Next Article in Journal
The Molecular Basis of Pediatric Brain Tumors: A Review with Clinical Implications
Previous Article in Journal
Computer-Aided Detection (CADe) of Small Metastatic Prostate Cancer Lesions on 3D PSMA PET Volumes Using Multi-Angle Maximum Intensity Projections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients

1
Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
2
Department of Clinical Chemistry, Inselspital, University of Bern, CH-3010 Bern, Switzerland
3
Department for Biomedical Research (DBMR), University of Bern, CH-3008 Bern, Switzerland
4
Department of Hematology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
5
Center of Laboratory Medicine (ZLM), Inselspital, University of Bern, CH-3010 Bern, Switzerland
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1565; https://doi.org/10.3390/cancers17091565
Submission received: 17 March 2025 / Revised: 24 April 2025 / Accepted: 28 April 2025 / Published: 4 May 2025

Simple Summary

The impact of minimal residual disease (MRD) post-treatment consolidation with high-dose chemotherapy and autologous stem cell transplantation (ASCT) on patient outcomes in high- and ultra-high-risk multiple myeloma (MM) remains poorly characterized. We investigated this by analyzing outcomes from our single-center patient cohort and showed that MRD status post-ASCT fails to predict longer versus shorter progression-free survival in MM patients with high- or ultra-high-risk cytogenetics. Our results suggest that a single MRD assessment might be insufficient in this patient subgroup, which could benefit from a closer and dynamic MRD monitoring.

Abstract

Background: Cytogenetic abnormalities and the persistence of minimal residual disease (MRD) following autologous stem cell transplantation (ASCT) are two established prognostically unfavorable biomarkers in multiple myeloma (MM). Previous studies have shown that post-transplant MRD status is a powerful predictor of progression-free survival (PFS) and overall survival (OS). However, the impact of MRD remains poorly characterized in MM patients with high- or ultra-high-risk cytogenetics. Objectives: This study investigated the prognostic value of post-transplant MRD in standard- versus high- and ultra-high-risk MM. To this aim, we performed a retrospective analysis of 137 MM patients who underwent high-dose chemotherapy (HDCT) and ASCT at our institution between January 2019 and December 2021. Cytogenetics were assessed by fluorescence in situ hybridization. High-risk genomic alterations included del(17p), t(4;14), t(14;16), t(14;20), gain(1q), and TP53 mutations, with two or more alterations defining the ultra-high-risk category. MRD was assessed in bone marrow aspirates post-ASCT using flow cytometry. Results: Eighty-two (60%) patients were categorized as being at standard risk, forty (29%) as high risk, and fifteen (11%) as ultra-high risk. Median follow-up was 47 months. MRD negativity was achieved in 76 (55%) patients. At 48 months, the overall PFS rate was 61% (72%, 50%, and 32% for the standard-, high-, and ultra-high-risk subgroups, respectively; p = 0.0004), while the OS rate was 85% (89%, 79%, and 80% in standard-, high-, and ultra-high-risk MM patients, respectively; p = 0.1494). Within the standard-risk subgroup, longer PFS was observed for patients achieving MRD negativity (p = 0.0172). High- and ultra-high-risk patients showed no significant differences in PFS when stratified by MRD status, possibly due to prompt progression to MRD positivity. Conclusions: Our results suggest that high- and ultra-high-risk MM patients might benefit from closer response monitoring, including dynamic MRD assessment. Further, high- and ultra-high-risk patients might require a more intensive peri-transplant treatment.

1. Introduction

Multiple myeloma (MM) is a malignant disease defined by the monoclonal expansion of plasma cells in the bone marrow. The incidence of MM displays geographical variations, but has increased globally since 1990 [1,2]. The integration of novel drugs into the treatment landscape of MM, including proteasome inhibitors, immunomodulatory drugs, and CD38-targeting antibodies, as well as, more recently CAR-T-cell therapy in later treatment lines, has resulted in a sustained increase in life expectancy in patients with MM [1].
The use of high-dose chemotherapy (HDCT) and autologous stem cell transplantation (ASCT) leads to significantly prolonged progression-free survival (PFS) following first-line induction therapy [3]. However, the majority of patients still relapse, and disease progression or treatment-related complications will ultimately lead to death [1].
The prognosis of MM patients has been shown to be influenced by several factors, including the characteristics of tumor cells, such as cytogenetic abnormalities and gene expression profile, in addition to extramedullary growth or lactate dehydrogenase levels [1]. There are notable differences between the risk stratification systems used, which differ not only in the serum markers included, but also in the cytogenetic genetic aberrations that are classified as high risk [4]. In accordance with the Revised International Staging System (R-ISS) by the International Myeloma Working Group (IMWG), the aberrations t(4;14), t(14;16) and del(17p) are defined as high risk [5]. The Mayo Clinic mSMART risk stratification system also considers t(14;20) and gain(1q) to be high-risk cytogenetics [6]. In recent years, the concept of ultra-high-risk MM has been introduced in cases where two or more high-risk aberrations are present. This designation, referred to as double-hit myeloma, is associated with a particularly poor prognosis in terms of progression-free survival (PFS) and overall survival (OS), when compared to that observed in single-hit multiple myeloma [6,7].
The assessment of minimal residual disease (MRD) has been demonstrated to be a powerful prognostic factor both for PFS and OS in multiple myeloma [8,9]. Several methods are used to determine MRD status, including multiparameter flow cytometry and next-generation sequencing (NGS). Regardless of the method used, various studies have indicated that the PFS of MM patients in complete remission with MRD negativity is markedly superior to that of MM patients in complete remission but with MRD positivity [10,11,12,13,14]. The impact of MRD status and depth of response on survival remains poorly defined in MM with high-risk or even ultra-high-risk cytogenetics [9]. This may be due to the fact that cytogenetic high-risk patients make up only 15–20% of the study population in large clinical trials [15].
Some studies have indicated that MRD positivity and high-risk cytogenetics are independent negative prognostic factors [16,17]. A meta-analysis illustrated that patients at standard risk with undetectable MRD have the best OS of all MM patients. Conversely, MRD-positive patients with high-risk cytogenetics demonstrate the most unfavorable prognoses [18]. In a previous study, Hu et al. observed that achieving MRD negativity also resulted in improved PFS and OS in high-risk patients [19]. Another study indicates that, despite achieving MRD negativity, PFS remains significantly inferior in high-risk patients compared to standard-risk patients [20]. Li et al. found no significant difference in PFS and OS between patients with high-risk disease who were MRD-negative and those with standard-risk disease who were MRD-positive [17]. Furthermore, Paiva et al. reported that the achievement of MRD negativity may potentially overcome high-risk cytogenetics [21]. Despite the concept of ultra-high-risk MM gaining increasing recognition in recent years, there have been only a few studies exploring the impact of MRD status on this particular group of individuals. In the MASTER phase 2 trial, transplant-eligible patients were treated with quadruplet induction therapy with daratumumab, carfilzomib, lenalidomide, and dexamethasone (Dara-KRd), followed by HDCT/ASCT, MRD-driven Dara-KRd consolidation, and lenalidomide maintenance if needed. Treatment-free monitoring was initiated in patients who had two consecutive negative MRD test results. The study demonstrated a significantly shortened PFS in those with ultra-high-risk cytogenetics compared to those with standard- or high-risk genetics [7].
The aim of this study is to investigate whether achieving MRD negativity should be the goal for all myeloma patients, or whether this aim could be waived for certain cytogenetic risk groups. A statistical analysis should help to facilitate a more comprehensive understanding of the prognostic significance of MRD negativity, considering the specific cytogenetic risk constellation. This approach may help optimize treatment goals for different patient subgroups, with the potential to enhance outcomes and avoid potential overtreatment.

