Assessment of minimal residual disease (MRD) is becoming standard diagnostic care for potentially curable neoplasms such as acute lymphoblastic leukemia. In multiple myeloma (MM), the majority of patients will inevitably relapse despite achievement of progressively higher complete remission (CR) rates. Novel treatment protocols with inclusion of antibodies and small molecules might well be able to further increase remission rates and potentially also cure rates. Therefore, MRD diagnostics becomes essential to assess treatment effectiveness. This review summarizes reports from the past 2 decades, which demonstrate that persistent MRD by multiparameter flow cytometry, polymerase chain reaction, next-generation sequencing, and positron emission tomography/computed tomography, predicts significantly inferior survival among CR patients. We describe the specific features of currently available techniques for MRD monitoring and outline the arguments favoring new criteria for response assessment that incorporate MRD levels. Extensive data indicate that MRD information can potentially be used as biomarker to evaluate the efficacy of different treatment strategies, help on treatment decisions, and act as surrogate for overall survival. The time has come to address within clinical trials the exact role of baseline risk factors and MRD monitoring for tailored therapy in MM, which implies systematic usage of highly sensitive, cost-effective, readily available, and standardized MRD techniques.

The development of new and effective therapies usually comes along with the need for more sensitive approaches to compare the efficacy of different treatment strategies, and implementation of individualized therapy monitoring strategies to prevent both under- and overtreatment. In the past decade, the landscape of drugs approved for the treatment of multiple myeloma (MM) has rapidly grown, and several agents with novel mechanisms of action are currently in the pipeline.1  This, together with the availability of drugs with well-balanced efficacy/toxicity profiles has recently led to the design of more complex and prolonged treatment strategies.2-7  However, the definition of clinical response criteria and clinical end points has largely remained the same over the past 15 years.8-10  Nevertheless, concepts such as “depth of response,” “minimal residual disease (MRD),” and “surrogate survival markers” have become the subject of extensive research and debate within the MM scientific community (Figure 1) and even the subject of a recent workshop with regulatory agencies.11-15  In this review, we address these concepts and define what remains to be accomplished for optimization of response criteria and full implementation of MRD monitoring in MM into routine clinical practice.

Figure 1

Graphical representation of the increasing number of publications in PUBMED and abstracts reported in the Annual Congress of the American Society of Hematology (ASH) on MM MRD during the past decades. (A) Publications per 5-year periods on MRD studies in MM (PUBMED). (B) Abstracts reported per year at the ASH meetings on MRD studies in MM.

Figure 1

Graphical representation of the increasing number of publications in PUBMED and abstracts reported in the Annual Congress of the American Society of Hematology (ASH) on MM MRD during the past decades. (A) Publications per 5-year periods on MRD studies in MM (PUBMED). (B) Abstracts reported per year at the ASH meetings on MRD studies in MM.

Close modal

For virtually all hematologic malignancies, a direct correlation exists between depth of response and prolonged survival. MM is no exception to such paradigm, and meta-analyses among transplant-eligible and nontransplant candidates have clearly established the link between deep responses such as complete remission (CR) and prolonged survival.16-18  Thus, high-dose therapy (HDT) followed by the incorporation of novel agents into autologous stem cell transplantation (ASCT) trials have significantly improved outcome by achieving higher CR rates.17,19-22  Recent trials with novel agent combinations alone have also resulted in high CR rates (comparable to those previously reported only with HDT/ASCT),23,24  even among patients older than 65,3,25  high-risk patients,26,27  and relapse/refractory MM.28,29  Despite all accumulated evidence, there are still some caveats that should be highlighted. First, achieving the deepest level of remission (ie, CR) is considered to be a prerequisite, not only to prolong survival but also to ultimately achieve cure. Indeed a recent update on Total Therapy trials provides evidence of curability in MM,7  and other long-term analyses have shown that 1 out of 3 patients in CR could potentially be cured (relapse free after 10-years of follow-up).30  Remarkably, also 10% of cases that reach suboptimal response after therapy, such as near CR or (very good) partial response (PR), are relapse free at 10 years.30  This has raised a second question about whether CR is actually needed to achieve long-term survival. Indeed, biologically well-defined patient subgroups with monoclonal gammopathy of undetermined significance (MGUS)-like baseline profiles or specific molecular subtypes can present long-term survival without achieving CR (Figure 2).31-34  However, these patients only represent 10% of total MM patients. Thus, for the vast majority of patients, higher CR rates are indeed needed to increase survival rates and approve (new) treatment regimens.19,21,22,35-38 

Figure 2

Schematic representation to illustrate the paradigm of the deeper the response, the longer the (progression-free) survival (filled lines). However, distinct biological subgroups exist, and their clinical course may differ from the paradigm (dotted lines): a, those patients with a baseline MGUS-like signature and prolonged survival irrespectively of CR; b, those patients with unsustained CR (high-risk cytogenetics and persistent MRD); c, MRD-positive patients who may also experience extended outcomes if small residual clones are quiescent (MGUS-like) or under control (eg, by immune cells); d, an MRD-negative result does not preclude the risk of relapse, and optimization of MRD monitoring together with follow-up MRD studies are likely crucial to predict relapses early on; e, long-term disease control (ie, functional cure) could potentially be achieved if therapy eradicates (detectable) MRD levels. This is a hypothetical model, which does not translate to the real behavior of individual patients.

Figure 2

Schematic representation to illustrate the paradigm of the deeper the response, the longer the (progression-free) survival (filled lines). However, distinct biological subgroups exist, and their clinical course may differ from the paradigm (dotted lines): a, those patients with a baseline MGUS-like signature and prolonged survival irrespectively of CR; b, those patients with unsustained CR (high-risk cytogenetics and persistent MRD); c, MRD-positive patients who may also experience extended outcomes if small residual clones are quiescent (MGUS-like) or under control (eg, by immune cells); d, an MRD-negative result does not preclude the risk of relapse, and optimization of MRD monitoring together with follow-up MRD studies are likely crucial to predict relapses early on; e, long-term disease control (ie, functional cure) could potentially be achieved if therapy eradicates (detectable) MRD levels. This is a hypothetical model, which does not translate to the real behavior of individual patients.

Close modal

A retrospective analysis of 3 randomized European trials for transplant-ineligible patients indicated that ∼40% of CR patients will relapse, and that 20% will die within 4 years after initial therapy.16  Similar results have also been reported for transplant-candidate patients.17,39  In addition, a small fraction of CR patients show early (<1 year) relapse with a very poor survival (≤2 years),40  and similar CR rates after different treatment regimens fail to predict for an overall distinct outcome.4  These data reveal that the quality of CR may largely vary between different regimens. The data also emphasize that current CR criteria, such as negative immunofixation in serum and urine, disappearance of any soft tissue plasmacytomas, and <5% plasma cells (PCs) in bone marrow (BM),8  fail to detect such differences, even among patients who will relapse soon (ie, those who display unsustained CR).

In 2006, the International Myeloma Working Group had already highlighted the need for a new definition of CR and introduced normalization of serum free light chains (sFLCs) and absence of clonal PCs in BM biopsies by immunohistochemistry and/or immunofluorescence as additional requirements to define more stringent CR criteria.8  Since then, only 1 large study has been able to show the superiority of the stringent over conventional CR criteria to define patients’ outcomes,39  whereas other groups failed to demonstrate the utility of the sFLC assay among immunofixation-negative patients,41-43  most likely because the latter groups did not include simultaneous assessment of PC clonality in BM biopsies. Importantly, the vast majority of CR patients after therapy show recovery of normal PCs that exceeds the percentage of clonal PCs,44  implying that solid clonality markers are needed, such as the clonotypic immunoglobulin (Ig) gene sequences. In addition, it has been suggested that the sFLC might be replaced by the heavy-light format45  and become merely a surrogate for recovery of the immune system rather than an MRD monitoring tool.46 

Overall, it becomes clear that the definition of CR would benefit from an improvement that matches the dramatic evolution observed in MM treatment. Such improvement can only be achieved by highly sensitive technologies able to detect MRD at very low levels. Recent data by Rawstron et al47  point out that quantitative assessment of tumor load with a cutoff of 10−4 (using multiparameter flow cytometry [MFC]) would be more informative than a positive vs negative categorization, suggesting that a lower cutoff provided by more sensitive assays (eg, next-generation sequencing [NGS] or high-sensitive MFC) will likely improve outcome prediction further. This has already been confirmed by Martinez-Lopez et al using NGS,48  who identified 3 groups of patients with different time to progression (TTP): patients with high (<10−3), intermediate (10−3 to 10−5), and low (>10−5) MRD levels showed significantly different TTP (27, 48, and 80 months, respectively). Accordingly, 10−5 should currently be considered as the target cutoff level for definition of MRD negativity.

