ASCTMRD negativity rateNGS (10−5)
MRD-Isa-KRd versus ASCT+Isa-KRd
MASTER-2 (NCT05231629)2Guide consolidation/maintenance therapyNDMM after Dara-VRdAfter InductionMRD+ASCT+Dara-Tec versus ASCT+Dara-RDepth of response; sustained MRD negativityNGS (10−5)
MRD-Dara-VRd+Dara-R versus ASCT+Dara-R
HEME-20 (NCT05192122)1Guide maintenance cessationNDMM MRD- after ASCTMaintenance (3 years)MRD+Continue maintenanceSustained MRD negativityNGS (10−5)
MRD-Stop maintenance
Abbreviations: MRD—Measurable Residual Disease; NDMM—Newly Diagnosed Multiple Myeloma patients; RRMM—Relapsed/Refractory Multiple Myeloma patients; NGF—Next Generation Flow; NGS—Next Generation Sequencing; ACST—Autologous Stem Cell Transplant; Dara—Daratumumab; R—Lenalidomide; d—Dexamethasone; Ixa—Ixazomib; K—Carfilzomib; E—Elotuzumab; Tec—Teclistamab; PD—Progressive Disease; OS—Overall Survival; EFS—Event Free Survival; NA—Not Applicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Caetano, J.; Barahona, F.; Lúcio, P.; João, C. Measurable Residual Disease Assessment in Multiple Myeloma: How Deep Is Enough? Hemato 2022, 3, 385-413. https://doi.org/10.3390/hemato3030027

AMA Style

Caetano J, Barahona F, Lúcio P, João C. Measurable Residual Disease Assessment in Multiple Myeloma: How Deep Is Enough? Hemato. 2022; 3(3):385-413. https://doi.org/10.3390/hemato3030027

Chicago/Turabian Style

Caetano, Joana, Filipa Barahona, Paulo Lúcio, and Cristina João. 2022. "Measurable Residual Disease Assessment in Multiple Myeloma: How Deep Is Enough?" Hemato 3, no. 3: 385-413. https://doi.org/10.3390/hemato3030027

Article Metrics

Back to TopTop