2. Results

2.1. Patient Characteristics

A total of 137 patients were included in the study. The basal patient characteristics are outlined in Table 1. Of the total cohort, 82 patients (60%) were classified as being at standard risk, while 40 patients (29%) were identified as high risk, and 15 patients (11%) as ultra-high risk. The following genetic alterations were catalogued as high risk: del(17p), t(4;14), t(14;16), t(14;20), and gain(1q), as well as the presence of TP53 mutations. The presence of two or more of these cytogenetic alterations was interpreted as ultra-high risk. Conversely, if none of the previously mentioned mutations were present, the patients were classified as having standard-risk genetics. An overview of the abnormalities detected is given in Appendix A Table A1, and a comprehensive list of the karyotype data can be found in Appendix A Table A2. The median age was 62 years. The majority of patients were male (67%) and were in stage II according to R-ISS (49%) at the time of diagnosis. A significant difference between the groups was observed in the proportion of patients who were in stage III according to R-ISS (standard risk 12% vs. high risk 33% vs. ultra-high risk 60%; p < 0.0001). The immunoglobulin IgG was the most prevalent (66%), while IgA was less common (17%). However, the proportion of IgA present between the groups varied significantly (standard risk 11% vs. high risk 20%, ultra-high risk 40%; p = 0.0177). IgM could only be detected in one patient. Significant differences between the risk groups were also found in the presence of light chains. Lambda light chains were detected most frequently in ultra-high-risk patients (standard risk 22% vs. high risk 33% vs. ultra-high risk 80%; p < 0.0001). In contrast, kappa light chains were identified in 76% of standard-risk, 68% of high-risk, and only 20% of ultra-high-risk patients (p = 0.0001). Anemia was present in 18% of patients, and was most prevalent among high- (33%) and ultra-high-risk (27%) patients, compared to 10% among patients with standard-risk cytogenetics (p = 0.0063).

2.2. Course of Treatment

All 137 patients received induction therapy prior to HDCT/ASCT. The regimens utilized are outlined in Appendix A Table A3. The most frequently used combination (86%) was bortezomib, lenalidomide, and dexamethasone (VRd). In preparation for stem cell apheresis, mobilization was typically achieved through the administration of a chemotherapeutic agent, such as vinorelbine or gemcitabine, in conjunction with G-CSF (58%). A further 30% of patients were mobilized with G-CSF alone, and 10% received the proteasome inhibitor ixazomib in addition to G-CSF. High-dose chemotherapy was mainly administered using treosulfan and melphalan (48%). Melphalan alone was utilized in 31% of cases, while a combination of bendamustine and melphalan was employed in the remaining 21%. A substantial number of patients (87%) received maintenance treatment with lenalidomide following HDCT/ASCT, while 9% of patients did not receive any maintenance therapy. The remaining patients were treated with other therapeutic agents, which are also listed in Appendix A Table A3. A total of 14 patients (10%) underwent additional ASCT, with 6% of standard-risk, 18% of high-risk, and 13% of ultra-high-risk patients receiving tandem ASCT. No significant differences were identified between the groups with respect to the therapeutic regimens used during the induction, mobilization, HDCT, and maintenance phases, or regarding the administration of tandem ASCT.

2.3. Response

Following induction, most patients achieved VGPR (47%), while 36% reached PR. Notably, 14% of patients were already in CR after induction. After HDCT/ASCT, 76% of patients achieved CR or better, with 42% attaining sCR. The remaining patients were in VGPR (14%) or PR (9%). One patient died on day 25 following HDCT, prior to the assessment of remission status. Among ultra-high-risk patients, 73% achieved CR or sCR, while 85% of the high-risk group and 72% of the standard-risk group reached this outcome. No significant differences were identified between the groups with regard to the stages of remission attained, including the achievement of post-transplant flow MRD negativity (determined on bone marrow day +15 to day +20 following ASCT). A total of 53% of patients classified as ultra-high-risk had an MRD-negative status after receiving HDCT/ASCT. Among high-risk patients, the MRD negativity rate was 60%, while in standard-risk patients it was 54% (p = 0.7910). Overall, 55% of patients were MRD-negative after HDCT/ASCT (Table 2).

2.4. Clinical Outcomes

The clinical outcomes are illustrated in Table 3. The median duration of follow-up was 47 months, with 46 months for standard-risk, 53 months for high-risk, and 49 months for ultra-high-risk patients.
During the follow-up period, 44 patients experienced a relapse, representing an incidence of 32%. Patients with an ultra-high-risk or high-risk constellation exhibited a significantly higher recurrence rate than those with a standard-risk constellation (standard risk 22% vs. high risk 43% vs. ultra-high risk 60%; p = 0.0037). There were also significant differences in PFS (p = 0.0002), which is illustrated in Figure 1a. As the median PFS was not reached in all groups, the 48-month PFS rate for the overall population was 61%. PFS was significantly inferior in high-risk patients compared to standard-risk patients, and even more so in the ultra-high-risk group (48-month PFS: standard risk 72% vs. high risk 50% vs. ultra-high risk 32%; p = 0.0004).
A total of 21 deaths occurred (15%). No statistically significant differences in mortality rates were observed between different risk groups (p = 0.1868), even though—proportionally—27% of ultra-high-risk patients, 20% of high-risk patients, and 11% of standard-risk patients died. Figure 1b visualizes the OS. The overall study population demonstrated an 85% 48-month OS rate, while median OS was not reached in any group. Despite the OS rate for high- and ultra-high-risk patients being lower than for standard-risk patients, no statistically significant differences could be identified (48-month OS: standard risk 89% vs. high risk 79% vs. ultra-high risk 80%; p = 0.1494).
Figure 2 illustrates the PFS and OS for patients with standard-risk cytogenetics stratified by MRD status. Within the standard-risk group, a significantly prolonged PFS was observed in patients who achieved MRD negativity compared to those who did not (p = 0.0172). Nevertheless, no statistically significant association was identified between OS and MRD status in patients with standard-risk genetics (p = 0.6371).
No significant differences were revealed within the high-risk cohort in either PFS or OS depending on MRD status post-transplant (Figure 3).
Likewise, no significant differences in PFS and OS were observed based on MRD status post-transplant within the ultra-high-risk group (Figure 4). However, a trend towards longer survival was observed in patients who achieved MRD negativity. Given the limited sample size of 15 individuals in the ultra-high-risk group, these findings should be cautiously interpreted.
As mentioned before, 14 patients (10%) underwent tandem ASCT. The survival curves of those patients can be found in Figure A1 in the Appendix A. Due to the limited number of patients who received tandem ASCT, no further statistical analysis was performed.