Over the past decade, MFC and Ig allele-specific oligonucleotide–based quantitative polymerase chain reaction (ASO-PCR) have emerged as the most attractive, well-suited, and sensitive approaches to detect MRD in the BM of MM patients during and after therapy. More recently, preliminary studies have also shown that NGS of Ig genes might be applicable for MRD detection in BM of MM patients. However, because of the frequency of extramedullary relapses, sensitive imaging techniques have also become relevant in assessing low levels of disease outside BM.

MFC

MFC is particularly well-suited to study biological samples containing PCs because this worldwide-available technique allows (1) simultaneous identification and characterization of single PCs based on multiple parameters, (2) evaluation of high cell numbers in a few hours, (3) quantitative assessment of different cell populations and their corresponding antigen expression levels, and (4) combined detection of cell surface and intracellular antigens.49 

In recent years, the sensitivity of MFC has increased because of simultaneous assessment of ≥8 markers and evaluation of greater numbers of cells than what was previously feasible with 4-color instruments.50  Single parameters cannot reliably distinguish clonal vs normal PCs, but multiparameter cytometry with evaluation of at least 8 markers in a single tube can readily identify aberrant PC phenotypes at MRD levels if sufficient cell numbers (eg, ≥5 × 106) are evaluated.49  Consensus exists that PC identification markers (CD38 plus CD138) plus discriminatory markers such as CD19, CD27, CD45, CD56, CD81, and CD117 should be simultaneously evaluated for accurate identification of BM PCs and unequivocal distinction between clonal and normal PCs.49-51  It should be noted that normal PCs have a considerably heterogeneous immunophenotype according to the PC maturation process,52,53  but this maturation pathway is highly conserved in all conditions from normal to regenerating and reactive BM samples.50  The understanding of normal PC maturation facilitates the universal MFC-based identification of aberrant PC phenotypes in MM patients (Table 1). This can be further confirmed via the clonal nature of the phenotypically aberrant PCs through cytoplasmic Ig light-chain restriction.50  Because the aberrant phenotypes of clonal PCs are readily distinguishable from normal PCs, flow MRD is applicable in virtually every MM patient without requiring patient-specific diagnostic phenotypic profiles (Table 1). Additionally, discrimination between normal and myeloma PCs is still feasible in the (rare) event of phenotypic shifts from diagnostic to posttreatment MRD samples.54  Most importantly, flow-MRD assays also provide an intra-assay quality check of the whole cell sample via simultaneous detection of B-cell precursors, erythroblasts, myeloid precursors, and/or mast cells. This information is critical to ensure sample quality and to identify hemodiluted BM aspirates that may lead to false-negative results.

Table 1

Individual features of currently available techniques to monitor MRD in MM

MFC (≥8-color)ASO-PCRNGSPET/CT
Applicability ∼100% 60% to 70% ∼90% ∼100%* 
Reproducibility among centers High High Not reported Moderate at MRD 
Availability in individual laboratories around the world High Intermediate Limited Intermediate 
Diagnostic sample Important but not mandatory Mandatory Mandatory Important but not mandatory 
Time 2-3 h ≥5 d (follow-up), 3-4 wk (target identification) ≥7 d 2 h 
Cost per sample ∼350 USD ∼500 USD (follow-up), ∼1500 USD at diagnosis (target identification) ∼700 USD ∼2000 USD 
Sensitivity 10−5 to 10−6 10−5 to 10−6 10−6 High (4 mm) 
Quantitative Yes (directly; high accuracy) Yes Yes Yes 
Fresh sample Needed (<36 h) Not needed Not needed NA 
Patchy sample Impacts Impacts Impacts No impact 
Global cell characterization Yes No No No 
Standardization Ongoing (EuroFlow/IMF) Yes, since 15 y (EuroMRD) Not reported No 
MFC (≥8-color)ASO-PCRNGSPET/CT
Applicability ∼100% 60% to 70% ∼90% ∼100%* 
Reproducibility among centers High High Not reported Moderate at MRD 
Availability in individual laboratories around the world High Intermediate Limited Intermediate 
Diagnostic sample Important but not mandatory Mandatory Mandatory Important but not mandatory 
Time 2-3 h ≥5 d (follow-up), 3-4 wk (target identification) ≥7 d 2 h 
Cost per sample ∼350 USD ∼500 USD (follow-up), ∼1500 USD at diagnosis (target identification) ∼700 USD ∼2000 USD 
Sensitivity 10−5 to 10−6 10−5 to 10−6 10−6 High (4 mm) 
Quantitative Yes (directly; high accuracy) Yes Yes Yes 
Fresh sample Needed (<36 h) Not needed Not needed NA 
Patchy sample Impacts Impacts Impacts No impact 
Global cell characterization Yes No No No 
Standardization Ongoing (EuroFlow/IMF) Yes, since 15 y (EuroMRD) Not reported No 

EuroFlow, see www.EuroFlow.org; EuroMRD, see www.EuroMRD.org; IMF, International Myeloma Foundation; PET/CT, positron emission tomography/computed tomography; USD, US dollars. NA, not appropriate.

*

Specifically for extramedullary disease.

Costs calculated based on both reagent and personnel costs for a medium-size laboratory receiving ∼150 to 200 MRD samples per year.

Defined as minimal percentage of cells detectable within or out of the quantitative range of the method or in size for imaging techniques.

A potential limitation of MFC is that current strategies are designed to characterize the PC compartment and could therefore miss potential MM cancer stem cells with more immature phenotypes, such as postgerminal center memory B cells (Table 1).55  Nevertheless, recent investigations conducted with sensitive ASO-PCR assessment of clonal Ig heavy (IGH) myeloma sequences among fluorescence-activated cell sorter–sorted peripheral blood (PB) B-cell subsets, revealed that such clonotypic cells are either absent or present below highly sensitive limits of detection.56 

Highly sensitive MFC-based MRD monitoring (down to 10−5) requires the availability of ≥8-color digital flow cytometers coupled to novel sample preparation procedures that allow fast and cost-effective, routine evaluation of >5 million nucleated cells (for detailed protocols, see www.EuroFlow.org; Table 1). This contrasts with previous MFC studies that defined MRD as the presence of a discrete population of clonal PCs at the 0.01% (ie, 10−4) limit of detection.

The need for extensive expertise to analyze flow cytometric data, together with the lack of well-standardized flow-MRD methods, has been pointed out as the main drawback of MFC immunophenotyping (Table 1).13  Furthermore, conventional visualization of flow cytometric data in bivariate (two-dimensional) dot plots becomes increasingly complex with increasing numbers of parameters.57,58  In recent years, new multivariate computational tools and visualization plots (eg, principal component analysis and canonical analysis) have been developed and integrated into innovative software packages for improved multidimensional identification and classification of different clusters of cells coexisting in a sample. These tools together with the use of normal and malignant reference databases further pave the way for automated detection and tracking of aberrant cell populations that deviate from the normal/reactive phenotypic profiles.50,58  Such innovative flow-MRD strategies are currently being developed by the EuroFlow Consortium under the Black Swan Research Initiative promoted by the International Myeloma Foundation, and it is likely to become the method of choice for accurate, high-sensitive, and automated flow-MRD monitoring in MM.

ASO-PCR

Rearrangements of germ-line V, (D), and J gene segments in the Ig gene complexes (IGH, IGK, and IGL) provide each B cell with specific V(D)J combinations, which together code for the many different variable domains of Ig molecules. The random insertion and deletion of nucleotides at the V(D)J junction sites create highly diverse junctional regions, which represent unique “fingerprint-like” sequences that are most probably different in each B-cell and thus also in each B-cell malignancy. Since the 1990s, these junctional regions (to be identified in each individual patient at diagnosis) have therefore been used as individual tumor-specific targets using Ig ASOs as primers, initially for nested PCR approaches and later for real-time quantitative PCR-based MRD analysis (ASO-PCR). Such Ig targets can be identified and sequenced with standardized technologies in >95% of lymphoid malignancies and used for the design of junctional region–specific oligonucleotides to be applied for sensitive PCR-based detection of low frequencies of malignant cells, down to 1 malignant cell in 104 to 105 normal cells (10−4 to 10−5) (Table 1).59  This time-consuming but sensitive approach has been highly successful for MRD diagnostics in immature B-lineage malignancies, such as acute lymphoblastic leukemia,57,59  and has also been applied in mature B-cell malignancies, such as MM,60,61  reaching good sensitivities and demonstrating predictive value post–stem cell transplantation (SCT)62,63  and in myeloma patients with PR to induction therapy.48,64 

However, B-cells can further mature their Ig molecules via somatic hypermutation (SHM) of the functional V(D)J exons, resulting in high-affinity antibodies that are typically produced by postgerminal center memory B cells and PCs. The SHM process also occurs in and around the junctional regions, and therefore, the mutations can change the DNA sequence at the positions of the PCR primers that are used in the MRD studies. This explains why standard PCR primer sets cannot detect each individual IGH, IGK, and IGL gene rearrangement in mature postgerminal center B-cell malignancies.65,66  This is particularly valid for MM, which represents the most mature B-cell stage with heavily mutated Ig genes. To reduce the problem of false-negative results, multiple Ig genes have been targeted in parallel (eg, IGH and IGK), and unmutated Ig rearrangements have also been used as targets (eg, incomplete D-J and deletion of the IGK gene [IGK-Kde] rearrangements).66,67  Nevertheless, it remains difficult to apply the ASO-PCR approach in all MM cases (Table 1), unless other (nonclassical) methods will be used for Ig target detection and sequencing. For example, it might be possible to use Ig leader primers in combination with Constant gene primers at the RNA level to avoid the SHM-mutated sequences; such an approach has not been tested so far.

NGS of Ig genes

Several years ago, high-throughput sequencing (HTS) was introduced for studying the diversity of antigen receptor genes.68  For this purpose, multiplex primer sets65  are used to detect all potential rearrangements in a sample (up to 105 or more). As a logical consequence, HTS has been applied for detection of clonal Ig gene rearrangements, including detection of MRD,69-71  assuming that each rearrangement can be detected and that such detection of rearranged Ig genes is proportional (ie, reflecting their frequency in the original sample). However, comparable to ASO-PCR, HTS is also based on an initial PCR step, using primers that have to anneal to the Ig gene sequences. Logically, some primers are more efficient than others, and SHM in Ig genes further hampers primer annealing in mature B-cell malignancies.65,66  This explains why HTS was not able to detect a reliable Ig PCR target in all MM patients, even when multiple Ig loci (IGH Vh-Jh, Dh-Jh, and IGK) were evaluated.48,70  Another problem in HTS is the quantitation of MRD because the clonal rearrangement is detected between polyclonal Ig rearrangements derived from remaining normal B-cells, of which the frequency might be highly variable, depending on the type of treatment. Finally, whereas the classical ASO-PCR has been standardized and is subjected to frequent (every 6 months) international quality assurance rounds (www.EuroMRD.org), HTS has not (yet) been standardized and lacks quality assurance rounds (Table 1), the MRD HTS data reported so far in MM being restricted to a commercial service-based approach/tool.48,70,72 

Recently, NGS has also been evaluated in PB (ie, plasma) from 45 MM patients who received carfilzomid-lenalidomide-dexamethasone (CRD) induction.72  This would represent an attractive minimally invasive approach to overcome the challenge of a patchy BM infiltration. However, preliminary data indicate that clonotypic sequences identified at baseline become undetectable with just a few cycles of chemotherapy, even among electrophoresis-positive patients. Thus, further research is warranted to establish the feasibility of PB (eg, cell- or free DNA–based) MRD monitoring.

MRI and PET/CT

The possibility of patchy BM infiltration or extramedullary involvement with an MRD-negative BM is an additional challenge for both MFC- and PCR-based MRD detection in single BM aspirates. This highlights the value of sensitive imaging techniques to redefine CR among MRD-negative cases by MFC, ASO-PCR, and NGS, both at the intramedullary and extramedullary levels, whenever the subjective nature of the assessments and the concerns regarding reproducibility are overcome. Magnetic resonance imaging (MRI) is the most sensitive noninvasive imaging technique for detection of bone involvement in the spine. It also provides relevant information on the extent and nature of soft tissue disease and the pattern of marrow infiltration (normal, focal, heterogeneous, or diffuse). However, it should be noted that focal lesions may remain hyperintense for several months after therapy, in both responding and nonresponding patients, because of treatment-induced necrosis and inflammation. This can explain some inconsistencies found between serological CR and MRI-based CR.73,74  Consequently, an interval of 3 months has been recommended before MRI monitoring.75 

The use of PET/CT combines the imaging of a particular molecular process (eg, fluorodeoxyglucose uptake) with the morphologic images provided by CT data. However, it is important to emphasize that for MRD monitoring (which will pay particular attention to fluorodeoxyglucose uptake rather than lytic bone lesions), both false-negative and false-positive results (in the case of other coexisting infectious or inflammatory processes) may be seen.76 

In contrast to traditional imaging techniques, a specific advantage of PET/CT relies on its ability to detect extramedullary disease, which is a sign of spread of the disease outside the BM with an adverse prognostic impact.77  A recent comparison between PET/CT and whole body MRI in transplant-candidate patients showed that, against conventional response criteria, PET/CT had the same sensitivity but higher specificity than whole body MRI. Although the utility of other MRI-based techniques is still under investigation (eg, dynamic contrast-enhanced MRI),78  the current perception is that PET/CT would represent the most effective imaging tool to monitor MRD in MM. However, standardization of response definitions by PET/CT and comparison with other sensitive BM-based MRD methods, including targeted biopsies, is still needed to implement this imaging technique across different clinical studies (Table 1).79 

Transplant-eligible patients

Early studies exploring the role of MRD in MM typically used PCR-based methods in the setting of autologous or allogeneic SCT (allo-SCT) because at that time these were considered the only effective treatment approaches (Figure 3).60,63,80-82  With few exceptions,83  most studies concerned relatively small patient series reflecting the challenging nature of PCR-based MRD methods for routine testing (see “ASO-PCR”), but they demonstrated that PCR-MRD monitoring was of prognostic value.60,80-82  In parallel, 3- and 4-color flow-MRD methods were introduced83,84  and shown to be of prognostic value by San Miguel et al85  and Rawstron et al (Figure 3).86  Subsequent comparisons between PCR- and MFC-based MRD monitoring showed that, except for a few discordant cases, both techniques provided highly concordant results (Figure 3).60,64,87,88  The initial positive experience by the Spanish and United Kingdom groups led to the implementation of their corresponding 4- and 6-color MFC approaches in large clinical trials. In the Programa para el Estudio de la Terapéutica en Hemopatías Malignas/Grupo Español de MM (PETHEMA/GEM) 2000 study, flow MRD was identified as the most relevant prognostic factor in a series of 295 newly diagnosed MM patients receiving uniform treatment including HDT/SCT.89  In this trial, MRD negativity after ASCT translated to significantly improved PFS and OS rates. Similarly, in the intensive pathway of the Medical Research Council, UK (MRC) Myeloma IX study, MRD negativity after HDT/ASCT was predictive of favorable PFS and OS.90  More recently, these observations were reproduced using ASO-PCR64  and NGS tools,48  which again confirmed the prognostic value of MRD assessment in transplant-eligible MM patients. Furthermore, Zamagni et al reported that post-ASCT, PET/CT monitoring was also an independent prognostic marker for PFS and OS.91  Recently, similar results have been reported in the allo-SCT setting where the presence of MRD following allo-SCT has been associated with a significantly adverse PFS and OS (Figure 3).92-94 

Figure 3

Number of studies published in PUBMED per MRD technique showing prognostic value for progression-free survival (PFS) and overall survival (OS) specifically among patients in CR after therapy. Numbers refer to the literature cited in the present review.