3. Discussion

Our data suggest that MM patients with high-risk cytogenetics have shorter PFS compared to patients with standard-risk alterations, an observation that aligns with prior studies [22,23,24]. Furthermore, our findings indicate that ultra-high-risk MM is associated with an even shorter PFS. Various studies have reported similar findings [7,25]. The MASTER phase 2 trial demonstrated a markedly shortened PFS in MM patients with ultra-high-risk cytogenetics compared to those with standard- or high-risk disease [7], suggesting that even quadruplet induction therapy might be insufficient to improve outcomes for this patient subgroup. This highlights the importance of identifying novel therapeutic strategies for ultra-high-risk MM, particularly in the context of persistent MRD positivity [7].
Given the limited follow-up period of our study, our data are hypothesis-generating, and data on long-term outcomes, such as OS, are immature. In fact, median OS was not reached in any of the subgroups. In other studies, a markedly inferior OS was observed in patients with ultra-high-risk MM [7,25].
In our study, achieving MRD negativity following HDCT and ASCT determined by marrow flow cytometry in patients with standard-risk MM was associated with superior PFS. In contrast, in patients with high-risk MM, achieving MRD negativity did not appear to be sufficient to predict better outcomes. A study conducted by Chakraborty et al. demonstrated a superior PFS by reaching MRD negativity in patients with high-risk myeloma, with the exception of those with del(17p) and patients with ultra-high-risk MM [9]. A comparable conclusion was drawn by the MASTER study. The MRD-driven study design enabled patients to enter treatment-free monitoring when two consecutive negative MRD test results were obtained during the consolidation or maintenance phases. Despite similar MRD negativity rates between patients with standard-, high- and ultra-high-risk cytogenetics, patients with ultra-high-risk MM still showed a shorter PFS and a more frequent MRD resurgence after entering the treatment-free monitoring phase. As suggested by the researchers, this can be explained by the inferior outcomes of MRD-positive patients and the high progression rates of MRD-negative patients [7]. One potential explanation for patients treated at our institution is that most MM patients receive maintenance therapy with lenalidomide for a minimum of two years. A recent study showed that patients who received lenalidomide for at least two years had a longer PFS compared to those with a shorter maintenance treatment [26]. This may be sufficient for MRD-negative standard-risk MM patients, but may not be intensive enough for high-risk or ultra-high-risk patients. An analysis of patients enrolled in the Myeloma XI trial showed a significant benefit in PFS for patients with isolated del(1p), del(17p), or t(4;14) who received lenalidomide maintenance compared to the observation group. Conversely, the findings indicated a limited benefit of lenalidomide maintenance in patients with isolated gain(1q) or ultra-high-risk MM [27]. Shah et al. have suggested that ultra-high-risk patients are unlikely to benefit from lenalidomide as a single-agent maintenance therapy [28]. These patients require more intensive maintenance regimens. Multiple studies evaluated the role of daratumumab in induction and/or maintenance therapy [29,30,31,32,33]. The multicenter OPTIMUM (MUKnine) phase II trial showed that daratumumab, low-dose cyclophosphamide, lenalidomide, bortezomib, and dexamethasone (Dara-CVRd) induction with post-ASCT daratumumab, lenalidomide, bortezomib and dexamethasone (Dara-VRd) consolidation markedly enhances PFS for ultra-high-risk MM patients compared to conventional therapy [31]. The phase 3 PERSEUS trial evaluated the treatment efficacy of Dara-VRd induction and consolidation followed by Dara-lenalidomide (Dara-R) maintenance compared to VRd induction and consolidation followed by lenalidomide (R) maintenance alone in transplant-eligible patients with multiple myeloma. The risk of disease progression and death was significantly lower, while the rates of CR and MRD negativity were significantly increased in the D-VRd treatment group. The randomization process was stratified according to several factors, including cytogenetic risk [29]. A post hoc analysis showed that the combination of D-VRd followed by Dara-R maintenance is more effective than VRd followed by R maintenance in terms of PFS for all cytogenetic risk groups. However, this superiority does not appear to be significant for ultra-high-risk patients and those with isolated gain1q21 [32]. As no re-randomization prior to maintenance therapy was conducted, it is not possible to draw conclusions regarding the extent to which Dara-lenalidomide as a maintenance strategy offers an advantage over lenalidomide alone, regardless of induction [29]. However, possible negative effects of daratumumab during the induction of stem cell mobilization and engraftment in ASCT have been shown [33]. Insights relevant to the efficacy of daratumumab in maintenance therapy are offered by the phase 3 AURIGA study. The trial investigated the efficacy of Dara-R versus R maintenance in transplant-eligible and anti-CD38 naive MM patients. Data from this trial demonstrated a higher MRD-negative conversion rate within 12 months and a longer PFS for patients who received Dara-R as a maintenance strategy compared to R alone. This finding was consistent across all investigated patient groups, including those with high-risk disease [30]. However, the effect on ultra-high-risk MM patients was not examined further.
As only a small proportion of our patients underwent tandem ASCT, a conclusion regarding the effects on outcome cannot be drawn. A retrospective study conducted by the Spanish Myeloma Group indicates that double ASCT may partially overcome the adverse prognosis associated with high-risk genetic abnormalities in comparison to single ASCT [34]. An older analysis by Cavo et al. described a significant PFS benefit associated with double ASCT compared to single ASCT in patients with t(4;14) and/or t(14;16) and/or del(17p), as well a benefit for patients with an additional amp(1q) and/or del(1p) [35].
Some limitations of our study are the relatively small sample size, potentially impacting subgroup analyses. Furthermore, the short follow-up period limits the ability to draw conclusions regarding OS. Comparability with other studies is only possible to a limited extent due to the different aberrations defined as high-risk and ultra-high-risk cytogenetics.
In addition to cytogenetic abnormalities, other factors contribute to a less favorable prognosis. These include a high presence of circulating plasma cells or extramedullary disease at the time of diagnosis. Furthermore, gene expression profiling (GEP) has also been demonstrated to stratify patients according to their risk [15,28,36,37]. Given the multitude of factors that can influence the prognosis of MM patients, it remains an important task to identify a suitable risk stratification strategy. Dynamic MRD monitoring throughout the treatment course might improve the prognostic value of MRD assessment in MM patients. A few studies have already investigated MRD-guided treatment (de-)escalation for MM patients [7,38]. However, the final report of the MASTER trial suggests that these approaches may facilitate therapy de-escalation for standard-risk patients who respond well, but that the results may not be optimal for ultra-high-risk patients [38]. For these patients, it is essential to ascertain suitable treatment strategies, particularly for MM patients with MRD-positive high- or ultra-high-risk disease.

4. Materials and Methods

4.1. Patients

This retrospective study included patients diagnosed with MM who underwent HDCT and ASCT at the University Hospital of Bern, Switzerland, between January 2019 and December 2021. Patients were excluded if data on cytogenetics were unavailable or if MRD assessment had not been performed. The study was approved by the local ethics committee of Berne, Switzerland (approval number: 2024-01246; decision date: 7 August 2024).

4.2. Cytogenetics

Cytogenetics were assessed in bone marrow aspirates at the time of diagnosis using fluorescence in situ hybridization (FISH). TP53 mutations were identified by next-generation sequencing, although this was not performed as a routine testing procedure. For the purpose of analysis, patients were assigned to one of three groups according to their genetic risk profiles based on cytogenetic abnormalities or the presence of a TP53 mutation: the standard-risk group, the high-risk group, or the ultra-high-risk group. High-risk aberrations were defined as del(17p), t(4;14), t(14;16), t(14;20), gain(1q), and TP53 mutations. In case of multiple high-risk aberrations, the patient was classified as ultra-high-risk. Conversely, the absence of high-risk mutations was interpreted as standard-risk genetics.

4.3. MRD

A bone marrow biopsy and flow cytometric immunophenotyping were performed at Bern University Hospital’s Center for Laboratory Medicine between days +15 and +20 post-ASCT to assess minimal residual disease (MRD) status. For MRD assessment, leukemia aberrant immunophenotype (LAIP)-positive cells were quantified within the white blood cell compartment. The presence of <10−5 aberrant plasma cells in the bone marrow was considered indicative of MRD negativity.

4.4. Endpoints

The primary endpoint of the study was PFS, and secondary endpoints were the correlation of PFS with risk categories and MRD status, as well as OS. PFS was defined as the time elapsed between HDCT and either a first relapse, progression, or death, irrespective of the cause of death. OS was defined as the time between HDCT and death, also regardless of the cause. The data cut-off date was 31 May 2024. In the absence of a current follow-up report, it was assumed that the patient had not experienced a relapse or had not died by the time of the analysis. Treatment responses were assessed using the IMWG response criteria [10].

4.5. Statistical Analysis

The data were collected and analyzed using Microsoft Excel 2016. Percentages were rounded to whole numbers. To ensure that the total percentage equaled 100%, the percentage of the largest non-rounded difference value was adjusted. The Kruskal–Wallis test, Chi-square test, and log-rank test were performed to assess comparisons between the standard-, high- and ultra-high-risk groups. Kaplan–Meier curves for PFS and OS were generated using GraphPad Prism version 8.0.1, and p-values were calculated using the Gehan–Breslow–Wilcoxon test.

5. Conclusions

Patients with high- and ultra-high-risk MM exhibited shorter PFS than patients with standard-risk disease. Moreover, while post-transplant flow MRD negativity correlated with longer PFS in standard-risk MM patients, single MRD assessments showed no correlation with PFS in high- and ultra-high-risk patients. These results suggest that high- and ultra-high risk MM patients might benefit from closer response monitoring, including dynamic MRD assessment. Further, these patients might require more intensive peri-transplant management.