Figure 3

Number of studies published in PUBMED per MRD technique showing prognostic value for progression-free survival (PFS) and overall survival (OS) specifically among patients in CR after therapy. Numbers refer to the literature cited in the present review.

Close modal

Importantly, all studies showed that PFS of MRD-negative patients at least doubled that of MRD-positive CR patients,40,48,64,88-91  reaching up to a striking 8-year difference in PFS among CR patients by their NGS-MRD status.48  Conversely, both MFC and ASO-PCR showed that CR patients with persistent MRD had significantly inferior OS vs MRD-negative cases.64,89,90  These results support the rationale for implementing MRD assessment to redefine and improve current CR criteria in MM.

Discordant results between MRD (by immunophenotypic, molecular, and imaging techniques) vs conventional response assessment has questioned the sensitivity and specificity of MRD monitoring over traditional paraprotein measurement. However, it has been shown that MRD-negative patients in near CR/PR have a favorable prognosis, which has been hypothesized to be because of the long M-protein half-lives that, in selected patients, could disappear over time43 ; other factors such as the phenomenon of continued response after HDT/ASCT without further therapy, as well as the impact of additional therapy, should also be considered. Irrespectively of all the above-mentioned factors, our most recent observations indicate that approximately two-thirds of MRD-negative cases in near CR/PR achieved CR in a median of 2 months (B.P., L. Rosiñol, M.B. Vidriales, M.A. Montalban, N.C. Gutierrez, M.L. Martín-Ramos, N. Puig, J. Martinez-Lopez, M.V. Mateos, L. Cordón, A. Oriol, M.J. Terol, M.A. Echeveste, J. De la Rubia, J.J. Lahuerta, J. Blade, and J.F. San Miguel, manuscript in preparation). Such observations also highlight the importance of the 2 consecutive protein response assessments before the institution of any new therapy to confirm a response category and also unravel how immunofixation alone may be suboptimal to evaluate the added value of sequential treatment strategies (eg, HDT/ASCT followed by consolidation and/or maintenance).

Elderly nontransplant candidate patients

The prognostic value of MRD assessment was not investigated outside of the SCT setting until recently, when the incorporation of novel agents into the treatment of patients who were not fit for HDT/ASCT showed increased CR rates and prolonged survival.3  Puig et al have recently demonstrated that among patients treated according to the PETHEMA/GEM2005MAS65 protocol, those in molecular CR after induction had a PFS not yet reached, whereas MRD-positive patients had a significantly shorter PFS (median 31 months; P = .03).64  Because MRD levels measured by ASO-PCR- and NGS-based approaches correlate well,70  when Martinez-Lopez studied young and elderly patients separately, the prognostic significance of achieving MRD negativity by deep-sequencing was equally observed.48  Regarding MFC, in the MRC myeloma IX protocol only a few patients achieved flow CR after induction regimens without proteasome inhibitors, and these showed nonsignificantly superior PFS.90  In contrast, in the PETHEMA/GEM2005MAS65 study patients were monitored after 6 induction cycles with bortezomib, melphalan, and prednisone or bortezomib, thalidomide, and prednisone, and, within a subset of 102 cases in CR/(very good) PR, 30% attained MRD negativity with PFS and OS rates at 3 years of 90% and 94%, respectively.41  A recent update of this study3  after a median follow-up >5 years shows median PFS and OS rates not yet reached for patients in flow CR after bortezomib, melphalan, and prednisone (but not bortezomib, thalidomide, and prednisone) induction. These results suggest that MRD monitoring is also clinically relevant in elderly patients. Because MRD-negative cases after 2 different regimens should experience similar outcomes,95  this study also unraveled that the 4-color MFC assay originally performed was underpowered for ultrasensitive detection of MRD.3  This has been recently confirmed by comparing deep-sequencing vs 4-color MFC-based MRD monitoring in younger and elderly MM patients,48  indicating that MRD prognostication is improved when more sensitive (ie, lower) limits of detection (ie, ≤1 tumor cell in 100 000 vs 10 000 normal cells; 10−5 vs 10−4) are reached.

Standard-risk vs high-risk cytogenetic patients

In MM, it has been suggested that attaining deep levels of remission (CR) could be critical only for patients with high-risk disease, whereas those with more indolent biology may not particularly benefit.14,15  However, after the PETHEMA/GEM reported that risk assessment by fluorescence in situ hybridization and flow-MRD monitoring were of independent prognostic value in transplant-eligible patients,89  Rawstron et al have reproduced and confirmed that the presence of MRD is a strong predictor of outcome in patients with both favorable and adverse cytogenetic profiles.90  In fact, the percentage of patients achieving MRD negativity was identical between standard- and high-risk cytogenetic patient subgroups (∼60%).90  Further analyses by the PETHEMA/GEM have shown that combined cytogenetic evaluation of PCs at diagnosis plus MRD assessment after HDT/ASCT (day +100) provided a powerful discriminator of outcomes, which also resulted in a highly effective approach to identify patients with unsustained CR and dismal outcomes (2-years median OS for cases with baseline high-risk cytogenetics plus persistent MRD).39  Collectively, these results confirm the superiority of MRD assessment over conventional response criteria to predict outcome in distinct MM genetic subgroups.

MRD and treatment schema

So far, no clinical trial has randomized MM patients according to their MRD status and, thereby, investigated the role of MRD for individualized therapy. However, many studies have shown the value of MRD diagnostics for evaluation of the efficacy of specific treatment stages and, therefore, potential treatment decisions (Figure 4). For example, both the Spanish and the United Kingdom study groups have shown that MRD kinetics before and after HDT/ASCT allow the identification of chemosensitive (MRD-negative cases at 2 time points), intermediate, and chemoresistant patients (MRD-positive patients at 2 time points).89,90  For the latter, it could be hypothesized that consolidation is needed to improve outcomes that, with maintenance alone, were significantly inferior vs the remaining cases.89,90  When such analysis is restricted to CR patients after induction, those failing to eradicate MRD levels before HDT/ASCT will show significantly superior PFS if MRD negativity is achieved after HDT/ASCT, and their outcome becomes superimposable to that of cases that were already MRD negative before HDT/ASCT (B.P., L. Rosiñol, M.B. Vidriales, M.A. Montalban, N.C. Gutierrez, M.L. Martín-Ramos, N. Puig, J. Martinez-Lopez, M.V. Mateos, L. Cordón, A. Oriol, M.J. Terol, M.A. Echeveste, J. De la Rubia, J.J. Lahuerta, J. Blade, and J.F. San Miguel, manuscript in preparation). These results suggest not only that MRD kinetics is more informative than single time-point assessments, but also that this information may be useful to address specific clinical questions (eg, early vs delayed HDT/ASCT for CR patients after induction).96 

Figure 4

MRD monitoring (ie, black solid line arrows) has been reported (numbers refer to the literature cited in the present review) to be prognostically informative among cytogenetically defined standard- and high-risk MM patients after induction chemotherapy, HDT/ASCT, and consolidation; during follow-up; and after salvage therapy.