Author Contributions

Conceptualization, T.P.; methodology, T.P., M.L. and L.J.K.; validation, T.P., L.J.K. and U.B.; formal analysis, L.J.K.; writing—original draft preparation, L.J.K.; writing—review and editing, D.A. and T.P. All authors contributed to writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the local ethics committee in Berne, Switzerland (approval number: 2024-01246; decision date: 7 August 2024).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All primary data can be provided upon request.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Table A1. Cytogenetics.
Table A1. Cytogenetics.
Parameter Total
(n = 137)
Standard-Risk Genetics
(n = 82)
High-Risk
Genetics
(n = 40)
Ultra-High-Risk Genetics b
(n = 15)
High-risk cytogenetic abnormality, n (%)55 (40)0 (0)40 (100)15 (100)
        del(17p)9 (7)-5 (13)4 (27)
        t(4;14)23 (17)-14 (35)9 (60)
        t(14;16)2 (1)-1 (3)1 (7)
        t(14;20)1 (1)-0 (0)1 (7)
        +1q31 (23)-17 (43)14 (93)
        TP53 mutation6 (4)-3 (8)3 (20)
Standard-risk cytogenetic abnormality a, n (%)117 (85)82 (100)23 (58)12 (80)
        t(11;14)26 (19)20 (24)5 (13)1 (7)
        t(6;14)0 (0)0 (0)0 (0)0 (0)
        del(1p)14 (10)6 (7)4 (10)4 (27)
        Hypodiploidy27 (20)11 (13)9 (23)7 (47)
                −1324 (18)9 (11)8 (20)7 (47)
        Hyperdiploidy53 (39)38 (46)10 (25)5 (33)
                +334 (25)24 (29)7 (18)3 (20)
                +525 (18)19 (23)3 (8)3 (20)
                +722 (16)15 (18)3 (8)4 (27)
                +925 (18)20 (24)3 (8)2 (13)
                +1122 (16)15 (18)3 (8)4 (27)
                +1527 (20)20 (24)5 (13)2 (13)
                +1925 (18)20 (24)3 (8)2 (13)
                +2115 (16)12 (15)1 (3)2 (13)
a Only the most frequently identified standard-risk abnormalities are shown. The following aberrations were detected in ≤5% of cases: t(9;14), Monosomy 2, Monosomy 4, Monosomy 6, Monosomy 8, Monosomy 9, Monosomy 10, Monosomy 12, Monosomy 14, Monosomy 16, Monosomy 17, Monosomy 18, Monosomy 20, Monosomy 22, -Y, -X, del(2q), del(6q), del(8q), del(11q), del(13q), del(14q), de(15q), del(16q), del(17q), del(20q), del(5p), del(7p), del(20p), Trisomy 2, Trisomy 4, Trisomy 6, Trisomy 8, Trisomy 10, Trisomy 12, Trisomy 13, Trisomy 14, Trisomy 16, Trisomy 17, Trisomy 18, Trisomy 20, Trisomy 22, Tetrasomy 7, Tetrasomy 9, Tetrasomy 11, Tetrasomy 14, Tetrasomy 15, Tetrasomy 16, Tetrasomy 18, Tetrasomy 19, Tetrasomy 20, gain(1p), gain(4p), gain(5p), gain(11p), gain(18p), gain(20p), gain(5q), gain(8q), gain(9q), gain(10q), gain(11q), gain(14q), gain(18q), gain(20q), +Y; b 2 patients had more than 2 high-risk aberrations.
Table A2. Karyotype data.
Table A2. Karyotype data.
PatientGenetic Risk ClassificationKaryotype in ISCN Format
1Standard risk54,XY,+3,+4,+7,+9,+15,+17,+19,+21,dup(8)(q),dup(11)(q)
2Standard risk57,XY,+3,+4,+5,+7,+9,+9,+11,+12,+15,+15,+19
3Standard risk?,XY,?
4Standard risk48,XY,+15,+19,+21,−Y
5Standard risk?,XX,+?
6Standard risk54,XY,+3,+4,+9,+11,+15,+19,+20,+21
7Standard risk46,XY,t(11;14)
8Standard risk46,XY,t(11;14),del(16)(q)
9Standard risk?,XX,?
10Standard risk46,XY,t(11;14)
11Standard risk49,XX,+3,+9,+15
12Standard risk50,XY,+5,+9,+15,+19,del(1)(p)
13Standard risk51,XY,+3,+5,+11,+15,+19
14Standard risk54,XY,+3,+5,+7,+9,+11,+15,+19,+Y,del(1)(p)
15Standard risk?,XX,+?
16Standard risk50,XX,+1,+3,+7,+9,−13,+15
17Standard risk53,XY,+3,+5,+7,+9,+15,−16,+19,+21,+Y
18Standard risk61,XY,+1,+2,+3,+4,+5,+6,+7,+9,+11,+15,+15,+17,+19,+19,+21
19Standard risk50,XX,+3,+9,+9,+15,dup(4)(p),dup(11)(q),dup(20)(q)
20Standard risk46,XY,t(11;14)
21Standard risk?,XY,?
22Standard risk46,XY,t(11;14)
23Standard risk44,XY,−13,−14,del(1)(p)
24Standard risk46,XX,t(11;14)
25Standard risk55,XY,+3,+5,+9,+9,+11,+15,+19,+21,+22
26Standard risk46,XY,t(11;14)
27Standard risk?,XX,?
28Standard risk?,XX,+?,del(1)(p)
29Standard risk46,XX,del(1)(p)
30Standard risk46,XX,t(11;14),del(16)(q)
31Standard risk46,XX,t(11;14)
32Standard risk?,XX,?
33Standard risk?,XY,?
34Standard risk46,XY,t(11;14)
35Standard risk46,XX,t(11;14)
36Standard risk49,XY,+4,+9,+11
37Standard risk?,XY,?
38Standard risk53,XY,+3,+5,+7,+9,+9,+15,+19
39Standard risk53,XY,+3,+5,+9,+11,+15,+19,+Y
40Standard risk43,XX,−13,−14,−22
41Standard risk?,XY,?
42Standard risk55,XY,+3,+5,+6,+9,+9,+15,+15,+19,+21
43Standard risk45,XX,−13,dup(18)(q),del(20)(p)
44Standard risk55,XY,+3,+5,+7,+9,+11,+15,+19,+19,+21
45Standard risk43,XY,t(11;14),−13,−14,−Y
46Standard risk?,XX,?
47Standard risk46,XX,t(11;14)
48Standard risk50,XY,+3,+7,+9,+15
49Standard risk49,XY,+3,+7,+9
50Standard risk?,XY,?
51Standard risk?,XY,?
52Standard risk?,XY,?
53Standard risk?,XY,?
54Standard risk48,XX,+5,+9,+11,−13,−16,+19,dup(1)(p),del(17)(q),dup(18)(p)
55Standard risk46,XY,t(11;14)
56Standard risk?,XY,?
57Standard risk48,XY,+9,+15
58Standard risk?,XY,?
59Standard risk58,XY,+2,+3,+5,+6,+7,+9,+9,+11,+15,+15,+19,+21
60Standard risk42,XX,−13,−14,−16,−X
61Standard risk?,XY,?
62Standard risk65,XY,+2,+3,+5,+7,+9,+9,+11,+11,+13,+14,+15,+15,+16,+18,+18,+19,+19,+20,+20
63Standard risk46,XY,t(11;14)
64Standard risk49,XY,+6,+9,+19
65Standard risk?,XY,+?
66Standard risk47,XY,+12
67Standard risk46,XY,+3,−13
68Standard risk50,XY,+14,+14,+16,+16