Figure 4

MRD monitoring (ie, black solid line arrows) has been reported (numbers refer to the literature cited in the present review) to be prognostically informative among cytogenetically defined standard- and high-risk MM patients after induction chemotherapy, HDT/ASCT, and consolidation; during follow-up; and after salvage therapy.

Close modal

Maintenance therapy represents another illustrating example. In the Gruppo Italiano Malattie EMatologiche dell'Adulto (GIMEMA) VEL-03-096 study, Ladetto et al reported PFS rates at median follow-up of 100% vs 57% for patients in molecular-CR vs MRD-positive cases, respectively.97  In a recent update, these authors confirmed the significantly superior PFS and OS observed for patients attaining molecular CR.98  Because no maintenance therapy was given in this study, one might hypothesize that for those cases that failed to achieve MRD negativity despite being in CR/near CR after consolidation, maintenance could have potentially been an effective approach to eradicate MRD levels and improve outcome. In line with this hypothesis, Rawstron et al have shown that 1 out of 4 MRD-positive patients randomized to the maintenance arm of the intensive treatment pathway of the MRC-myeloma IX study turned into MRD negative and experienced significantly prolonged PFS vs the abstention arm.90 

Overall, the experience of several cooperative groups using different MRD techniques indicates that persistence of MRD is always an adverse prognostic feature, even among CR patients. Consequently, it would be safer to make clinical decisions based on MRD positivity rather than on MRD negativity because the patchy pattern of BM infiltration typically observed in MM leads to a degree of uncertainty regarding MRD-negative results (ie, are clonal PCs truly absent, or is it because of nonrepresentative BM sampling?). Some of these limitations could be potentially overcome in flow- and/or molecular-MRD-negative cases by parallel usage of sensitive imaging techniques, although these approaches may also give false-negative results.76,79,91  Thus, it may be envisioned that if treatment decisions are made according to patients’ MRD status, follow-up MRD studies would also become useful to detect MRD reappearance preceding clinical relapse.98 

Recently, Barlogie et al have shown that the vast majority of CR patients (94%) achieving long-term survival (10 years relapse free), were also MRD negative.7  By contrast, at least one-third of MM patients achieving CR after initial therapy will not experience a survival benefit because of persistent MRD. However, attaining deep remissions is not a prerequisite for some patients to achieve long-term disease control,7,30  and more accurate identification of such patients should also become a research priority (Figure 2).

MRD clearance is achievable in the era of novel and more effective treatment strategies and it is predictive of superior outcomes. Thus, MRD could potentially be used as a biomarker to evaluate the efficacy of treatment at different stages (induction, transplantation, consolidation, and/or maintenance; Figure 4) and as a surrogate for OS.

Because of their poor prognosis, 2 specific patient subgroups could be ideal to investigate the role of MRD monitoring as a clinical end point for novel treatment modalities and a surrogate biomarker for OS: patients with baseline high-risk cytogenetics and those with relapsed disease. Both patient subgroups reflect the unmet need for novel agents; at the same time, achieving MRD negativity has also resulted in superior outcome in both groups.40,89,90,99,100  The choice of MRD technology for monitoring will depend on how individual centers’ priorities adjust to the specific advantages that each tool has to offer, highly sensitive and automated flow MRD being particularly attractive in assessing BM response (Table 1). In turn, extensive research is still warranted to determine how to best integrate medullary and extramedullary MRD monitoring.

In other hematologic malignancies, baseline risk factors and MRD monitoring have an established and complementary role to individualized treatment. Over the past 2 decades, several groups have consistently confirmed the added value of MRD in MM. Therefore, now also in MM, the time has come to establish the role of baseline risk factors and MRD monitoring for tailored therapy. This requires the introduction of standardized, highly sensitive, cost-effective, and broadly available MRD techniques in all clinical trials.

This work was supported by grants from the Cooperative Research Thematic Network (RD12/0036/0058 and RD12/0036/0048 of the Red de Cancer [Cancer Network of Excellence] FEDER); Instituto de Salud Carlos III, Spain, Instituto de Salud Carlos III/Subdirección General de Investigación Sanitaria (FIS: PI060339, 06/1354, 02/0905, 01/0089/01-02, PS09/01897/01370, G03/136; Sara Borrell: CD13/00340); and Asociación Española Contra el Cáncer (GCB120981SAN), Spain. The study was also supported internationally by the International Myeloma Foundation Black Swan Research Initiative and Junior Grant Proposals and the Multiple Myeloma Research Foundation research fellow award.

Contribution: B.P., J.J.M.v.D., and A.O. wrote, reviewed, and approved the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Alberto Orfao, Cancer Research Center, Paseo de la Universidad de Coimbra s/n, 37007 Salamanca, Spain; e-mail: orfao@usal.es.

1
Ocio
 
EM
Richardson
 
PG
Rajkumar
 
SV
, et al. 
New drugs and novel mechanisms of action in multiple myeloma in 2013: a report from the International Myeloma Working Group (IMWG).
Leukemia
2014
, vol. 
28
 
3
(pg. 
525
-
542
)
2
Cavo
 
M
Pantani
 
L
Petrucci
 
MT
, et al. 
GIMEMA (Gruppo Italiano Malattie Ematologiche dell’Adulto) Italian Myeloma Network
Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma.
Blood
2012
, vol. 
120
 
1
(pg. 
9
-
19
)
3
Mateos
 
MV
Oriol
 
A
Martinez-López
 
J
, et al. 
GEM2005 trial update comparing VMP/VTP as induction in elderly multiple myeloma patients: do we still need alkylators?
Blood
2014
, vol. 
124
 
12
(pg. 
1887
-
1893
)
4
Palumbo
 
A
Cavallo
 
F
Gay
 
F
, et al. 
Autologous transplantation and maintenance therapy in multiple myeloma.
N Engl J Med
2014
, vol. 
371
 
10
(pg. 
895
-
905
)
5
Mellqvist
 
UH
Gimsing
 
P
Hjertner
 
O
, et al. 
Nordic Myeloma Study Group
Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial.
Blood
2013
, vol. 
121
 
23
(pg. 
4647
-
4654
)
6
Roussel
 
M
Lauwers-Cances
 
V
Robillard
 
N
, 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
, vol. 
32
 
25
(pg. 
2712
-
2717
)
7
Barlogie
 
B
Mitchell
 
A
van Rhee
 
F
Epstein
 
J
Morgan
 
GJ
Crowley
 
J
Curing myeloma at last: defining criteria and providing the evidence.
Blood
2014
, vol. 
124
 
20
(pg. 
3043
-
3051
)
8
Durie
 
BG
Harousseau
 
JL
Miguel
 
JS
, et al. 
International Myeloma Working Group
International uniform response criteria for multiple myeloma.
Leukemia
2006
, vol. 
20
 
9
(pg. 
1467
-
1473
)
9
Cavo
 
M
Rajkumar
 
SV
Palumbo
 
A
, et al. 
International Myeloma Working Group
International Myeloma Working Group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation.
Blood
2011
, vol. 
117
 
23
(pg. 
6063
-
6073
)
10
Bladé
 
J
Samson
 
D
Reece
 
D
, et al. 
Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant
Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation.
Br J Haematol
1998
, vol. 
102
 
5
(pg. 
1115
-
1123
)
11
Lonial
 
S
Gleason
 
C
Down to the bitter end.
Blood
2014
, vol. 
123
 
20
(pg. 
3061
-
3062
)
12
Landgren
 
O
Gormley
 
N
Turley
 
D
, et al. 
Flow cytometry detection of minimal residual disease in multiple myeloma: lessons learned at FDA-NCI roundtable symposium.
Am J Hematol
2014
, vol. 
89
 
12
(pg. 
1159
-
1160
)
13
Flanders
 
A
Stetler-Stevenson
 
M
Landgren
 
O
Minimal residual disease testing in multiple myeloma by flow cytometry: major heterogeneity.
Blood
2013
, vol. 
122
 
6
(pg. 
1088
-
1089
)
14
Kumar
 
SK
Rajkumar
 
SV
The current status of minimal residual disease assessment in myeloma.
Leukemia
2014
, vol. 
28
 
2
(pg. 
239
-
240
)
15
Lonial
 
S
Anderson
 
KC
Association of response endpoints with survival outcomes in multiple myeloma.
Leukemia
2014
, vol. 
28
 
2
(pg. 
258
-
268
)
16
Gay
 
F
Larocca
 
A
Wijermans
 
P
, 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
, vol. 
117
 
11
(pg. 
3025
-
3031
)
17
Lahuerta
 
JJ
Mateos
 
MV
Martínez-López
 
J
, 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
, vol. 
26
 
35
(pg. 
5775
-
5782
)
18
van de Velde
 
HJ
Liu
 
X
Chen
 
G
Cakana
 
A
Deraedt
 
W
Bayssas
 
M
Complete response correlates with long-term survival and progression-free survival in high-dose therapy in multiple myeloma.
Haematologica
2007
, vol. 
92
 
10
(pg. 
1399
-
1406
)
19
Rosiñol
 
L
Oriol
 
A
Teruel
 
AI
, et al. 
Programa para el Estudio y la Terapéutica de las Hemopatías Malignas/Grupo Español de Mieloma (PETHEMA/GEM) group
Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study.
Blood
2012
, vol. 
120
 
8
(pg. 
1589
-
1596
)
20
Barlogie
 
B
Anaissie
 
E
Haessler
 
J
, et al. 
Complete remission sustained 3 years from treatment initiation is a powerful surrogate for extended survival in multiple myeloma.
Cancer
2008
, vol. 
113
 
2
(pg. 
355
-
359
)
21
Sonneveld
 
P
Goldschmidt
 
H
Rosiñol
 
L
, et al. 
Bortezomib-based versus nonbortezomib-based induction treatment before autologous stem-cell transplantation in patients with previously untreated multiple myeloma: a meta-analysis of phase III randomized, controlled trials.
J Clin Oncol
2013
, vol. 
31
 
26
(pg. 
3279
-
3287
)
22
Cavo
 
M
Tacchetti
 
P
Patriarca
 
F
, et al. 
GIMEMA Italian Myeloma Network
Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study.
Lancet
2010
, vol. 
376
 
9758
(pg. 
2075
-
2085
)
23
Jakubowiak
 
AJ
Griffith
 
KA
Reece
 
DE
, et al. 
Lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone in newly diagnosed multiple myeloma: a phase 1/2 Multiple Myeloma Research Consortium trial.
Blood
2011
, vol. 
118
 
3
(pg. 
535
-
543
)
24
Richardson
 
PG
Weller
 
E
Lonial
 
S
, et al. 
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma.
Blood
2010
, vol. 
116
 
5
(pg. 
679
-
686
)
25
Palumbo
 
A
Bringhen
 
S
Larocca
 
A
, et al. 
Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: updated follow-up and improved survival.
J Clin Oncol
2014
, vol. 
32
 
7
(pg. 
634
-
640
)
26
Usmani
 
SZ
Crowley
 
J
Hoering
 
A
, et al. 
Improvement in long-term outcomes with successive Total Therapy trials for multiple myeloma: are patients now being cured?
Leukemia
2013
, vol. 
27
 
1
(pg. 
226
-
232
)
27
Nooka
 
AK
Kaufman
 
JL
Muppidi
 
S
, et al. 
Consolidation and maintenance therapy with lenalidomide, bortezomib and dexamethasone (RVD) in high-risk myeloma patients.
Leukemia
2014
, vol. 
28
 
3
(pg. 
690
-
693
)
28
Stewart
 
AK
Rajkumar
 
SV
Dimopoulos
 
MA
, et al. 
ASPIRE Investigators
Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma.
N Engl J Med
2015
, vol. 
372
 
2
(pg. 
142
-
152
)
29
Harousseau
 
JL
Dimopoulos
 
MA
Wang
 
M
, et al. 
Better quality of response to lenalidomide plus dexamethasone is associated with improved clinical outcomes in patients with relapsed or refractory multiple myeloma.
Haematologica
2010
, vol. 
95
 
10
(pg. 
1738
-
1744
)
30
Martinez-Lopez
 
J
Blade
 
J
Mateos
 
MV
, et al. 
Grupo Español de MM; Programa para el Estudio de la Terapé utica en Hemopatía Maligna
Long-term prognostic significance of response in multiple myeloma after stem cell transplantation.
Blood
2011
, vol. 
118
 
3
(pg. 
529
-
534
)
31
Zhan
 
F
Barlogie
 
B
Arzoumanian
 
V
, et al. 
Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis.
Blood
2007
, vol. 
109
 
4
(pg. 
1692
-
1700
)
32
Kastritis
 
E
Zagouri
 
F
Symeonidis
 
A
, et al. 
Greek Myeloma Study Group
Preserved levels of uninvolved immunoglobulins are independently associated with favorable outcome in patients with symptomatic multiple myeloma.
Leukemia
2014
, vol. 
28
 
10
(pg. 
2075
-
2079
)
33
Paiva
 
B
Vidriales
 
MB
Mateo
 
G
, et al. 
GEM (Grupo Español de MM)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Groups
The persistence of immunophenotypically normal residual bone marrow plasma cells at diagnosis identifies a good prognostic subgroup of symptomatic multiple myeloma patients.
Blood
2009
, vol. 
114
 
20
(pg. 
4369
-
4372
)
34
Paiva
 
B
Vídriales
 
MB
Rosiñol
 
L
, et al. 
Grupo Español de MM/Programa para el Estudio de la Terapéutica en Hemopatías Malignas Cooperative Study Group
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
, vol. 
27
 
10
(pg. 
2056
-
2061
)
35
Dimopoulos
 
M
Spencer
 
A
Attal
 
M
, et al. 
Multiple Myeloma (010) Study Investigators
Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma.
N Engl J Med
2007
, vol. 
357
 
21
(pg. 
2123
-
2132
)
36
Richardson
 
PG
Sonneveld
 
P
Schuster
 
MW
, et al. 
Assessment of Proteasome Inhibition for Extending Remissions (APEX) Investigators
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma.
N Engl J Med
2005
, vol. 
352
 
24
(pg. 
2487
-
2498
)
37
Facon
 
T
Mary
 
JY
Hulin
 
C
, et al. 
Intergroupe Francophone du Myélome
Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial.
Lancet
2007
, vol. 
370
 
9594
(pg. 
1209
-
1218
)
38
San Miguel
 
JF
Schlag
 
R
Khuageva
 
NK
, et al. 
VISTA Trial Investigators
Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma.
N Engl J Med
2008
, vol. 
359
 
9
(pg. 
906
-
917
)
39
Kapoor
 
P
Kumar
 
SK
Dispenzieri
 
A
, et al. 
Importance of achieving stringent complete response after autologous stem-cell transplantation in multiple myeloma.
J Clin Oncol
2013
, vol. 
31
 
36
(pg. 
4529
-
4535
)
40
Paiva
 
B
Gutiérrez
 
NC
Rosiñol
 
L
, et al. 
PETHEMA/GEM (Programa para el Estudio de la Terapéutica en Hemopatías Malignas/Grupo Español de Mieloma) Cooperative Study Groups
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
, vol. 
119
 
3
(pg. 
687
-
691
)
41
de Larrea
 
CF
Cibeira
 
MT
Elena
 
M
, et al. 
Abnormal serum free light chain ratio in patients with multiple myeloma in complete remission has strong association with the presence of oligoclonal bands: implications for stringent complete remission definition.
Blood
2009
, vol. 
114
 
24
(pg. 
4954
-
4956
)
42
Giarin
 
MM
Giaccone
 
L
Sorasio
 
R
, et al. 
Serum free light chain ratio, total kappa/lambda ratio, and immunofixation results are not prognostic factors after stem cell transplantation for newly diagnosed multiple myeloma.
Clin Chem
2009
, vol. 
55
 
8
(pg. 
1510
-
1516
)
43
Paiva
 
B
Martinez-Lopez
 
J
Vidriales
 
MB
, et al. 
Comparison of immunofixation, serum free light chain, and immunophenotyping for response evaluation and prognostication in multiple myeloma.
J Clin Oncol
2011
, vol. 
29
 
12
(pg. 
1627
-
1633
)
44
San-Miguel
 
JF
Paiva
 
B
Gutiérrez
 
NC
New tools for diagnosis and monitoring of multiple myeloma.
Am Soc Clin Oncol Educ Book
2013
45
Ludwig
 
H
Milosavljevic
 
D
Zojer
 
N
, et al. 
Immunoglobulin heavy/light chain ratios improve paraprotein detection and monitoring, identify residual disease and correlate with survival in multiple myeloma patients.
Leukemia
2013
, vol. 
27
 
1
(pg. 
213
-
219
)
46
Tovar
 
N
de Larrea
 
CF
Aróstegui
 
JI
, et al. 
Natural history and prognostic impact of oligoclonal humoral response in patients with multiple myeloma after autologous stem cell transplantation: long-term results from a single institution.
Haematologica
2013
, vol. 
98
 
7
(pg. 
1142
-
1146
)
47
Rawstron
 
AC
Gregory
 
WM
de Tute
 
RM
, et al. 
Minimal residual disease (MRD) in myeloma by flow cytometry: independent prediction of survival benefit per log reduction.
Blood
2015
, vol. 
125
 
12
(pg. 
1932
-
1935
)
48
Martinez-Lopez
 
J
Lahuerta
 
JJ
Pepin
 
F
, et al. 
Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma.
Blood
2014
, vol. 
123
 
20
(pg. 
3073
-
3079
)
49
Paiva
 
B
Almeida
 
J
Pérez-Andrés
 
M
, et al. 
Utility of flow cytometry immunophenotyping in multiple myeloma and other clonal plasma cell-related disorders.
Cytometry B Clin Cytom
2010
, vol. 
78
 
4
(pg. 
239
-
252
)
50
van Dongen
 
JJ
Lhermitte
 
L
Böttcher
 
S
, et al. 
EuroFlow Consortium (EU-FP6, LSHB-CT-2006-018708)
EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes.
Leukemia
2012
, vol. 
26
 
9
(pg. 
1908
-
1975
)
51
Rawstron
 
AC
Orfao
 
A
Beksac
 
M
, et al. 
European Myeloma Network
Report of the European Myeloma Network on multiparametric flow cytometry in multiple myeloma and related disorders.
Haematologica
2008
, vol. 
93
 
3
(pg. 
431
-
438
)
52
Mei
 
HE
Yoshida
 
T
Sime
 
W
, et al. 
Blood-borne human plasma cells in steady state are derived from mucosal immune responses.
Blood
2009
, vol. 
113
 
11
(pg. 
2461
-
2469
)
53
Caraux
 
A
Klein
 
B
Paiva
 
B
, et al. 
Myeloma Stem Cell Network
Circulating human B and plasma cells. Age-associated changes in counts and detailed characterization of circulating normal CD138- and CD138+ plasma cells.
Haematologica
2010
, vol. 
95
 
6
(pg. 
1016
-
1020
)
54
Paíno
 
T
Paiva
 
B
Sayagués
 
JM
, et al. 
Phenotypic identification of subclones in multiple myeloma with different chemoresistant, cytogenetic and clonogenic potential [published online ahead of print November 12, 2014].
Leukemia
55
Leung-Hagesteijn
 
C
Erdmann
 
N
Cheung
 
G
, et al. 
Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma.
Cancer Cell
2013
, vol. 
24
 
3
(pg. 
289
-
304
)
56
Thiago
 
L
Perez-Andres
 
M
Balanzategui
 
A
, et al. 
Circulating clonotypic B cells in multiple myeloma and monoclonal gammopathy of undetermined significance.
Haematologica
2014
, vol. 
99
 
1
(pg. 
155
-
162
)
57
Szczepariski
 
T
Orfão
 
A
van der Velden
 
VH
San Miguel
 
JF
van Dongen
 
JJ
Minimal residual disease in leukaemia patients.
Lancet Oncol
2001
, vol. 
2
 
7
(pg. 
409
-
417
)
58
Costa
 
ES
Pedreira
 
CE
Barrena
 
S
, 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
, vol. 
24
 
11
(pg. 
1927
-
1933
)
59
van der Velden
 
VH
Panzer-Grümayer
 
ER
Cazzaniga
 
G
, et al. 
Optimization of PCR-based minimal residual disease diagnostics for childhood acute lymphoblastic leukemia in a multi-center setting.
Leukemia
2007
, vol. 
21
 
4
(pg. 
706
-
713
)
60
Sarasquete
 
ME
García-Sanz
 
R
González
 
D
, et al. 
Minimal residual disease monitoring in multiple myeloma: a comparison between allelic-specific oligonucleotide real-time quantitative polymerase chain reaction and flow cytometry.
Haematologica
2005
, vol. 
90
 
10
(pg. 
1365
-
1372
)
61
Ladetto
 
M
Dinovan
 
JW
Haring
 
S
, et al. 
Real-Time polymerase chain reaction of immunoglobulin rearrangements for quantitative evaluation of minimal residual disease in multiple myeloma.
Biol Blood Marrow Transplant.
2000
, vol. 
6
 
3
(pg. 
241
-
253
)
62
Bakkus
 
MH
Bouko
 
Y
Samson
 
D
, et al. 
Post-transplantation tumour load in bone marrow, as assessed by quantitative ASO-PCR, is a prognostic parameter in multiple myeloma.
Br J Haematol
2004
, vol. 
126
 
5
(pg. 
665
-
674
)
63
Galimberti
 
S
Benedetti
 
E
Morabito
 
F
, et al. 
Prognostic role of minimal residual disease in multiple myeloma patients after non-myeloablative allogeneic transplantation.
Leuk Res
2005
, vol. 
29
 
8
(pg. 
961
-
966
)
64
Puig
 
N
Sarasquete
 
ME
Balanzategui
 
A
, et al. 
Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myeloma. A comparative analysis with flow cytometry.
Leukemia
2014
, vol. 
28
 
2
(pg. 
391
-
397
)
65
van Dongen
 
JJ
Langerak
 
AW
Brüggemann
 
M
, et al. 
Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2257
-
2317
)
66
Evans
 
PA
Pott
 
C
Groenen
 
PJ
, et al. 
Significantly improved PCR-based clonality testing in B-cell malignancies by use of multiple immunoglobulin gene targets. Report of the BIOMED-2 Concerted Action BHM4-CT98-3936.
Leukemia
2007
, vol. 
21
 
2
(pg. 
207
-
214
)
67
Puig
 
N
Sarasquete
 
ME
Alcoceba
 
M
, et al. 
Kappa deleting element as an alternative molecular target for minimal residual disease assessment by real-time quantitative PCR in patients with multiple myeloma.
Eur J Haematol
2012
, vol. 
89
 
4
(pg. 
328
-
335
)
68
Boyd
 
SD
Marshall
 
EL
Merker
 
JD
, et al. 
Measurement and clinical monitoring of human lymphocyte clonality by massively parallel VDJ pyrosequencing.
Sci Transl Med
2009
, vol. 
1
 
12
pg. 
12ra23
 
69
Logan
 
AC
Gao
 
H
Wang
 
C
, et al. 
High-throughput VDJ sequencing for quantification of minimal residual disease in chronic lymphocytic leukemia and immune reconstitution assessment.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
52
(pg. 
21194
-
21199
)
70
Ladetto
 
M
Brüggemann
 
M
Monitillo
 
L
, et al. 
Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1299
-
1307
)
71
Faham
 
M
Zheng
 
J
Moorhead
 
M
, et al. 
Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
26
(pg. 
5173
-
5180
)
72
Korde
 
N
Mailankody
 
S
Roschewski
 
M
, et al. 
Minimal residual disease (MRD) testing in newly diagnosed multiple myeloma (MM) patients: a prospective head-to-head assessment of cell-based, molecular, and molecular-imaging modalities [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 2105
73
Hillengass
 
J
Ayyaz
 
S
Kilk
 
K
, et al. 
Changes in magnetic resonance imaging before and after autologous stem cell transplantation correlate with response and survival in multiple myeloma.
Haematologica
2012
, vol. 
97
 
11
(pg. 
1757
-
1760
)
74
Bartel
 
TB
Haessler
 
J
Brown
 
TL
, et al. 
F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma.
Blood
2009
, vol. 
114
 
10
(pg. 
2068
-
2076
)
75
Zamagni
 
E
Cavo
 
M
The role of imaging techniques in the management of multiple myeloma.
Br J Haematol
2012
, vol. 
159
 
5
(pg. 
499
-
513
)
76
Caers
 
J
Withofs
 
N
Hillengass
 
J
, et al. 
The role of positron emission tomography-computed tomography and magnetic resonance imaging in diagnosis and follow up of multiple myeloma.
Haematologica
2014
, vol. 
99
 
4
(pg. 
629
-
637
)
77
Bladé
 
J
Fernández de Larrea
 
C
Rosiñol
 
L
Cibeira
 
MT
Jiménez
 
R
Powles
 
R
Soft-tissue plasmacytomas in multiple myeloma: incidence, mechanisms of extramedullary spread, and treatment approach.
J Clin Oncol
2011
, vol. 
29
 
28
(pg. 
3805
-
3812
)
78
Hillengass
 
J
Bäuerle
 
T
Bartl
 
R
, et al. 
Diffusion-weighted imaging for non-invasive and quantitative monitoring of bone marrow infiltration in patients with monoclonal plasma cell disease: a comparative study with histology.
Br J Haematol
2011
, vol. 
153
 
6
(pg. 
721
-
728
)
79
Moreau
 
P
PET-CT in MM: a new definition of CR.
Blood
2011
, vol. 
118
 
23
(pg. 
5984
-
5985
)
80
Cavo
 
M
Terragna
 
C
Martinelli
 
G
, et al. 
Molecular monitoring of minimal residual disease in patients in long-term complete remission after allogeneic stem cell transplantation for multiple myeloma.
Blood
2000
, vol. 
96
 
1
(pg. 
355
-
357
)
81
Fenk
 
R
Ak
 
M
Kobbe
 
G
, et al. 
Levels of minimal residual disease detected by quantitative molecular monitoring herald relapse in patients with multiple myeloma.
Haematologica
2004
, vol. 
89
 
5
(pg. 
557
-
566
)
82
Martinelli
 
G
Terragna
 
C
Zamagni
 
E
, et al. 
Molecular remission after allogeneic or autologous transplantation of hematopoietic stem cells for multiple myeloma.
J Clin Oncol
2000
, vol. 
18
 
11
(pg. 
2273
-
2281
)
83
Corradini
 
P
Cavo
 
M
Lokhorst
 
H
, et al. 
Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT)
Molecular remission after myeloablative allogeneic stem cell transplantation predicts a better relapse-free survival in patients with multiple myeloma.
Blood
2003
, vol. 
102
 
5
(pg. 
1927
-
1929
)
84
Almeida
 
J
Orfao
 
A
Ocqueteau
 
M
, et al. 
High-sensitive immunophenotyping and DNA ploidy studies for the investigation of minimal residual disease in multiple myeloma.
Br J Haematol
1999
, vol. 
107
 
1
(pg. 
121
-
131
)
85
San Miguel
 
JF
Almeida
 
J
Mateo
 
G
, 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
, vol. 
99
 
5
(pg. 
1853
-
1856
)
86
Rawstron
 
AC
Davies
 
FE
DasGupta
 
R
, et al. 
Flow cytometric disease monitoring in multiple myeloma: the relationship between normal and neoplastic plasma cells predicts outcome after transplantation.
Blood
2002
, vol. 
100
 
9
(pg. 
3095
-
3100
)
87
Lioznov
 
M
Badbaran
 
A
Fehse
 
B
Bacher
 
U
Zander
 
AR
Kröger
 
NM
Monitoring of minimal residual disease in multiple myeloma after allo-SCT: flow cytometry vs PCR-based techniques.
Bone Marrow Transplant
2008
, vol. 
41
 
10
(pg. 
913
-
916
)
88
Silvennoinen
 
R
Lundan
 
T
Kairisto
 
V
, et al. 
Comparative analysis of minimal residual disease detection by multiparameter flow cytometry and enhanced ASO RQ-PCR in multiple myeloma.
Blood Cancer J
2014
, vol. 
4
 
10
pg. 
e250
 
89
Paiva
 
B
Vidriales
 
MB
Cerveró
 
J
, et al. 
GEM (Grupo Español de MM)/PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas) Cooperative Study Groups
Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation.
Blood
2008
, vol. 
112
 
10
(pg. 
4017
-
4023
)
90
Rawstron
 
AC
Child
 
JA
de Tute
 
RM
, et al. 
Minimal residual disease assessed by multiparameter flow cytometry in multiple myeloma: impact on outcome in the Medical Research Council Myeloma IX Study.
J Clin Oncol
2013
, vol. 
31
 
20
(pg. 
2540
-
2547
)
91
Zamagni
 
E
Patriarca
 
F
Nanni
 
C
, et al. 
Prognostic relevance of 18-F FDG PET/CT in newly diagnosed multiple myeloma patients treated with up-front autologous transplantation.
Blood
2011
, vol. 
118
 
23
(pg. 
5989
-
5995
)
92
Giaccone
 
L
Brunello
 
L
Passera
 
R
, et al. 
Immunophenotypic response after allografting in multiple myeloma [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3371
93
Bruno
 
B
Ferrero
 
S
Drandi
 
D
, et al. 
Prospective molecular monitoring of minimal residual disease after non-myeloablative allografting in newly diagnosed multiple myeloma [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 44
94
Putkonen
 
M
Kairisto
 
V
Juvonen
 
V
, et al. 
Depth of response assessed by quantitative ASO-PCR predicts the outcome after stem cell transplantation in multiple myeloma.
Eur J Haematol
2010
, vol. 
85
 
5
(pg. 
416
-
423
)
95
Böttcher
 
S
Ritgen
 
M
Fischer
 
K
, et al. 
Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial.
J Clin Oncol
2012
, vol. 
30
 
9
(pg. 
980
-
988
)
96
van Rhee
 
F
Giralt
 
S
Barlogie
 
B
The future of autologous stem cell transplantation in myeloma.
Blood
2014
, vol. 
124
 
3
(pg. 
328
-
333
)
97
Ladetto
 
M
Pagliano
 
G
Ferrero
 
S
, et al. 
Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma.
J Clin Oncol
2010
, vol. 
28
 
12
(pg. 
2077
-
2084
)
98
Ferrero
 
S
Ladetto
 
M
Drandi
 
D
, 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
, vol. 
29
 
3
(pg. 
689
-
695
)
99
Paiva
 
B
Chandia
 
M
Puig
 
N
, et al. 
The prognostic value of multiparameter flow cytometry minimal residual disease assessment in relapse multiple myeloma.
Haematologica
2015
, vol. 
100
 
2
(pg. 
e53
-
e55
)
100
Ashcroft
 
J
de Tute
 
RM
Cairns
 
DA
, et al. 
The utility of minimal residual disease (MRD) assessment at first relapse: results from the BSBMT/Ukmf Myeloma X (intensive) trial [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 3378
Sign in via your Institution