69Standard risk?,XY,?
70Standard risk45,XY,−13,del(13)(q)
71Standard risk?,XX,?
72Standard risk54,XY,+3,+5,+6,+9,+11,+15,+17,+19,del(1)(p)
73Standard risk54,XY,+3,+5,+7,+9,+11,+15,+19,+21
74Standard risk?,XY,t(9;14),+?
75Standard risk56,XY,+3,+4,+5,+6,+7,+9,+11,+15,+19,+21,dup(8)(q)
76Standard risk46,XX,t(11;14)
77Standard risk46,XX,dup(8)(q)
78Standard risk46,XY,t(11;14)
79Standard risk46,XY,t(11;14)
80Standard risk46,XX,t(11;14)
81Standard risk47,XY,t(11;14),+19
82Standard risk53,XY,+3,+5,+7,+9,+11,+15,+19
83High risk46,XX,dup(1)(q),dup(14)(q),del(16)(q)
84High risk45,XY,−13,dup(1)(q)
85High risk46,XY,t(4;14)
86High risk46,XY,del(17)(p)
87High risk46,XY,t(4;14),del(14)(q)
88High risk46,XX,del(17)(p)
89High risk46,XX,t(4;14)
90High risk?,XY,?
91High risk46,XX,t(4;14)
92High risk46,XX,t(4;14)
93High risk46,XX,t(11;14),dup(1)(q)
94High risk46,XX,del(8)(q),del(14)(q),del(17)(p)
95High risk47,XY,t(11;14),+11,dup(1)(q),dup(11)(q)
96High risk46,XY,dup(1)(q)
97High risk46,XY,t(11;14),dup(1)(q)
98High risk?,XY,?
99High risk46,XX,t(14;16)
100High risk46,XY,t(4;14)
101High risk?,XX,?
102High risk45,XY,−13,−Y,dup(1)(q)
103High risk45,XY,t(4;14),+3,−13,−18
104High risk50,XY,+3,+7,+9,+15,del(1)(p),dup(1)(q)
105High risk46,XY,t(4;14)
106High risk46,XX,t(4;14)
107High risk46,XY,del(1)(p),dup(1)(q)
108High risk57,XX,+3,+5,+7,+7,+9,+10,+15,+17,+18,+19,+19,dup(1)(q)
109High risk44,XX,t(11;14),−13,−X,dup(1)(q)
110High risk46,XX,t(4;14)
111High risk46,XY,t(11;14),dup(1)(q),del(13)(q)
112High risk51,XX,+3,+5,+7,+11,+19,dup(1)(q),del(13)(q)
113High risk46,XY,dup(4)(p),del(17)(p),del(20)(q)
114High risk51,XY,t(4;14),+3,+4,+15,+19,+21
115High risk46,XY,dup(1)(q)
116High risk46,XX,t(4;14),+8,−13
117High risk50,XX,t(4;14),+3,+5,+11,−13,+15,+19,del(1)(p)
118High risk49,XY,+3,+7,+9,−13,+15,dup(1)(q)
119High risk46,XY,t(4;14)
120High risk46,XY,dup(1)(q)
121High risk44,XY,−12,−14,+18,−Y,del(1)(p),dup(1)(q),del(2)(q),del(5)(p),dup(5)(q),del(6)(q),
del(7)(p),dup(9)(q),del(11)(q),del(15)(q),del(16)(q),del(20)(p)
122High risk33,XY,−1,−2,−4,−6,−8,−10,−12,−13,−14,−16,−17,−20,−22,del(17)(p)
123Ultra-high risk44,XY,−13,−16,dup(1)(q),del(17)(p)
124Ultra-high risk53,XY,t(14;20),+3,+5,+7,+8,+11,−13,+18,+19,+21,dup(1)(q),dup(5)(p),dup(9)(q),
dup(10)(q)
125Ultra-high risk45,XX,t(4;14),−13,dup(1)(q)
126Ultra-high risk45,XY,t(4;14),−13,del(1)(p),dup(1)(q)
127Ultra-high risk46,XX,t(4;14),del(1)(p),dup(1)(q)
128Ultra-high risk46,XX,t(14;16),dup(1)(q)
129Ultra-high risk46,XX,t(4;14),dup(1)(q)
130Ultra-high risk46,XY,t(4;14),dup(1)(q)
131Ultra-high risk42,XY,t(4;14),−2,−9,−13,−Y,del(1)(p),dup(1)(q),del(13)(p)
132Ultra-high risk45,XY,t(4;14),−13,dup(1)(q)
133Ultra-high risk56,XY,t(4;14),+2,+3,+5,+7,+9,+11,+15,+17,+19,+21,dup(1)(q)
134Ultra-high risk52,XY,t(11;14),+4,+7,+8,+9,+11,+15,del(17)(p)
135Ultra-high risk45,XY,t(4;14),−13,dup(1)(q)
136Ultra-high risk54,XY,+3,+5,+7,+11,+15,+15,+19,+19,dup(1)(q),del(17)(p)
137Ultra-high risk?,XX,+?,del(1)(p),dup(1)(q),del(17)(p)
Table A3. Course of treatment.
Table A3. Course of treatment.
Parameter.Total
(n = 137)
Standard-Risk Genetics
(n = 82)
High-Risk Genetics
(n = 40)
Ultra-High-Risk Genetics
(n = 15)
p-Value
First-line induction therapy received, n (%)137 (100)82 (100)40 (100)15 (100)-
        VRD118 (86)68 (83)35 (87)15 (100)0.2036
        Dara-VRD5 (4)3 (4)2 (5)0 (0)-
        VCD8 (6)6 (7)2 (5)0 (0)-
        VTD1 (1)1 (1)0 (0)0 (0)-
        VD3 (2)2 (2)1 (3)0 (0)-
        Dara-RD2 (1)2 (2)0 (0)0 (0)-
Second-line induction therapy received, n (%)5 (4)4 (5)1 (3)0 (0)-
Third-line induction therapy received, n (%)3 (2)3 (4)0 (0)0 (0)-
Stem cell mobilisation a, n (%)
        G-CSF only41 (30)24 (29)12 (30)5 (33)0.9512
        Chemotherapy b, G-CSF80 (58)51 (62)22 (55)7 (47)0.4662
        Proteasome inhibitor c, G-CSF13 (10)6 (8)4 (10)3 (20)0.3024
HDCT
        Melphalan43 (31)24 (29)14 (35)5 (33)0.8026
        Bendamustin, Melphalan29 (21)18 (22)7 (18)4 (27)0.7318
        Treosulfan, Melphalan65 (48)40 (49)19 (47)6 (40)0.8220
Tandem ASCT received, n (%)14 (10)5 (6)7 (18)2 (13)0.1361
Maintenance therapy d
        Lenalidomide119 (87)75 (92)31 (78)13 (87)0.1006
        Bortezomib3 (2)1 (1)0 (0)2 (13)-
        Carfilzomib2 (2)1 (1)1 (3)0 (0)-
        Ixazomib3 (2)0 (0)2 (5)1 (7)-
        Daratumumab6 (4)3 (4)2 (5)1 (7)-
        Pomalidomide, Elotuzumab1 (1)1 (1)0 (0)0 (0)-
        None12 (9)5 (6)6 (15)1 (7)0.2518
VRD: bortezomib, lenalidomide, dexamethasone; Dara-VRD: daratumumab, bortezomib, lenalidomide, dexamethasone; VCD: bortezomib, cyclophosphamide, dexamethasone; VTD: bortezomib, thalidomide, dexamethasone; VD: bortezomib, dexamethasone; Dara-RD: daratumumab, lenalidomide, dexamethasone; HDCT: high-dose chemotherapy; ASCT: autologous stem cell transplantation; a data missing for 3 patients; b using vinorelbine or gemcitabine; c using ixazomib; d some patients received several drugs in combination.
Figure A1. (a) Progression-free survival and (b) overall survival for standard- vs. high- vs. ultra-high-risk and single vs. tandem ASCT.
Figure A1. (a) Progression-free survival and (b) overall survival for standard- vs. high- vs. ultra-high-risk and single vs. tandem ASCT.
Cancers 17 01565 g0a1

References

  1. van de Donk, N.W.C.J.; Pawlyn, C.; Yong, K.L. Multiple Myeloma. Lancet 2021, 397, 410–427. [Google Scholar] [CrossRef] [PubMed]
  2. Cowan, A.J.; Allen, C.; Barac, A.; Basaleem, H.; Bensenor, I.; Curado, M.P.; Foreman, K.; Gupta, R.; Harvey, J.; Hosgood, H.D.; et al. Global Burden of Multiple Myeloma: A Systematic Analysis for the Global Burden of Disease Study 2016. JAMA Oncol. 2018, 4, 1221. [Google Scholar] [CrossRef]
  3. Richardson, P.G.; Jacobus, S.J.; Weller, E.A.; Hassoun, H.; Lonial, S.; Raje, N.S.; Medvedova, E.; McCarthy, P.L.; Libby, E.N.; Voorhees, P.M.; et al. Triplet Therapy, Transplantation, and Maintenance until Progression in Myeloma. N. Engl. J. Med. 2022, 387, 132–147. [Google Scholar] [CrossRef]
  4. Hagen, P.; Zhang, J.; Barton, K. High-Risk Disease in Newly Diagnosed Multiple Myeloma: Beyond the R-ISS and IMWG Definitions. Blood Cancer J. 2022, 12, 83. [Google Scholar] [CrossRef] [PubMed]
  5. 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]
  6. Rajkumar, S.V. Multiple Myeloma: 2022 Update on Diagnosis, Risk Stratification, and Management. Am. J. Hematol. 2022, 97, 1086–1107. [Google Scholar] [CrossRef] [PubMed]
  7. 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. 2022, 40, 2901–2912. [Google Scholar] [CrossRef]
  8. Derman, B.A.; Fonseca, R. Measurable Residual Disease and Decision-Making in Multiple Myeloma. Hematol. Oncol. Clin. N. Am. 2024, 38, 477–495. [Google Scholar] [CrossRef]
  9. 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. Impact of Post-Transplant Response and Minimal Residual Disease on Survival in Myeloma with High-Risk Cytogenetics. Biol. Blood Marrow Transplant. 2017, 23, 598–605. [Google Scholar] [CrossRef]
  10. Kumar, S.; Paiva, B.; Anderson, K.C.; Durie, B.; Landgren, O.; Moreau, P.; Munshi, N.; Lonial, S.; Bladé, J.; Mateos, M.-V.; et al. International Myeloma Working Group Consensus Criteria for Response and Minimal Residual Disease Assessment in Multiple Myeloma. Lancet Oncol. 2016, 17, e328–e346. [Google Scholar] [CrossRef]
  11. Paiva, B.; Vidriales, M.-B.; Cerveró, J.; Mateo, G.; Pérez, J.J.; Montalbán, M.A.; Sureda, A.; Montejano, L.; Gutiérrez, N.C.; García de Coca, A.; 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] [PubMed]
  12. 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] [PubMed]
  13. 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]
  14. 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]
  15. Risky Business: Examining the Challenge with Defining High-Risk and Ultra High–Risk Multiple Myeloma. Available online: https://dailynews.ascopubs.org/doi/10.1200/ADN.23.201523 (accessed on 4 November 2024).
  16. 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]
  17. 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] [PubMed]
  18. 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]
  19. Hu, B.; Thall, P.; Milton, D.R.; Sasaki, K.; Bashir, Q.; Shah, N.; Patel, K.; Popat, U.; Hosing, C.; Nieto, Y.; et al. High-Risk Myeloma and Minimal Residual Disease Postautologous-HSCT Predict Worse Outcomes. Leuk. Lymphoma 2019, 60, 442–452. [Google Scholar] [CrossRef]
  20. Kunacheewa, C.; Lee, H.C.; Patel, K.; Thomas, S.; Amini, B.; Srour, S.; Bashir, Q.; Nieto, Y.; Qazilbash, M.H.; Weber, D.M.; et al. Minimal Residual Disease Negativity Does Not Overcome Poor Prognosis in High-Risk Multiple Myeloma: A Single-Center Retrospective Study. Clin. Lymphoma Myeloma Leuk. 2020, 20, e221–e238. [Google Scholar] [CrossRef]
  21. 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]
  22. Joseph, N.S.; Kaufman, J.L.; Dhodapkar, M.V.; Hofmeister, C.C.; Almaula, D.K.; Heffner, L.T.; Gupta, V.A.; Boise, L.H.; Lonial, S.; Nooka, A.K. Long-Term Follow-Up Results of Lenalidomide, Bortezomib, and Dexamethasone Induction Therapy and Risk-Adapted Maintenance Approach in Newly Diagnosed Multiple Myeloma. JCO 2020, 38, 1928–1937. [Google Scholar] [CrossRef] [PubMed]
  23. Avet-Loiseau, H.; Fonseca, R.; Siegel, D.; Dimopoulos, M.A.; Špička, I.; Masszi, T.; Hájek, R.; Rosiñol, L.; Goranova-Marinova, V.; Mihaylov, G.; et al. Carfilzomib Significantly Improves the Progression-Free Survival of High-Risk Patients in Multiple Myeloma. Blood 2016, 128, 1174–1180. [Google Scholar] [CrossRef]
  24. Shah, V.; Sherborne, A.L.; Walker, B.A.; Johnson, D.C.; Boyle, E.M.; Ellis, S.; Begum, D.B.; Proszek, P.Z.; Jones, J.R.; Pawlyn, C.; et al. Prediction of Outcome in Newly Diagnosed Myeloma: A Meta-Analysis of the Molecular Profiles of 1905 Trial Patients. Leukemia 2018, 32, 102–110. [Google Scholar] [CrossRef] [PubMed]
  25. Panopoulou, A.; Easdale, S.; Ethell, M.; Nicholson, E.; Potter, M.; Giotas, A.; Woods, H.; Thornton, T.; Pawlyn, C.; Boyd, K.D.; et al. Impact of Ultra High-Risk Genetics on Real-World Outcomes of Transplant-Eligible Multiple Myeloma Patients. Hemasphere 2023, 7, e831. [Google Scholar] [CrossRef] [PubMed]
  26. 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]
  27. Panopoulou, A.; Cairns, D.A.; Holroyd, A.; Nichols, I.; Cray, N.; Pawlyn, C.; Cook, G.; Drayson, M.; Boyd, K.; Davies, F.E.; et al. Optimizing the Value of Lenalidomide Maintenance by Extended Genetic Profiling: An Analysis of 556 Patients in the Myeloma XI Trial. Blood 2023, 141, 1666–1674. [Google Scholar] [CrossRef]
  28. Shah, V.; Sherborne, A.L.; Johnson, D.C.; Ellis, S.; Price, A.; Chowdhury, F.; Kendall, J.; Jenner, M.W.; Drayson, M.T.; Owen, R.G.; et al. Predicting Ultrahigh Risk Multiple Myeloma by Molecular Profiling: An Analysis of Newly Diagnosed Transplant Eligible Myeloma XI Trial Patients. Leukemia 2020, 34, 3091–3096. [Google Scholar] [CrossRef]
  29. Sonneveld, P.; Dimopoulos, M.A.; Boccadoro, M.; Quach, H.; Ho, P.J.; Beksac, M.; Hulin, C.; Antonioli, E.; Leleu, X.; Mangiacavalli, S.; et al. Daratumumab, Bortezomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N. Engl. J. Med. 2024, 390, 301–313. [Google Scholar] [CrossRef]
  30. Badros, A.Z.; Foster, L.; Anderson, L.D., Jr.; Chaulagain, C.P.; Pettijohn, E.M.; Cowan, A.J.; Costello, C.L.; Larson, S.; Sborov, D.W.; Shain, K.H.; et al. Daratumumab with Lenalidomide as Maintenance after Transplant in Newly Diagnosed Multiple Myeloma: The AURIGA Study. Blood 2024, 145, 300–310. [Google Scholar] [CrossRef]
  31. Kaiser, M.F.; Hall, A.; Walker, K.; Sherborne, A.; Tute, R.M.D.; Newnham, N.; Roberts, S.; Ingleson, E.; Bowles, K.; Garg, M.; et al. Daratumumab, Cyclophosphamide, Bortezomib, Lenalidomide, and Dexamethasone as Induction and Extended Consolidation Improves Outcome in Ultra-High-Risk Multiple Myeloma. J. Clin. Oncol. 2023, 41, 3945–3955. [Google Scholar] [CrossRef]
  32. Dimopoulos, M.A.; Sonneveld, P.; Rodríguez-Otero, P.; Quach, H.; Ho, P.H.; Beksac, M.; Hulin, C.; Antonioli, R.; Leleu, X.; Mangiacavalli, S.; et al. Daratumumab (DARA)/Bortezomib/Lenalidomide/Dexamethasone (D-VRd) with D-R Maintenance in Transplant-Eligible (TE) Newly Diagnosed Multiple Myeloma (NDMM): Analysis of PERSEUS Based on Cytogenetic Risk. In Proceedings of the 2024 European Hematology Association Annual Meeting, Madrid, Spain, 13–16 June 2024. [Google Scholar]
  33. Mehl, J.; Akhoundova, D.; Bacher, U.; Jeker, B.; Rhyner Agocs, G.; Ruefer, A.; Soltermann, S.; Soekler, M.; Winkler, A.; Daskalakis, M.; et al. Daratumumab during Myeloma Induction Therapy Is Associated with Impaired Stem Cell Mobilization and Prolonged Post-Transplant Hematologic Recovery. Cancers 2024, 16, 1854. [Google Scholar] [CrossRef] [PubMed]
  34. Villalba, A.; Gonzalez-Rodriguez, A.P.; Arzuaga-Mendez, J.; Puig, N.; Arnao, M.; Arguiñano, J.M.; Jimenez, M.; Canet, M.; Teruel, A.I.; Sola, M.; et al. Single versus Tandem Autologous Stem-Cell Transplantation in Patients with Newly Diagnosed Multiple Myeloma and High-Risk Cytogenetics. A Retrospective, Open-Label Study of the PETHEMA/Spanish Myeloma Group (GEM). Leuk. Lymphoma 2022, 63, 3438–3447. [Google Scholar] [CrossRef] [PubMed]
  35. Cavo, M.; Gay, F.M.; Patriarca, F.; Zamagni, E.; Montefusco, V.; Dozza, L.; Galli, M.; Bringhen, S.; Testoni, N.; Grasso, M.; et al. Double Autologous Stem Cell Transplantation Significantly Prolongs Progression-Free Survival and Overall Survival in Comparison with Single Autotransplantation in Newly Diagnosed Multiple Myeloma: An Analysis of Phase 3 EMN02/HO95 Study. Blood 2017, 130, 401. [Google Scholar] [CrossRef]
  36. Garcés, J.-J.; Cedena, M.-T.; Puig, N.; Burgos, L.; Perez, J.J.; Cordon, L.; Flores-Montero, J.; Sanoja-Flores, L.; Calasanz, M.-J.; Ortiol, A.; et al. Circulating Tumor Cells for the Staging of Patients with Newly Diagnosed Transplant-Eligible Multiple Myeloma. JCO 2022, 40, 3151–3161. [Google Scholar] [CrossRef] [PubMed]
  37. Varettoni, M.; Corso, A.; Pica, G.; Mangiacavalli, S.; Pascutto, C.; Lazzarino, M. Incidence, Presenting Features and Outcome of Extramedullary Disease in Multiple Myeloma: A Longitudinal Study on 1003 Consecutive Patients. Ann. Oncol. 2010, 21, 325–330. [Google Scholar] [CrossRef]
  38. 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. Minimal Residual Disease Response-Adapted Therapy in Newly Diagnosed Multiple Myeloma (MASTER): Final Report of the Multicentre, Single-Arm, Phase 2 Trial. Lancet Haematol. 2023, 10, e890–e901. [Google Scholar] [CrossRef]
Figure 1. (a) Progression-free survival and (b) overall survival for standard- vs. high- vs. ultra-high-risk patients.
Figure 1. (a) Progression-free survival and (b) overall survival for standard- vs. high- vs. ultra-high-risk patients.
Cancers 17 01565 g001
Figure 2. (a) PFS and (b) OS of the standard-risk group by MRD status determined by flow cytometry of bone marrow on day +15 to day +20 post-transplant.
Figure 2. (a) PFS and (b) OS of the standard-risk group by MRD status determined by flow cytometry of bone marrow on day +15 to day +20 post-transplant.
Cancers 17 01565 g002
Figure 3. (a) PFS and (b) OS of the high-risk group by MRD status.
Figure 3. (a) PFS and (b) OS of the high-risk group by MRD status.
Cancers 17 01565 g003
Figure 4. (a) PFS and (b) OS of the ultra-high-risk group by MRD status.
Figure 4. (a) PFS and (b) OS of the ultra-high-risk group by MRD status.
Cancers 17 01565 g004
Table 1. Patient characteristics at initial diagnosis.
Table 1. Patient characteristics at initial diagnosis.
ParameterTotalStandard-Risk
Genetics
High-Risk Genetics aUltra-High-Risk Genetics bp-Value
Patients, n (%)137 (100)82 (60)40 (29)15 (11)
Median age, years (range)62 (31–75)61 (35–75)62 (31–75)63 (44–75)0.9460
Sex, n (%)
        Male92 (67)58 (71)24 (60)10 (67)0.4952
        Female45 (33)24 (29)16 (40)5 (33)0.4952
R-ISS, n (%)
        I33 (24)24 (29)8 (20)1 (7) 0.1314
        II67 (49)43 (53)19 (47)5 (33)0.3873
        III32 (23)10 (12)13 (33)9 (60)<0.0001
        Unknown c5 (4)5 (6)0 (0)0 (0)-
Paraprotein subtype, n (%)
        Lambda light chain43 (31)18 (22)13 (33)12 (80)<0.0001
        Kappa light chain92 (67)62 (76)27 (68)3 (20)0.0001
        IgG91 (66)60 (73)22 (55)9 (60)0.1170
        IgA23 (17)9 (11)8 (20)6 (40)0.0177
        IgM1 (1)0 (0)1 (2)0 (0)-
        Light-chain-only20 (15)11 (13)9 (23)0 (0)0.0973
        Unknown2 (1)2 (3)0 (0)0 (0)-
Hypercalcemia c, n (%)18 (13)9 (11)7 (18)2 (13)0.6055
Renal insufficiency d, n (%)15 (11)6 (7)6 (15)3 (20)0.2184
Anemia e, n (%) 25 (18)8 (10)13 (33)4 (27)0.0063
Osteolytic lesions, n (%)106 (77)64 (78)33 (83)9 (60)0.2011
Bone marrow infiltration, median, % (range)60 (2–100)50 (2–100)60 (5–100)60 (15–80)0.1080
R-ISS: Revised International Staging System; a 1 of t(4;14), t(14;16), t(14;20), del(17p), gain(1q), or TP53 mutation; b ≥2 of t(4;14), t(14;16), t(14;20), del(17p), gain(1q), or TP53 mutation; c calcium > 2.75 mmol/L; d serum creatinine > 177 µmol/L; e hemoglobin < 100 g/L.
Table 2. Response and MRD.
Table 2. Response and MRD.
ParameterTotal
(n = 137)
Standard-Risk Genetics
(n = 82)
High-Risk Genetics
(n = 40)
Ultra-High-Risk Genetics
(n = 15)
p-Value
Remission status after induction, n (%)
        CR19 (14)11 (13)6 (15)2 (13)0.9701
        VGPR64 (47)38 (46)21 (52)5 (33)0.4445
        PR49 (36)30 (37)12 (30)7 (47)0.5019
        SD5 (3)3 (4)1 (3)1 (7)-
        PD0 (0)0 (0)0 (0)0 (0)-
Remission status after HDCT/ASCT a, n (%)
        sCR58 (42)34 (41)18 (45)6 (40)0.9160
        CR46 (34)25 (31)16 (40)5 (33)0.5795
        VGPR19 (14)13 (16)5 (12)1 (7)0.6112
        PR13 (9)10 (12)1 (3)2 (13)0.1987
        SD0 (0)0 (0)0 (0)0 (0)-
        PD0 (0)0 (0)0 (0)0 (0)-
MRD b status post-transplant
        MRD-negative76 (55)44 (54)24 (60)8 (53)0.7910
        MRD-positive61 (45)38 (46)16 (40)7 (47)0.7910
MRD in patients with tandem ASCT, n (%)
        MRD-negative after first HDCT/ASCT4 (3)0 (0)3 (8)1 (7)-
        MRD-negative after second HDCT/ASCT7 (5)2 (2)4 (10)1 (7)-
        MRD-positive after second HDCT/ASCT7 (5)3 (4)3 (8)1 (7)-
sCR: stringent complete remission; CR: complete remission; VGPR: very good partial remission; PR: partial remission; SD: stable disease; PD: progressive disease; HDCT: high-dose chemotherapy; ASCT: autologous stem cell transplantation; MRD: minimal residual disease by marrow flow cytometry at days 15–20 post-transplant; a one patient died before the remission status was assessed; b after single ASCT or tandem ASCT.
Table 3. Outcomes.
Table 3. Outcomes.
ParameterTotal
(n = 137)
Standard-Risk
Genetics
(n = 82)
High-Risk Genetics
(n = 40)
Ultra-
High-Risk Genetics
(n = 15)
p-Value
Best response after HDCT/ASCT, n (%)
        sCR75 (55)43 (52)24 (60)8 (53)0.7284
        CR47 (34)27 (33)14 (35)6 (40)0.8635
        never in CR15 (11)12 (15)2 (5)1 (7)0.2374
Relapse after HDCT/ASCT, n (%)44 (32)18 (22)17 (43)9 (60)0.0037
48-month PFS rate, %617250320.0004
Death, n (%)21 (15)9 (11) 8 (20)4 (27)0.1868
        Disease progression10 (7)3 (4)5 (13)2 (13)-
        HSCT-related cause1 (1)1 (1)0 (0)0 (0)-
        Other or unknown cause10 (7)5 (6)3 (8)2 (13)-
48-month OS rate, %858979800.1494
Median follow-up after HDCT/ASCT, months (range)47
(0.8–65)
46
(4–64)
53
(5–65)
49
(0.8–59)
0.4512
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kündgen, L.J.; Akhoundova, D.; Hoffmann, M.; Legros, M.; Shaforostova, I.; Seipel, K.; Bacher, U.; Pabst, T. Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients. Cancers 2025, 17, 1565. https://doi.org/10.3390/cancers17091565

AMA Style

Kündgen LJ, Akhoundova D, Hoffmann M, Legros M, Shaforostova I, Seipel K, Bacher U, Pabst T. Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients. Cancers. 2025; 17(9):1565. https://doi.org/10.3390/cancers17091565

Chicago/Turabian Style

Kündgen, Lea Jasmin, Dilara Akhoundova, Michele Hoffmann, Myriam Legros, Inna Shaforostova, Katja Seipel, Ulrike Bacher, and Thomas Pabst. 2025. "Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients" Cancers 17, no. 9: 1565. https://doi.org/10.3390/cancers17091565

APA Style

Kündgen, L. J., Akhoundova, D., Hoffmann, M., Legros, M., Shaforostova, I., Seipel, K., Bacher, U., & Pabst, T. (2025). Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients. Cancers, 17(9), 1565. https://doi.org/10.3390/cancers17091565

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop