Abstract

Background

Smoldering multiple myeloma (SMM), an asymptomatic precursor of multiple myeloma (MM), carries a variable risk of progression to MM. There is little consensus on the efficacy or optimal timing of treatment in SMM. We systematically reviewed the landscape of all clinical trials in SMM. We compared the efficacy of treatment regimens studied in SMM to results from these regimens when used in newly diagnosed multiple myeloma (NDMM), to determine whether the data suggest deeper responses in SMM versus NDMM.

Methods

All prospective interventional clinical trials for SMM, including published studies, meeting abstracts, and unpublished trials listed on ClinicalTrials.gov up to April 1, 2023, were identified. Trial-related variables were captured, including treatment strategy and efficacy results. Relevant clinical endpoints were defined as overall survival (OS) and quality of life.

Results

Among 45 SMM trials identified, 38 (84.4%) assessed active myeloma drugs, while 7 (15.6%) studied bone-modifying agents alone. Of 18 randomized trials in SMM, only one (5.6%) had a primary endpoint of OS; the most common primary endpoint was progression-free survival (n = 7, 38.9%). Among 32 SMM trials with available results, 9 (28.1%) met their prespecified primary endpoint, of which 5 were single-arm studies. Six treatment regimens were tested in both SMM and NDMM; 5 regimens yielded a lower rate of very good partial response rate or better (≥VGPR) in SMM compared to the corresponding NDMM trial (32% vs 63%, 43% vs 53%, 40% vs 63%, 86% vs 89%, 92% vs 95%, and 94% vs 87%, respectively).

Conclusion

In this systematic review of all prospective interventional clinical trials in SMM, we found significant variability in trial design, including randomization status, primary endpoints, and types of intervention used. Despite the statistical limitations, comparison of treatment regimens revealed no compelling evidence that the treatment is more effective when introduced early in SMM compared to NDMM.

Implications for Practice

Our findings suggest that at this time, given changes to imaging and diagnostic classification, there is very limited randomized evidence that support the treatment of SMM, outside of well-designed clinical trials. Further randomized studies, ideally with an observation control arm, that are powered for appropriate primary endpoints (eg, overall survival and quality of life) are warranted to better understand potential benefits or risks of early intervention in SMM.

Introduction

Smoldering multiple myeloma (SMM) is a precursor condition to multiple myeloma (MM), occurring in approximately one in 200 individuals over the age of 40.1 SMM carries a highly variable risk of progressing to MM. Historical studies suggested that approximately 50% of patients with SMM would develop MM within 5 years of diagnosis. However, this is likely an overestimate due to advances in imaging and the 2014 International Myeloma Working Group (IMWG) diagnostic reclassification, which increased the number of people diagnosed with MM.2-4 Although MM is often considered incurable, dramatic advances in treatment raise the question of whether cure may be achievable with novel agents.5,6 In some solid cancers, detection at an earlier stage leading to intervention—especially surgery—can lead to cure, such as in colorectal cancer.7,8 Similarly, it has been hypothesized that early treatment of SMM before progression to MM may either avert morbidity associated with MM and/or increase the possibility of cure.9

There are various hypotheses that biological characteristics of SMM might make it more susceptible to treatment than MM; it is thus often perceived that a given therapy might be more effective in SMM than MM.10-12 Based on the concept of early interception plus the recognition that MM always originates from a precursor state, there is strong interest in early intervention for SMM. For example, the ASCENT trial of aggressive 4-drug therapy for SMM was based on the hypothesis that intense therapy applied at this precursor phase could potentially eradicate the malignant clone and lead to long term remissions or even cure.10 Furthermore, theoretically preventing or curing MM before it leads to morbidity, such as fractures and renal failure, would be ideal.

Two prospective trials suggested that progression to MM may be delayed by treating SMM. In the randomized QUIREDEX study of 119 patients with high-risk SMM (HR-SMM), the median time to progression (TTP) was significantly longer in patients treated with lenalidomide and dexamethasone compared to patients who were observed.13 In a second, more recent trial of 182 patients, of whom 56 had HR-SMM, the use of lenalidomide reduced the risk of progression of SMM to MM.14 However, neither trial was adequately powered to assess overall survival (OS), and their relevance to today’s patients is limited, given that the patients previously diagnosed with highest-risk SMM would now be classified as having MM, and many patients previously thought asymptomatic may now be classified as having MM due to more sensitive imaging.15,16 Furthermore, as these trials did not clearly outline the nature of progression events, “progression” could range from an asymptomatic lab change, such as a drop in hemoglobin, to more clinically meaningful issues such as fractures or permanent renal failure. Since these events vary significantly in severity, it remains unclear whether the therapy is preventing or delaying asymptomatic, reversible lab changes or symptomatic, irreversible organ damage. Therefore, whether or not to treat patients with high-risk SMM remains controversial.17

It ultimately remains unknown whether treatments that are known to be effective against MM are indeed more effective when used in SMM compared to reserving their use for patients whose disease progresses to MM. Furthermore, there is great variability in the methodology of clinical trials that seek to evaluate early interventions in SMM, making it difficult to incorporate the evidence from these largely single-arm trials into clinical practice.18,19 In order to capture both completed and in-progress clinical trials in SMM, we systematically reviewed all prospective clinical trials in SMM. To interrogate the hypothesis that treatment may be more effective when used in SMM, we compared the results of treatment regimens used to treat SMM to that regimen’s corresponding outcomes in newly diagnosed MM (NDMM), wherever applicable and possible.

Methods

Search strategy and selection criteria

A comprehensive search strategy to identify prospective trials was constructed in Embase (Embase.com, Elsevier) by a health librarian and a clinician using truncated keywords, phrases, proximity searching and subject headings for SMM, and the CADTH Clinical Trials search filter.20 This strategy was translated to MEDLINE, Cochrane Central Register of Controlled Trials, and the Web of Science Core Collection with initial searches performed on November 10, 2022, and a follow-up search of ClinicalTrials.gov on April 1, 2023 (see Supplementary Appendix I for detailed search strategies). There was no start date for our search strategy, as we aimed to include all published and unpublished work on SMM. Thus, the start of our first SMM trial was October 1, 1983. For all SMM studies, a search was done on PubMed and Google Scholar for corresponding prospective trials in NDMM using the same regimen on April 30, 2023. No publication date or language limits were used. All results were exported to EndNote version 20 (Clarivate, Philadelphia, USA); duplicates were removed with EndNote’s duplicate detection algorithms and manual inspection.

Two independent reviewers (A.K. and A.T.) screened all studies with discrepancies resolved by a third reviewer (G.R.M.). This systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations21 and a PROSPERO protocol (CRD42022373951) outlining our search and analysis plan a priori was submitted before search initiation. This study was not submitted for Institutional Review Board approval as it was not human participant research.

Our search strategy included all prospective therapeutic clinical trials from 1980 to April 1, 2023. Other study types were excluded (eg, observational cohort and registry). Only studies with patients meeting the diagnostic criteria for SMM were included, while studies of all other plasma cell dyscrasias such as MM were excluded. Studies also had to evaluate potentially active myeloma drugs (including drugs considered to potentially be active but not proven, such as celecoxib and metformin) or bone-modifying agents; studies of other treatment types were excluded. Results posted on non-peer reviewed sources such as trial registry sites and abstracts from conference proceedings captured via our search strategy were included. Trials that were in progress that did not yet have results but had a clear description of methodology and interventions were included.

Statistical analysis

Two authors (A.K. and A.T.) extracted the following data using Microsoft Excel (Microsoft, Washington, USA): trial design (randomized or nonrandomized), number of patients, median age, treatment regimen, duration of treatment, length of follow-up, study enrollment period, primary endpoint type and outcomes, overall and treatment-related deaths, funding source, and study location. Clinical endpoints were defined according to FDA guidelines as overall survival and symptom endpoints (patient-reported outcomes), while surrogate endpoints included overall response rate (ORR), progression-free survival (PFS), TTP, measurable residual disease (MRD), and safety.22

The primary objective of our study was to systematically review clinical trials in SMM, ascertaining whether these studies were randomized, whether they met their prespecified primary endpoint (that the study was statistically powered to assess), and the type of intervention studied, which we categorized as intensive multiagent therapy, preventative low-intensity therapy, or bone-strengthening agents. A key secondary objective was to compare the results of individual regimens used to treat SMM to the corresponding outcomes of that regimen when used in NDMM. The prospective trial in NDMM that had the greatest similarity in dosing and administration to the comparable trial in SMM was used for comparison. We descriptively compared rates of very good partial response or greater (≥VGPR), and also collected ORR and complete response rates for each regimen. We chose to compare response rates and MRD rates rather than PFS, as progression is defined differently in MM (defined by formal progression criteria based on laboratory values and/or end-organ damage), in contrast to SMM, where it is often defined as progression to MM.4 Furthermore, time-to-event endpoints such as PFS in SMM can incorporate lead-time bias due to earlier diagnosis.

All completed randomized trials were assessed for bias using the Cochrane Handbook for Systematic Review of Interventions, version 6.2 and Cochrane risk-of-bias tool,23,24 and all completed nonrandomized trials were appraised across all domains of bias using the Cochrane Collaboration Risk of Bias in Nonrandomized Studies of Intervention (ROBINS-I) tool.25

Results

From an initial search finding 1492 results, 45 studies10,11,13,14,26-66 were included after excluding duplicates and studies not meeting SMM diagnostic or treatment inclusion criteria (Figure 1). Most trials studied active myeloma drugs (n = 38, 84.4%)10,11,13,14,26-29,32,33,35-39,41-52,54,56-65 while the remainder studied bone-modifying agents (n = 7, 15.6%; Table 1).30,31,34,40,53,55,66

Table 1.

Characteristics of included studies.

Characteristic (n = 45)No. of trials (%)
Study status
 Completed23 (51.1)
 Active, not recruiting, preliminary results available7 (15.6)
 Recruiting, preliminary results available2 (4.4)
 Active, not recruiting, no results available2 (4.4)
 Recruiting, no results available11 (24.4)
Study design
 Randomized18 (40.0)
  Blinded (amongst randomized)4 (22.2)
  Treatment control arm (amongst randomized)9 (50.0)
  Observation control arm (amongst randomized)9 (50.0)
 Nonrandomized27 (60.0)
  Single-arm23 (51.1)
  Observation control arm1 (2.2)
  Two-group study2 (4.4)
  Multi-group study1 (2.2)
 Median sample size (IQR)54 (27.5-104.5)
  Randomized studies102 (51-177)
  Nonrandomized studies38 (20-61)
 Year study started patient enrollment
  Pre-20005 (11.1)
  2001-201316 (35.6)
  Post-201424 (53.3)
Publication format
 Manuscript20 (44.4)
 Abstract13 (28.9)
 ClinicalTrials.gov only12 (26.7)
Treatment approach
 Single agent18 (40.0)
 Multiagent20 (44.4)
 Bone-modifying agent7 (15.6)
Primary endpoint
 Response rate16 (35.6)
  Randomized3 (16.7)
  Nonrandomized13 (48.1)
 PFS10 (22.2)
  Randomized7 (38.9)
  Nonrandomized3 (11.1)
 MRD3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 OS1 (2.2)
  Randomized1 (5.6)
  Nonrandomized0 (0.0)
 TTP3 (6.7)
  Randomized3 (16.7)
  Nonrandomized0 (0.0)
 Translational2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
 M-protein level3 (6.7)
  Randomized2 (11.1)
  Nonrandomized1 (3.7)
 Safety3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 Multiple2 (4.4)
  Randomized2 (11.1)
  Nonrandomized0 (0.0)
 Other2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
Funding source
 Industry23 (51.1)
 Nonindustry19 (42.2)
 Unknown3 (6.7)
Location
 USA29 (64.4)
 International including USA5 (11.1)
 Non-USA only11 (24.4)
Characteristic (n = 45)No. of trials (%)
Study status
 Completed23 (51.1)
 Active, not recruiting, preliminary results available7 (15.6)
 Recruiting, preliminary results available2 (4.4)
 Active, not recruiting, no results available2 (4.4)
 Recruiting, no results available11 (24.4)
Study design
 Randomized18 (40.0)
  Blinded (amongst randomized)4 (22.2)
  Treatment control arm (amongst randomized)9 (50.0)
  Observation control arm (amongst randomized)9 (50.0)
 Nonrandomized27 (60.0)
  Single-arm23 (51.1)
  Observation control arm1 (2.2)
  Two-group study2 (4.4)
  Multi-group study1 (2.2)
 Median sample size (IQR)54 (27.5-104.5)
  Randomized studies102 (51-177)
  Nonrandomized studies38 (20-61)
 Year study started patient enrollment
  Pre-20005 (11.1)
  2001-201316 (35.6)
  Post-201424 (53.3)
Publication format
 Manuscript20 (44.4)
 Abstract13 (28.9)
 ClinicalTrials.gov only12 (26.7)
Treatment approach
 Single agent18 (40.0)
 Multiagent20 (44.4)
 Bone-modifying agent7 (15.6)
Primary endpoint
 Response rate16 (35.6)
  Randomized3 (16.7)
  Nonrandomized13 (48.1)
 PFS10 (22.2)
  Randomized7 (38.9)
  Nonrandomized3 (11.1)
 MRD3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 OS1 (2.2)
  Randomized1 (5.6)
  Nonrandomized0 (0.0)
 TTP3 (6.7)
  Randomized3 (16.7)
  Nonrandomized0 (0.0)
 Translational2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
 M-protein level3 (6.7)
  Randomized2 (11.1)
  Nonrandomized1 (3.7)
 Safety3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 Multiple2 (4.4)
  Randomized2 (11.1)
  Nonrandomized0 (0.0)
 Other2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
Funding source
 Industry23 (51.1)
 Nonindustry19 (42.2)
 Unknown3 (6.7)
Location
 USA29 (64.4)
 International including USA5 (11.1)
 Non-USA only11 (24.4)

Abbreviations: IQR, interquartile range; M-protein, monoclonal protein; MRD, measurable residual disease; OS, overall survival; PFS, progression-free survival; TTP, time to progression.

Table 1.

Characteristics of included studies.

Characteristic (n = 45)No. of trials (%)
Study status
 Completed23 (51.1)
 Active, not recruiting, preliminary results available7 (15.6)
 Recruiting, preliminary results available2 (4.4)
 Active, not recruiting, no results available2 (4.4)
 Recruiting, no results available11 (24.4)
Study design
 Randomized18 (40.0)
  Blinded (amongst randomized)4 (22.2)
  Treatment control arm (amongst randomized)9 (50.0)
  Observation control arm (amongst randomized)9 (50.0)
 Nonrandomized27 (60.0)
  Single-arm23 (51.1)
  Observation control arm1 (2.2)
  Two-group study2 (4.4)
  Multi-group study1 (2.2)
 Median sample size (IQR)54 (27.5-104.5)
  Randomized studies102 (51-177)
  Nonrandomized studies38 (20-61)
 Year study started patient enrollment
  Pre-20005 (11.1)
  2001-201316 (35.6)
  Post-201424 (53.3)
Publication format
 Manuscript20 (44.4)
 Abstract13 (28.9)
 ClinicalTrials.gov only12 (26.7)
Treatment approach
 Single agent18 (40.0)
 Multiagent20 (44.4)
 Bone-modifying agent7 (15.6)
Primary endpoint
 Response rate16 (35.6)
  Randomized3 (16.7)
  Nonrandomized13 (48.1)
 PFS10 (22.2)
  Randomized7 (38.9)
  Nonrandomized3 (11.1)
 MRD3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 OS1 (2.2)
  Randomized1 (5.6)
  Nonrandomized0 (0.0)
 TTP3 (6.7)
  Randomized3 (16.7)
  Nonrandomized0 (0.0)
 Translational2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
 M-protein level3 (6.7)
  Randomized2 (11.1)
  Nonrandomized1 (3.7)
 Safety3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 Multiple2 (4.4)
  Randomized2 (11.1)
  Nonrandomized0 (0.0)
 Other2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
Funding source
 Industry23 (51.1)
 Nonindustry19 (42.2)
 Unknown3 (6.7)
Location
 USA29 (64.4)
 International including USA5 (11.1)
 Non-USA only11 (24.4)
Characteristic (n = 45)No. of trials (%)
Study status
 Completed23 (51.1)
 Active, not recruiting, preliminary results available7 (15.6)
 Recruiting, preliminary results available2 (4.4)
 Active, not recruiting, no results available2 (4.4)
 Recruiting, no results available11 (24.4)
Study design
 Randomized18 (40.0)
  Blinded (amongst randomized)4 (22.2)
  Treatment control arm (amongst randomized)9 (50.0)
  Observation control arm (amongst randomized)9 (50.0)
 Nonrandomized27 (60.0)
  Single-arm23 (51.1)
  Observation control arm1 (2.2)
  Two-group study2 (4.4)
  Multi-group study1 (2.2)
 Median sample size (IQR)54 (27.5-104.5)
  Randomized studies102 (51-177)
  Nonrandomized studies38 (20-61)
 Year study started patient enrollment
  Pre-20005 (11.1)
  2001-201316 (35.6)
  Post-201424 (53.3)
Publication format
 Manuscript20 (44.4)
 Abstract13 (28.9)
 ClinicalTrials.gov only12 (26.7)
Treatment approach
 Single agent18 (40.0)
 Multiagent20 (44.4)
 Bone-modifying agent7 (15.6)
Primary endpoint
 Response rate16 (35.6)
  Randomized3 (16.7)
  Nonrandomized13 (48.1)
 PFS10 (22.2)
  Randomized7 (38.9)
  Nonrandomized3 (11.1)
 MRD3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 OS1 (2.2)
  Randomized1 (5.6)
  Nonrandomized0 (0.0)
 TTP3 (6.7)
  Randomized3 (16.7)
  Nonrandomized0 (0.0)
 Translational2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
 M-protein level3 (6.7)
  Randomized2 (11.1)
  Nonrandomized1 (3.7)
 Safety3 (6.7)
  Randomized0 (0.0)
  Nonrandomized3 (11.1)
 Multiple2 (4.4)
  Randomized2 (11.1)
  Nonrandomized0 (0.0)
 Other2 (4.4)
  Randomized0 (0.0)
  Nonrandomized2 (7.4)
Funding source
 Industry23 (51.1)
 Nonindustry19 (42.2)
 Unknown3 (6.7)
Location
 USA29 (64.4)
 International including USA5 (11.1)
 Non-USA only11 (24.4)

Abbreviations: IQR, interquartile range; M-protein, monoclonal protein; MRD, measurable residual disease; OS, overall survival; PFS, progression-free survival; TTP, time to progression.

Flow diagram summarizing strategy for identification and inclusion of studies.
Figure 1.

Flow diagram depicting search strategy and study inclusion.

Eighteen studies (40.0%)13,14,27-33,50,52,54-56,58,59,64,65 were randomized trials while 27 studies (60.0%)10,11,26,34-49,51,53,57,60-63,66 were nonrandomized trials. Of 18 randomized trials, the most common primary endpoint was PFS (n = 7, 38.9%),14,27,30,31,50,52,56 followed by ORR (n = 3, 16.7%),59,64,65 TTP (n = 3, 16.7%),13,29,55 M-protein level (n = 2, 11.1%),32,58 and OS (n = 1, 5.6%)54; 2 studies (11.1%)28,33 had multiple primary endpoints. Fourteen (78%) randomized trials13,14,28-31,33,50,52,54,56,59,64,65 were open label and 4 (22%)27,32,55,58 were blinded. Only one study (5.6%)54 had OS as its primary endpoint. Amongst 27 nonrandomized trials, the most common primary endpoint was ORR (n = 13, 48.1%),10,11,35-37,39,40,46-48,60,62,66 followed by safety (n = 3, 11.1%),42,51,63 PFS (n = 3, 11.1%),45,49,61 MRD (n = 3, 11.1%),23,43,57 a translational outcome (n = 2, 7.4%),38,41 other outcomes (n = 2, 7.4%),34,53 and M-protein level (n = 1, 3.7%).44

Thirty-two studies10,11,13,14,26-49,53,57,65,66 had reported results: 23 (71.9%)13,14,27-45,65,66 were completed trials with final results available and 9 (28.1%)10,11,26,46-49,53,57 were ongoing with only preliminary results. Among these 32 trials, single-agent treatment (n = 14, 43.8%),14,16,27,28,32,35,37-39,41,44,47,48,65 multiagent approaches (n = 12, 37.5%),13,15,26,29,33,36,42,43,45,46,49,57 and bone-modifying agents (n = 6, 18.8%)30,31,34,40,53,55,66 were tested. Most were nonrandomized (n = 22, 68.8%).10,11,26,34-49,53,57,66Table 2 lists the characteristics of the 10 (31.3%) completed randomized trials13,14,27-33,65 with final results available, including primary outcome results.

Table 2.

Characteristics of completed randomized SMM trials.

Author,
year
Trial name/NCT numberTotal number of patientsStudy designIntervention regimenControl regimenPrimary endpointPrimary endpoint met?Primary endpoint results (intervention vs control)
Mateos et al, 201313QUIREDEX
NCT00480363
119Parallel assignmentLenalidomide + dexamethasoneObservationTTPYESAfter a median follow-up time of 12.5 years (range: 10.4-13.6), the median TTP to MM was 2.1 years vs 9.5 years (HR: 0.28, 95% CI: 0.18-0.44, P < .0001)
Brighton et al, 201927NCT0148427574Parallel assignment; double-blindedSiltuximabPlaceboPFSNOThe 1-year PFS rate was 84.5% [95% CI, 68.6-92.8] vs 74.4% (95% CI, 57.3-85.5). The study did not meet the prespecified hypothesis that siltuximab would increase the 1-year PFS rate by at least 14%
Landgren et al, 202028CENTAURUS
NCT02316106
123Parallel assignment, open labelDaratumumab (intense, intermediate, and short)NACo-primary endpoints (CR and PD/death rate per patient-year)NOWith longer follow-up (median follow-up, 25.9 months), CR rates were 4.9%, 9.8%, and 0% for intense, intermediate, and short dosing, respectively. PD/death rates were 0.059 (80% CI, 0.025-0.092), 0.107 (80% CI, 0.058-0.155), and 0.150 (80% CI, 0.089-0.211), translating to a median PFS ≥ 24 months in all arms (P < .0001 for all arms)
Witzig et al, 201329NCT0043245868Parallel assignment, open labelThalidomide and zoledronic acidZoledronic acidTTPYESMedian TTP was 2.4 years (95% CI: 1.4-3.6) vs 1.2 years (95% CI: 0.7-2.5; HR, 2.05; 95% CI: 1.1-3.8; P-value: .02)
D’arena et al, 201130NA197Parallel assignment, open labelPamidronateObservationPFSNOWith a minimum follow-up of 5 years for living patients, there were 56/89 (62.9%) progressions vs 55/88 (62.5%; P = NS). Median TTP was 46 and 48 months, respectively (P = NS)
Musto et al,31 2008EUCTR2006-003854-33-IT163Parallel assignment, open labelZoledronic acidObservationPFSNOAfter a median follow-up of 64.7 person-months, 44.4% of patients in the intervention group and 45.1% of the control group progressed to “symptomatic” myeloma requiring chemotherapy (P = .9307)
Horwitz et al, 201232NCT0009904723Parallel assignment, double blindCelecoxibPlaceboM-protein levelsNOCelecoxib had no significant effect on the median monoclonal protein concentration, which went from a median of 1.44-1.65 g/dL with placebo and 2.42-2.24 g/dL with celecoxib (P = .36)
Lonial et al, 202014NCT01169337182Parallel assignment, open labelLenalidomideObservationPFSYESPFS was significantly longer with lenalidomide compared with observation (HR, 0.28; 95% CI, 0.12-0.62; P = .002). 1-, 2-, and 3-year progression-free survival was 98%, 93%, and 91% vs 89%, 76%, and 66%, respectively
NAKIROMONO
NCT01222286
30Parallel assignment, open labelIPH2101 0.2 mg/kg or
IPH2101 2 mg/kg
NAResponseNO0% of patients achieved an objective response with either dose of IPH2101
Hjorth et al, 199333NA50Assignment based on disease classificationInitial melphalan-prednisoneObservation until disease progression then melphalan-prednisoneMultiple (response rate, plateau phase/response duration, and overall survival)NOResponse rate: response was seen in 13 out of 25 (52%) patients vs 12 out of 22 (55%). There was no difference in survival between the 2 treatment groups. Response duration: the duration of relapse-free survival after the cessation of treatment was similar in both groups (P = .17; median duration 21 months vs 31 months). Survival: there was no difference in survival between the 2 treatment groups
Author,
year
Trial name/NCT numberTotal number of patientsStudy designIntervention regimenControl regimenPrimary endpointPrimary endpoint met?Primary endpoint results (intervention vs control)
Mateos et al, 201313QUIREDEX
NCT00480363
119Parallel assignmentLenalidomide + dexamethasoneObservationTTPYESAfter a median follow-up time of 12.5 years (range: 10.4-13.6), the median TTP to MM was 2.1 years vs 9.5 years (HR: 0.28, 95% CI: 0.18-0.44, P < .0001)
Brighton et al, 201927NCT0148427574Parallel assignment; double-blindedSiltuximabPlaceboPFSNOThe 1-year PFS rate was 84.5% [95% CI, 68.6-92.8] vs 74.4% (95% CI, 57.3-85.5). The study did not meet the prespecified hypothesis that siltuximab would increase the 1-year PFS rate by at least 14%
Landgren et al, 202028CENTAURUS
NCT02316106
123Parallel assignment, open labelDaratumumab (intense, intermediate, and short)NACo-primary endpoints (CR and PD/death rate per patient-year)NOWith longer follow-up (median follow-up, 25.9 months), CR rates were 4.9%, 9.8%, and 0% for intense, intermediate, and short dosing, respectively. PD/death rates were 0.059 (80% CI, 0.025-0.092), 0.107 (80% CI, 0.058-0.155), and 0.150 (80% CI, 0.089-0.211), translating to a median PFS ≥ 24 months in all arms (P < .0001 for all arms)
Witzig et al, 201329NCT0043245868Parallel assignment, open labelThalidomide and zoledronic acidZoledronic acidTTPYESMedian TTP was 2.4 years (95% CI: 1.4-3.6) vs 1.2 years (95% CI: 0.7-2.5; HR, 2.05; 95% CI: 1.1-3.8; P-value: .02)
D’arena et al, 201130NA197Parallel assignment, open labelPamidronateObservationPFSNOWith a minimum follow-up of 5 years for living patients, there were 56/89 (62.9%) progressions vs 55/88 (62.5%; P = NS). Median TTP was 46 and 48 months, respectively (P = NS)
Musto et al,31 2008EUCTR2006-003854-33-IT163Parallel assignment, open labelZoledronic acidObservationPFSNOAfter a median follow-up of 64.7 person-months, 44.4% of patients in the intervention group and 45.1% of the control group progressed to “symptomatic” myeloma requiring chemotherapy (P = .9307)
Horwitz et al, 201232NCT0009904723Parallel assignment, double blindCelecoxibPlaceboM-protein levelsNOCelecoxib had no significant effect on the median monoclonal protein concentration, which went from a median of 1.44-1.65 g/dL with placebo and 2.42-2.24 g/dL with celecoxib (P = .36)
Lonial et al, 202014NCT01169337182Parallel assignment, open labelLenalidomideObservationPFSYESPFS was significantly longer with lenalidomide compared with observation (HR, 0.28; 95% CI, 0.12-0.62; P = .002). 1-, 2-, and 3-year progression-free survival was 98%, 93%, and 91% vs 89%, 76%, and 66%, respectively
NAKIROMONO
NCT01222286
30Parallel assignment, open labelIPH2101 0.2 mg/kg or
IPH2101 2 mg/kg
NAResponseNO0% of patients achieved an objective response with either dose of IPH2101
Hjorth et al, 199333NA50Assignment based on disease classificationInitial melphalan-prednisoneObservation until disease progression then melphalan-prednisoneMultiple (response rate, plateau phase/response duration, and overall survival)NOResponse rate: response was seen in 13 out of 25 (52%) patients vs 12 out of 22 (55%). There was no difference in survival between the 2 treatment groups. Response duration: the duration of relapse-free survival after the cessation of treatment was similar in both groups (P = .17; median duration 21 months vs 31 months). Survival: there was no difference in survival between the 2 treatment groups

Abbreviations: CR, complete response; HR, hazard ratio; NA, not available; NS, not significant; M-protein, monoclonal protein; PD, progressive disease; PFS, progression-free survival; TTP, time to progression.

Table 2.

Characteristics of completed randomized SMM trials.

Author,
year
Trial name/NCT numberTotal number of patientsStudy designIntervention regimenControl regimenPrimary endpointPrimary endpoint met?Primary endpoint results (intervention vs control)
Mateos et al, 201313QUIREDEX
NCT00480363
119Parallel assignmentLenalidomide + dexamethasoneObservationTTPYESAfter a median follow-up time of 12.5 years (range: 10.4-13.6), the median TTP to MM was 2.1 years vs 9.5 years (HR: 0.28, 95% CI: 0.18-0.44, P < .0001)
Brighton et al, 201927NCT0148427574Parallel assignment; double-blindedSiltuximabPlaceboPFSNOThe 1-year PFS rate was 84.5% [95% CI, 68.6-92.8] vs 74.4% (95% CI, 57.3-85.5). The study did not meet the prespecified hypothesis that siltuximab would increase the 1-year PFS rate by at least 14%
Landgren et al, 202028CENTAURUS
NCT02316106
123Parallel assignment, open labelDaratumumab (intense, intermediate, and short)NACo-primary endpoints (CR and PD/death rate per patient-year)NOWith longer follow-up (median follow-up, 25.9 months), CR rates were 4.9%, 9.8%, and 0% for intense, intermediate, and short dosing, respectively. PD/death rates were 0.059 (80% CI, 0.025-0.092), 0.107 (80% CI, 0.058-0.155), and 0.150 (80% CI, 0.089-0.211), translating to a median PFS ≥ 24 months in all arms (P < .0001 for all arms)
Witzig et al, 201329NCT0043245868Parallel assignment, open labelThalidomide and zoledronic acidZoledronic acidTTPYESMedian TTP was 2.4 years (95% CI: 1.4-3.6) vs 1.2 years (95% CI: 0.7-2.5; HR, 2.05; 95% CI: 1.1-3.8; P-value: .02)
D’arena et al, 201130NA197Parallel assignment, open labelPamidronateObservationPFSNOWith a minimum follow-up of 5 years for living patients, there were 56/89 (62.9%) progressions vs 55/88 (62.5%; P = NS). Median TTP was 46 and 48 months, respectively (P = NS)
Musto et al,31 2008EUCTR2006-003854-33-IT163Parallel assignment, open labelZoledronic acidObservationPFSNOAfter a median follow-up of 64.7 person-months, 44.4% of patients in the intervention group and 45.1% of the control group progressed to “symptomatic” myeloma requiring chemotherapy (P = .9307)
Horwitz et al, 201232NCT0009904723Parallel assignment, double blindCelecoxibPlaceboM-protein levelsNOCelecoxib had no significant effect on the median monoclonal protein concentration, which went from a median of 1.44-1.65 g/dL with placebo and 2.42-2.24 g/dL with celecoxib (P = .36)
Lonial et al, 202014NCT01169337182Parallel assignment, open labelLenalidomideObservationPFSYESPFS was significantly longer with lenalidomide compared with observation (HR, 0.28; 95% CI, 0.12-0.62; P = .002). 1-, 2-, and 3-year progression-free survival was 98%, 93%, and 91% vs 89%, 76%, and 66%, respectively
NAKIROMONO
NCT01222286
30Parallel assignment, open labelIPH2101 0.2 mg/kg or
IPH2101 2 mg/kg
NAResponseNO0% of patients achieved an objective response with either dose of IPH2101
Hjorth et al, 199333NA50Assignment based on disease classificationInitial melphalan-prednisoneObservation until disease progression then melphalan-prednisoneMultiple (response rate, plateau phase/response duration, and overall survival)NOResponse rate: response was seen in 13 out of 25 (52%) patients vs 12 out of 22 (55%). There was no difference in survival between the 2 treatment groups. Response duration: the duration of relapse-free survival after the cessation of treatment was similar in both groups (P = .17; median duration 21 months vs 31 months). Survival: there was no difference in survival between the 2 treatment groups
Author,
year
Trial name/NCT numberTotal number of patientsStudy designIntervention regimenControl regimenPrimary endpointPrimary endpoint met?Primary endpoint results (intervention vs control)
Mateos et al, 201313QUIREDEX
NCT00480363
119Parallel assignmentLenalidomide + dexamethasoneObservationTTPYESAfter a median follow-up time of 12.5 years (range: 10.4-13.6), the median TTP to MM was 2.1 years vs 9.5 years (HR: 0.28, 95% CI: 0.18-0.44, P < .0001)
Brighton et al, 201927NCT0148427574Parallel assignment; double-blindedSiltuximabPlaceboPFSNOThe 1-year PFS rate was 84.5% [95% CI, 68.6-92.8] vs 74.4% (95% CI, 57.3-85.5). The study did not meet the prespecified hypothesis that siltuximab would increase the 1-year PFS rate by at least 14%
Landgren et al, 202028CENTAURUS
NCT02316106
123Parallel assignment, open labelDaratumumab (intense, intermediate, and short)NACo-primary endpoints (CR and PD/death rate per patient-year)NOWith longer follow-up (median follow-up, 25.9 months), CR rates were 4.9%, 9.8%, and 0% for intense, intermediate, and short dosing, respectively. PD/death rates were 0.059 (80% CI, 0.025-0.092), 0.107 (80% CI, 0.058-0.155), and 0.150 (80% CI, 0.089-0.211), translating to a median PFS ≥ 24 months in all arms (P < .0001 for all arms)
Witzig et al, 201329NCT0043245868Parallel assignment, open labelThalidomide and zoledronic acidZoledronic acidTTPYESMedian TTP was 2.4 years (95% CI: 1.4-3.6) vs 1.2 years (95% CI: 0.7-2.5; HR, 2.05; 95% CI: 1.1-3.8; P-value: .02)
D’arena et al, 201130NA197Parallel assignment, open labelPamidronateObservationPFSNOWith a minimum follow-up of 5 years for living patients, there were 56/89 (62.9%) progressions vs 55/88 (62.5%; P = NS). Median TTP was 46 and 48 months, respectively (P = NS)
Musto et al,31 2008EUCTR2006-003854-33-IT163Parallel assignment, open labelZoledronic acidObservationPFSNOAfter a median follow-up of 64.7 person-months, 44.4% of patients in the intervention group and 45.1% of the control group progressed to “symptomatic” myeloma requiring chemotherapy (P = .9307)
Horwitz et al, 201232NCT0009904723Parallel assignment, double blindCelecoxibPlaceboM-protein levelsNOCelecoxib had no significant effect on the median monoclonal protein concentration, which went from a median of 1.44-1.65 g/dL with placebo and 2.42-2.24 g/dL with celecoxib (P = .36)
Lonial et al, 202014NCT01169337182Parallel assignment, open labelLenalidomideObservationPFSYESPFS was significantly longer with lenalidomide compared with observation (HR, 0.28; 95% CI, 0.12-0.62; P = .002). 1-, 2-, and 3-year progression-free survival was 98%, 93%, and 91% vs 89%, 76%, and 66%, respectively
NAKIROMONO
NCT01222286
30Parallel assignment, open labelIPH2101 0.2 mg/kg or
IPH2101 2 mg/kg
NAResponseNO0% of patients achieved an objective response with either dose of IPH2101
Hjorth et al, 199333NA50Assignment based on disease classificationInitial melphalan-prednisoneObservation until disease progression then melphalan-prednisoneMultiple (response rate, plateau phase/response duration, and overall survival)NOResponse rate: response was seen in 13 out of 25 (52%) patients vs 12 out of 22 (55%). There was no difference in survival between the 2 treatment groups. Response duration: the duration of relapse-free survival after the cessation of treatment was similar in both groups (P = .17; median duration 21 months vs 31 months). Survival: there was no difference in survival between the 2 treatment groups

Abbreviations: CR, complete response; HR, hazard ratio; NA, not available; NS, not significant; M-protein, monoclonal protein; PD, progressive disease; PFS, progression-free survival; TTP, time to progression.

Among 32 trials with results available, 9 studies (28.1%)13,14,26,29,37,38,42,43,48 met their prespecified primary endpoint while 18 studies (56.3%)10,27,28,30-35,39-41,44,46,47,53,65,66 had not; 2 (6.3%)49,57 had not yet reported primary results, and 3 (9.4%)11,36,45 had unclear primary outcome results. Among 9 trials that met their primary endpoint, 7 (77.8%)13,14,29,37,38,42,43 were published in manuscripts and 2 (22.2%)26,48 in abstracts. Of the remaining 23 trials, 16 (69.6%)27,28,30-36,39-41,44,45,65,66 have been completed without clear evidence of having met their primary endpoint. Of these 16 trials, 11 (68.8%)27,28,30,31,33,35,36,39,41,44,66 published in manuscripts, 4 (25.0%)32,34,40,45 in abstract form, and one (6.3%)65 on ClinicalTrials.gov only.

Of 13 ongoing studies50-52,54-56,58-64 without results available, 8 (61.5%)50,52,54-56,58,59,64 are randomized. The majority of these 8 randomized studies list surrogate markers as the primary endpoint, including PFS (n = 3, 37.5%),50,52,56 ORR (n = 2, 25.0%),59,64 TTP (n = 1, 12.5%),55 and M-protein level (n = 1, 12.5%).58

Among 8 SMM ongoing randomized clinical trials without available results, 552,54,56,59,64 use active control arms (lenalidomide plus dexamethasone in NCT04270409, NCT03673826, NCT03937635, NCT05469893 and iberdomide in NCT04776395) whereas 3 ongoing trials assign observation or placebo to the control arm (NCT03301220, NCT04850846, and NCT03792763).

The median sample size of 10 completed randomized studies was 102 patients (IQR 55-153); of these trials, 4 (40.0%)28,31,32,65 had a low overall risk of bias, 3 (30.0%)14,27,29 had some concerns, and 3 (30.0%)13,30,33 studies had a high risk of bias, according to the Cochrane risk-of-bias tool (Supplementary Appendix II). Of 13 completed nonrandomized trials,34–45,66 median sample size was 31 patients (IQR 22-50). The majority (n = 10, 76.9%)36–41,43–45,66 had a moderate risk of bias, 2 studies35,42 were at serious risk of bias, and one study34 had insufficient information to allow appraisal (Supplementary Appendix III).

Among 6 treatment regimens with ≥VGPR data available in both SMM and NDMM settings (Table 3; Supplementary Appendix IV), 5 regimens yielded a lower ≥VGPR in SMM compared to NDMM: lenalidomide plus dexamethasone (32% in SMM vs 63% in NDMM); elotuzumab, lenalidomide and dexamethasone (43% vs 53%); ixazomib, lenalidomide and dexamethasone (40% vs 63%); carfilzomib, lenalidomide, dexamethasone, autologous stem cell transplant (ASCT; 86% vs 89%); and daratumumab, carfilzomib, lenalidomide and dexamethasone without ASCT (92% vs 95%; Figure 2); whereas one regimen showed the reverse: carfilzomib, lenalidomide and dexamethasone without ASCT (94% in SMM vs 87% in NDMM). MRD results were sparsely and heterogeneously assessed73; however, of the 3 regimens with MRD-negativity rates available in both SMM and NDMM settings, one regimen (daratumumab, carfilzomib, lenalidomide and dexamethasone without ASCT) had higher MRD-negativity rates in SMM, one (carfilzomib, lenalidomide, dexamethasone, and ASCT) had higher MRD-negativity rates in NDMM, and the other (carfilzomib, lenalidomide, and dexamethasone) had similar rates in both settings.10,26,43,68,71

Table 3.

Comparison of regimen efficacy in smoldering myeloma versus newly diagnosed myeloma.

RegimenNCT number of smoldering trial and NDMM trialDosing in smoldering myeloma trialDosing in multiple myeloma trialORR in SMMORR in NDMM≥VGPR in SMM≥VGPR in NDMM≥CRR in SMM≥CRR in NDMMMRD data in SMMMRD data in NDMM
Lenalidomide-dexamethasoneNCT00480363
NCT00064038
Len = 25 mg (days 1-21), dex 20 mg (days 1-4, 12-15, and 12-15) for nine 28-day cycles, then maintenance len = 10 mg (days 1-21) for 2 years13Len = 25 mg (days 1-28), dex 40 mg (days 1-4, 9-12, and 17-20) of 28-day cycle for 3 cycles, followed by maintenance of dex 40 mg (days 1-4 and 15-18) and len 25 mg (days 1-21) of 28-day cycle continuously until progression/intolerance6779%
(95% CI, 66%-89%) at end of induction with 9 cycles
78%
(95% CI, 67%-86%) after one year of therapy
32%
(95% CI, 20%-45%)
63%
(95% CI, 52%-74%)
21%
(95% CI, 11%-34%)
26%
(95% CI, 17%-37%)
NRNR
LenalidomideNCT01169337Len = 25 mg (days 1-21) of 28-day cycle14No corresponding trial exists for lenalidomide monotherapy in NDMM50% (95% CI, 39%-61%) at any point while on therapy during 2 yearsNA4.5%
(95% CI, 1%-11%)
NA0%NANRNA
Carfilzomib-lenalidomide-dexamethasone (KRd)NCT01572480
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg days 1-21, dex (20 mg C1-4; C5-8 twice a week) for 8 cycles → Len (10 mg) × 24 cycles43K = 36 mg/m2 (first 2 doses at 20mg/m2) on days 1, 2, 8, 9, 15, and 16, len = 25 mg days 1-21, dex (20 mg twice a week) for 12 cycles, separate randomization for maintenance68100%
(95% CI, 93%-100%) after 8 cycles of therapy
94%
(95% CI, 89%-97%) after 1 year of therapy, prior to second randomization
94.4%
(95% CI, 85%-99%)
87%
(95% CI, 81%-92%)
75.9%
(95% CI, 62%-87%)
57%
(95% CI, 49%-65%)
70.4% MRD negative CRRs at 10−569% at 10−5 after completion of 12 cycles of KRd
Elotuzumab-lenalidomide-dexamethasone (ERd)NCT02279394
NCT01335399
Elotuzumab standard dosinga, Dex = 40 mg days 1, 8, 15, and 22, cycles 1-2, then 40 mg oral days 1, 8, and 15, cycles 3-8. Len monotherapy continued for 2 years of therapy45Elotuzumab standard dosinga, Len = 25 mg days 1-21, Dex = 40 mg days 1, 8, 15, and 22 of each cycle until progression/toxicity6984%
(95% CI, 70%-93%) at any point within 2 years of therapy
83%
(95% CI, 79%-87%) at any point during therapy
43%
(95% CI, 29%-58%)
53%
(95% CI, 47%-58%)
6%
(95% CI, 1%-17%)
18%
(95% CI, 14%-22%)
NRNR
Ixazomib-lenalidomide-dexamethasone
(IRd)
NCT02916771
NCT01850524
Ixazomib = 4 mg days 1, 8, and 15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle; dex discontinued after cycle 8, total of 24 cycles49Ixazomib = 4 mg days 1,8,15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle. Dex discontinued and ixazomib/len reduced to 3 mg/10 mg respectively, continued until progression/intolerance7090.9%
(95% CI, 80%-97%) at any point during the study treatment period
82.1%
(95% CI, 78%-86%) at any point during study treatment period
40%
(95% CI, 27%-54%)
63%
(95% CI, 58%-68%)
21.8%
(95% CI, 12%-35%)
25.6%
(95% CI, 21%-31%)
NR101 patients (28.8% of patients in IRd arm) had MRD evaluated: 52.5% MRD negative at 10−5
Carfilzomib-lenalidomide-dexamethasone + ASCT
(GEM-CESAR trial, KRd-ASCT)
NCT02415413
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg (days 1-21), dex = 40 mg weekly for six 28-day cycles → Mel-200/ASCT → KRD x 2 cycles →Rd × 2 years, (len = 10 mg/day, dex = 20 mg/week)26Carfilzomib = 36 mg/m2 (first 2 doses = 20 mg/m2) days 1, 2, 8, 9, 15, and 16, len = 25 mg (days 1-21), dex = 20 mg twice a week for four 28-day cycles → Mel-200/ASCT→ KRD × 2 cycles → randomization for maintenance6894%
(95% CI, 88%-98%) after end of consolidation therapy
97%
(95% CI, 93%-99%) after end of consolidation therapy
86%
(95% CI, 78%-93%)
89%
(95% CI, 83%-93%)
70%
(95% CI, 61%-80%)
54%
(95% CI, 46%-62%)
63%
MRD neg rates at 10−5 post ASCT
23% sustained MRD at 10−6 4 years after ASCT
80%
MRD neg rates at 10−5 post ASCT
Daratumumab-carfilzomib-lenalidomide-dexamethasone
without transplant
(ASCENT trial, DKRd)
NCT03289299
NCT03290950
K = 20/56 mg/m2 once weekly, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 28-day cycles. Dara standard dosingb, len dose reduced and dex discontinued after 12 cycles; total 2 years therapy10Carfilzomib = 56 mg/m2 days 1, 8, and 15, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 8 cycles.7197%
(95% CI, 90%-99%) at any point during study treatment period
100%
(95% CI, 91%-100%) after 8 cycles of therapy
92%
(95% CI, 84%-97%)
95%
(95% CI, 83%-99%)
63%
(95% CI, 52%-73%)
NR84% at 10−5
At median time to MRD negativity of 6.6 months
71% at 10−5
after 8 cycles of therapy
Daratumumab-lenalidomide-bortezomib-dexamethasone without transplant
(DVRd)
NCT04775550Dara standard dosingb, bortezomib days 1, 8, and 15 for cycles 1-6 and then biweekly until completion of cycle 24. Len is administered days 1-21 and dex is administered weekly until cycle 2472No corresponding trial exists in NDMM90%
(95% CI, 68%-99%) at any point during study treatment period
NA50%
(95% CI, 27%-73%)
NA25%
(95% CI, 8%-49%)
NA50% at 10−5
25% at 10−6
after≥6 months follow-up, in those for whom MRD assessed
NA
RegimenNCT number of smoldering trial and NDMM trialDosing in smoldering myeloma trialDosing in multiple myeloma trialORR in SMMORR in NDMM≥VGPR in SMM≥VGPR in NDMM≥CRR in SMM≥CRR in NDMMMRD data in SMMMRD data in NDMM
Lenalidomide-dexamethasoneNCT00480363
NCT00064038
Len = 25 mg (days 1-21), dex 20 mg (days 1-4, 12-15, and 12-15) for nine 28-day cycles, then maintenance len = 10 mg (days 1-21) for 2 years13Len = 25 mg (days 1-28), dex 40 mg (days 1-4, 9-12, and 17-20) of 28-day cycle for 3 cycles, followed by maintenance of dex 40 mg (days 1-4 and 15-18) and len 25 mg (days 1-21) of 28-day cycle continuously until progression/intolerance6779%
(95% CI, 66%-89%) at end of induction with 9 cycles
78%
(95% CI, 67%-86%) after one year of therapy
32%
(95% CI, 20%-45%)
63%
(95% CI, 52%-74%)
21%
(95% CI, 11%-34%)
26%
(95% CI, 17%-37%)
NRNR
LenalidomideNCT01169337Len = 25 mg (days 1-21) of 28-day cycle14No corresponding trial exists for lenalidomide monotherapy in NDMM50% (95% CI, 39%-61%) at any point while on therapy during 2 yearsNA4.5%
(95% CI, 1%-11%)
NA0%NANRNA
Carfilzomib-lenalidomide-dexamethasone (KRd)NCT01572480
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg days 1-21, dex (20 mg C1-4; C5-8 twice a week) for 8 cycles → Len (10 mg) × 24 cycles43K = 36 mg/m2 (first 2 doses at 20mg/m2) on days 1, 2, 8, 9, 15, and 16, len = 25 mg days 1-21, dex (20 mg twice a week) for 12 cycles, separate randomization for maintenance68100%
(95% CI, 93%-100%) after 8 cycles of therapy
94%
(95% CI, 89%-97%) after 1 year of therapy, prior to second randomization
94.4%
(95% CI, 85%-99%)
87%
(95% CI, 81%-92%)
75.9%
(95% CI, 62%-87%)
57%
(95% CI, 49%-65%)
70.4% MRD negative CRRs at 10−569% at 10−5 after completion of 12 cycles of KRd
Elotuzumab-lenalidomide-dexamethasone (ERd)NCT02279394
NCT01335399
Elotuzumab standard dosinga, Dex = 40 mg days 1, 8, 15, and 22, cycles 1-2, then 40 mg oral days 1, 8, and 15, cycles 3-8. Len monotherapy continued for 2 years of therapy45Elotuzumab standard dosinga, Len = 25 mg days 1-21, Dex = 40 mg days 1, 8, 15, and 22 of each cycle until progression/toxicity6984%
(95% CI, 70%-93%) at any point within 2 years of therapy
83%
(95% CI, 79%-87%) at any point during therapy
43%
(95% CI, 29%-58%)
53%
(95% CI, 47%-58%)
6%
(95% CI, 1%-17%)
18%
(95% CI, 14%-22%)
NRNR
Ixazomib-lenalidomide-dexamethasone
(IRd)
NCT02916771
NCT01850524
Ixazomib = 4 mg days 1, 8, and 15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle; dex discontinued after cycle 8, total of 24 cycles49Ixazomib = 4 mg days 1,8,15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle. Dex discontinued and ixazomib/len reduced to 3 mg/10 mg respectively, continued until progression/intolerance7090.9%
(95% CI, 80%-97%) at any point during the study treatment period
82.1%
(95% CI, 78%-86%) at any point during study treatment period
40%
(95% CI, 27%-54%)
63%
(95% CI, 58%-68%)
21.8%
(95% CI, 12%-35%)
25.6%
(95% CI, 21%-31%)
NR101 patients (28.8% of patients in IRd arm) had MRD evaluated: 52.5% MRD negative at 10−5
Carfilzomib-lenalidomide-dexamethasone + ASCT
(GEM-CESAR trial, KRd-ASCT)
NCT02415413
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg (days 1-21), dex = 40 mg weekly for six 28-day cycles → Mel-200/ASCT → KRD x 2 cycles →Rd × 2 years, (len = 10 mg/day, dex = 20 mg/week)26Carfilzomib = 36 mg/m2 (first 2 doses = 20 mg/m2) days 1, 2, 8, 9, 15, and 16, len = 25 mg (days 1-21), dex = 20 mg twice a week for four 28-day cycles → Mel-200/ASCT→ KRD × 2 cycles → randomization for maintenance6894%
(95% CI, 88%-98%) after end of consolidation therapy
97%
(95% CI, 93%-99%) after end of consolidation therapy
86%
(95% CI, 78%-93%)
89%
(95% CI, 83%-93%)
70%
(95% CI, 61%-80%)
54%
(95% CI, 46%-62%)
63%
MRD neg rates at 10−5 post ASCT
23% sustained MRD at 10−6 4 years after ASCT
80%
MRD neg rates at 10−5 post ASCT
Daratumumab-carfilzomib-lenalidomide-dexamethasone
without transplant
(ASCENT trial, DKRd)
NCT03289299
NCT03290950
K = 20/56 mg/m2 once weekly, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 28-day cycles. Dara standard dosingb, len dose reduced and dex discontinued after 12 cycles; total 2 years therapy10Carfilzomib = 56 mg/m2 days 1, 8, and 15, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 8 cycles.7197%
(95% CI, 90%-99%) at any point during study treatment period
100%
(95% CI, 91%-100%) after 8 cycles of therapy
92%
(95% CI, 84%-97%)
95%
(95% CI, 83%-99%)
63%
(95% CI, 52%-73%)
NR84% at 10−5
At median time to MRD negativity of 6.6 months
71% at 10−5
after 8 cycles of therapy
Daratumumab-lenalidomide-bortezomib-dexamethasone without transplant
(DVRd)
NCT04775550Dara standard dosingb, bortezomib days 1, 8, and 15 for cycles 1-6 and then biweekly until completion of cycle 24. Len is administered days 1-21 and dex is administered weekly until cycle 2472No corresponding trial exists in NDMM90%
(95% CI, 68%-99%) at any point during study treatment period
NA50%
(95% CI, 27%-73%)
NA25%
(95% CI, 8%-49%)
NA50% at 10−5
25% at 10−6
after≥6 months follow-up, in those for whom MRD assessed
NA

Abbreviations: ASCT, autologous stem cell transplant; C, cycle; CRR, complete response rate; Dara (D), daratumumab; Dex (d), dexamethasone; E, elotuzumab; I, ixazomib; K, carfilzomib; Len (R), lenalidomide; Mel, melphalan; MRD; measurable residual disease; NA, not available; NCT, national clinical trial; NDMM, newly diagnosed multiple myeloma; NR, not reported; ORR, overall response rate; SMM, smoldering multiple myeloma; VGPR, very good partial response.

aElotozumab standard dosing is as follows; cycles 1 and 2: 10 mg/kg IV once a week on days 1, 8, 15, and 22 of a 28-day cycle, cycle 3 and beyond: 10 mg/kg IV once every 2 weeks on days 1 and 15 of a 28-day cycle until disease progression or unacceptable toxicity.

bDaratumumab standard dosing is weekly for first 8 weeks, every 2 weeks for next 16 weeks, and then monthly.

Table 3.

Comparison of regimen efficacy in smoldering myeloma versus newly diagnosed myeloma.

RegimenNCT number of smoldering trial and NDMM trialDosing in smoldering myeloma trialDosing in multiple myeloma trialORR in SMMORR in NDMM≥VGPR in SMM≥VGPR in NDMM≥CRR in SMM≥CRR in NDMMMRD data in SMMMRD data in NDMM
Lenalidomide-dexamethasoneNCT00480363
NCT00064038
Len = 25 mg (days 1-21), dex 20 mg (days 1-4, 12-15, and 12-15) for nine 28-day cycles, then maintenance len = 10 mg (days 1-21) for 2 years13Len = 25 mg (days 1-28), dex 40 mg (days 1-4, 9-12, and 17-20) of 28-day cycle for 3 cycles, followed by maintenance of dex 40 mg (days 1-4 and 15-18) and len 25 mg (days 1-21) of 28-day cycle continuously until progression/intolerance6779%
(95% CI, 66%-89%) at end of induction with 9 cycles
78%
(95% CI, 67%-86%) after one year of therapy
32%
(95% CI, 20%-45%)
63%
(95% CI, 52%-74%)
21%
(95% CI, 11%-34%)
26%
(95% CI, 17%-37%)
NRNR
LenalidomideNCT01169337Len = 25 mg (days 1-21) of 28-day cycle14No corresponding trial exists for lenalidomide monotherapy in NDMM50% (95% CI, 39%-61%) at any point while on therapy during 2 yearsNA4.5%
(95% CI, 1%-11%)
NA0%NANRNA
Carfilzomib-lenalidomide-dexamethasone (KRd)NCT01572480
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg days 1-21, dex (20 mg C1-4; C5-8 twice a week) for 8 cycles → Len (10 mg) × 24 cycles43K = 36 mg/m2 (first 2 doses at 20mg/m2) on days 1, 2, 8, 9, 15, and 16, len = 25 mg days 1-21, dex (20 mg twice a week) for 12 cycles, separate randomization for maintenance68100%
(95% CI, 93%-100%) after 8 cycles of therapy
94%
(95% CI, 89%-97%) after 1 year of therapy, prior to second randomization
94.4%
(95% CI, 85%-99%)
87%
(95% CI, 81%-92%)
75.9%
(95% CI, 62%-87%)
57%
(95% CI, 49%-65%)
70.4% MRD negative CRRs at 10−569% at 10−5 after completion of 12 cycles of KRd
Elotuzumab-lenalidomide-dexamethasone (ERd)NCT02279394
NCT01335399
Elotuzumab standard dosinga, Dex = 40 mg days 1, 8, 15, and 22, cycles 1-2, then 40 mg oral days 1, 8, and 15, cycles 3-8. Len monotherapy continued for 2 years of therapy45Elotuzumab standard dosinga, Len = 25 mg days 1-21, Dex = 40 mg days 1, 8, 15, and 22 of each cycle until progression/toxicity6984%
(95% CI, 70%-93%) at any point within 2 years of therapy
83%
(95% CI, 79%-87%) at any point during therapy
43%
(95% CI, 29%-58%)
53%
(95% CI, 47%-58%)
6%
(95% CI, 1%-17%)
18%
(95% CI, 14%-22%)
NRNR
Ixazomib-lenalidomide-dexamethasone
(IRd)
NCT02916771
NCT01850524
Ixazomib = 4 mg days 1, 8, and 15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle; dex discontinued after cycle 8, total of 24 cycles49Ixazomib = 4 mg days 1,8,15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle. Dex discontinued and ixazomib/len reduced to 3 mg/10 mg respectively, continued until progression/intolerance7090.9%
(95% CI, 80%-97%) at any point during the study treatment period
82.1%
(95% CI, 78%-86%) at any point during study treatment period
40%
(95% CI, 27%-54%)
63%
(95% CI, 58%-68%)
21.8%
(95% CI, 12%-35%)
25.6%
(95% CI, 21%-31%)
NR101 patients (28.8% of patients in IRd arm) had MRD evaluated: 52.5% MRD negative at 10−5
Carfilzomib-lenalidomide-dexamethasone + ASCT
(GEM-CESAR trial, KRd-ASCT)
NCT02415413
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg (days 1-21), dex = 40 mg weekly for six 28-day cycles → Mel-200/ASCT → KRD x 2 cycles →Rd × 2 years, (len = 10 mg/day, dex = 20 mg/week)26Carfilzomib = 36 mg/m2 (first 2 doses = 20 mg/m2) days 1, 2, 8, 9, 15, and 16, len = 25 mg (days 1-21), dex = 20 mg twice a week for four 28-day cycles → Mel-200/ASCT→ KRD × 2 cycles → randomization for maintenance6894%
(95% CI, 88%-98%) after end of consolidation therapy
97%
(95% CI, 93%-99%) after end of consolidation therapy
86%
(95% CI, 78%-93%)
89%
(95% CI, 83%-93%)
70%
(95% CI, 61%-80%)
54%
(95% CI, 46%-62%)
63%
MRD neg rates at 10−5 post ASCT
23% sustained MRD at 10−6 4 years after ASCT
80%
MRD neg rates at 10−5 post ASCT
Daratumumab-carfilzomib-lenalidomide-dexamethasone
without transplant
(ASCENT trial, DKRd)
NCT03289299
NCT03290950
K = 20/56 mg/m2 once weekly, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 28-day cycles. Dara standard dosingb, len dose reduced and dex discontinued after 12 cycles; total 2 years therapy10Carfilzomib = 56 mg/m2 days 1, 8, and 15, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 8 cycles.7197%
(95% CI, 90%-99%) at any point during study treatment period
100%
(95% CI, 91%-100%) after 8 cycles of therapy
92%
(95% CI, 84%-97%)
95%
(95% CI, 83%-99%)
63%
(95% CI, 52%-73%)
NR84% at 10−5
At median time to MRD negativity of 6.6 months
71% at 10−5
after 8 cycles of therapy
Daratumumab-lenalidomide-bortezomib-dexamethasone without transplant
(DVRd)
NCT04775550Dara standard dosingb, bortezomib days 1, 8, and 15 for cycles 1-6 and then biweekly until completion of cycle 24. Len is administered days 1-21 and dex is administered weekly until cycle 2472No corresponding trial exists in NDMM90%
(95% CI, 68%-99%) at any point during study treatment period
NA50%
(95% CI, 27%-73%)
NA25%
(95% CI, 8%-49%)
NA50% at 10−5
25% at 10−6
after≥6 months follow-up, in those for whom MRD assessed
NA
RegimenNCT number of smoldering trial and NDMM trialDosing in smoldering myeloma trialDosing in multiple myeloma trialORR in SMMORR in NDMM≥VGPR in SMM≥VGPR in NDMM≥CRR in SMM≥CRR in NDMMMRD data in SMMMRD data in NDMM
Lenalidomide-dexamethasoneNCT00480363
NCT00064038
Len = 25 mg (days 1-21), dex 20 mg (days 1-4, 12-15, and 12-15) for nine 28-day cycles, then maintenance len = 10 mg (days 1-21) for 2 years13Len = 25 mg (days 1-28), dex 40 mg (days 1-4, 9-12, and 17-20) of 28-day cycle for 3 cycles, followed by maintenance of dex 40 mg (days 1-4 and 15-18) and len 25 mg (days 1-21) of 28-day cycle continuously until progression/intolerance6779%
(95% CI, 66%-89%) at end of induction with 9 cycles
78%
(95% CI, 67%-86%) after one year of therapy
32%
(95% CI, 20%-45%)
63%
(95% CI, 52%-74%)
21%
(95% CI, 11%-34%)
26%
(95% CI, 17%-37%)
NRNR
LenalidomideNCT01169337Len = 25 mg (days 1-21) of 28-day cycle14No corresponding trial exists for lenalidomide monotherapy in NDMM50% (95% CI, 39%-61%) at any point while on therapy during 2 yearsNA4.5%
(95% CI, 1%-11%)
NA0%NANRNA
Carfilzomib-lenalidomide-dexamethasone (KRd)NCT01572480
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg days 1-21, dex (20 mg C1-4; C5-8 twice a week) for 8 cycles → Len (10 mg) × 24 cycles43K = 36 mg/m2 (first 2 doses at 20mg/m2) on days 1, 2, 8, 9, 15, and 16, len = 25 mg days 1-21, dex (20 mg twice a week) for 12 cycles, separate randomization for maintenance68100%
(95% CI, 93%-100%) after 8 cycles of therapy
94%
(95% CI, 89%-97%) after 1 year of therapy, prior to second randomization
94.4%
(95% CI, 85%-99%)
87%
(95% CI, 81%-92%)
75.9%
(95% CI, 62%-87%)
57%
(95% CI, 49%-65%)
70.4% MRD negative CRRs at 10−569% at 10−5 after completion of 12 cycles of KRd
Elotuzumab-lenalidomide-dexamethasone (ERd)NCT02279394
NCT01335399
Elotuzumab standard dosinga, Dex = 40 mg days 1, 8, 15, and 22, cycles 1-2, then 40 mg oral days 1, 8, and 15, cycles 3-8. Len monotherapy continued for 2 years of therapy45Elotuzumab standard dosinga, Len = 25 mg days 1-21, Dex = 40 mg days 1, 8, 15, and 22 of each cycle until progression/toxicity6984%
(95% CI, 70%-93%) at any point within 2 years of therapy
83%
(95% CI, 79%-87%) at any point during therapy
43%
(95% CI, 29%-58%)
53%
(95% CI, 47%-58%)
6%
(95% CI, 1%-17%)
18%
(95% CI, 14%-22%)
NRNR
Ixazomib-lenalidomide-dexamethasone
(IRd)
NCT02916771
NCT01850524
Ixazomib = 4 mg days 1, 8, and 15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle; dex discontinued after cycle 8, total of 24 cycles49Ixazomib = 4 mg days 1,8,15, len = 25 mg days 1-21, dex = 40 mg days 1, 8, 15, and 22 of 28-day cycle. Dex discontinued and ixazomib/len reduced to 3 mg/10 mg respectively, continued until progression/intolerance7090.9%
(95% CI, 80%-97%) at any point during the study treatment period
82.1%
(95% CI, 78%-86%) at any point during study treatment period
40%
(95% CI, 27%-54%)
63%
(95% CI, 58%-68%)
21.8%
(95% CI, 12%-35%)
25.6%
(95% CI, 21%-31%)
NR101 patients (28.8% of patients in IRd arm) had MRD evaluated: 52.5% MRD negative at 10−5
Carfilzomib-lenalidomide-dexamethasone + ASCT
(GEM-CESAR trial, KRd-ASCT)
NCT02415413
NCT02203643
K = 20/36 mg/m2 twice weekly, len = 25 mg (days 1-21), dex = 40 mg weekly for six 28-day cycles → Mel-200/ASCT → KRD x 2 cycles →Rd × 2 years, (len = 10 mg/day, dex = 20 mg/week)26Carfilzomib = 36 mg/m2 (first 2 doses = 20 mg/m2) days 1, 2, 8, 9, 15, and 16, len = 25 mg (days 1-21), dex = 20 mg twice a week for four 28-day cycles → Mel-200/ASCT→ KRD × 2 cycles → randomization for maintenance6894%
(95% CI, 88%-98%) after end of consolidation therapy
97%
(95% CI, 93%-99%) after end of consolidation therapy
86%
(95% CI, 78%-93%)
89%
(95% CI, 83%-93%)
70%
(95% CI, 61%-80%)
54%
(95% CI, 46%-62%)
63%
MRD neg rates at 10−5 post ASCT
23% sustained MRD at 10−6 4 years after ASCT
80%
MRD neg rates at 10−5 post ASCT
Daratumumab-carfilzomib-lenalidomide-dexamethasone
without transplant
(ASCENT trial, DKRd)
NCT03289299
NCT03290950
K = 20/56 mg/m2 once weekly, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 28-day cycles. Dara standard dosingb, len dose reduced and dex discontinued after 12 cycles; total 2 years therapy10Carfilzomib = 56 mg/m2 days 1, 8, and 15, len = 25 mg days 1-21, dara = 16 mg/kg weekly, dex = 40 mg weekly for 8 cycles.7197%
(95% CI, 90%-99%) at any point during study treatment period
100%
(95% CI, 91%-100%) after 8 cycles of therapy
92%
(95% CI, 84%-97%)
95%
(95% CI, 83%-99%)
63%
(95% CI, 52%-73%)
NR84% at 10−5
At median time to MRD negativity of 6.6 months
71% at 10−5
after 8 cycles of therapy
Daratumumab-lenalidomide-bortezomib-dexamethasone without transplant
(DVRd)
NCT04775550Dara standard dosingb, bortezomib days 1, 8, and 15 for cycles 1-6 and then biweekly until completion of cycle 24. Len is administered days 1-21 and dex is administered weekly until cycle 2472No corresponding trial exists in NDMM90%
(95% CI, 68%-99%) at any point during study treatment period
NA50%
(95% CI, 27%-73%)
NA25%
(95% CI, 8%-49%)
NA50% at 10−5
25% at 10−6
after≥6 months follow-up, in those for whom MRD assessed
NA

Abbreviations: ASCT, autologous stem cell transplant; C, cycle; CRR, complete response rate; Dara (D), daratumumab; Dex (d), dexamethasone; E, elotuzumab; I, ixazomib; K, carfilzomib; Len (R), lenalidomide; Mel, melphalan; MRD; measurable residual disease; NA, not available; NCT, national clinical trial; NDMM, newly diagnosed multiple myeloma; NR, not reported; ORR, overall response rate; SMM, smoldering multiple myeloma; VGPR, very good partial response.

aElotozumab standard dosing is as follows; cycles 1 and 2: 10 mg/kg IV once a week on days 1, 8, 15, and 22 of a 28-day cycle, cycle 3 and beyond: 10 mg/kg IV once every 2 weeks on days 1 and 15 of a 28-day cycle until disease progression or unacceptable toxicity.

bDaratumumab standard dosing is weekly for first 8 weeks, every 2 weeks for next 16 weeks, and then monthly.

Bar graph depicting ORR and VGPR in SMM vs NDMM, with confidence intervals.
Figure 2.

Comparison of overall response rate and very good partial response (or greater) rate in corresponding SMM versus NDMM trials. Abbreviations: ASCT, autologous stem cell transplant; DKRd, daratumumab-carfilzomib-lenalidomide-dexamethasone; ERd, elotuzumab-lenalidomide-dexamethasone; IRd, ixazomib-lenalidomide-dexamethasone; KRd, carfilzomib-lenalidomide-dexamethasone; NDMM, newly diagnosed multiple myeloma; ORR, overall response rate; Rd, lenalidomide-dexamethasone; SMM, smoldering multiple myeloma; VGPR, very good partial response.

Of 45 total studies, 8 studies11,26,32,34,40,45,49,65 that had primary endpoint results available either on ClinicalTrials.gov or in abstract format by December 2021 (a cutoff date that was chosen to allow time for data analysis and publication) remained unpublished by April 2023. These 8 studies were then further described (Supplementary Appendix V); 7 out of 8 (87.5%)11,32,34,40,45,49,65 either did not meet their primary endpoint or did not clearly specify whether they met their primary endpoint.

The mode of progression to MM (biochemical vs clinical) was inconsistently described across studies, precluding quantitative analysis. Of 32 studies with results available, only 7 studies (21.9%)14,28,30,31,34,43,66 detailed the nature of progression (Supplementary Appendix VI).

Discussion

In this systematic review of clinical trials in SMM, we find that most SMM trials are single-arm, nonrandomized studies not adequately powered to assess clinically meaningful endpoints. Surrogate endpoints were the primary endpoint for almost all (97.7%) trials. Furthermore, nearly three-quarters of SMM studies with reported results failed to meet their primary endpoint or did not clearly report whether the primary endpoint was met. Seven studies that did not meet their endpoint have not been published at the time of our analysis, suggesting possible publication bias.

Importantly, when a regimen has been assessed in both SMM and NDMM contexts, response rates were similar, suggesting there is an absence of convincing data to support the hypothesis that SMM is “more responsive” to therapy than MM, even when considering the limitations of cross-trial comparison. We also find that most of these studies have at least a moderate risk of bias, further reinforcing the need for high-quality randomized studies in SMM.

Early intervention for SMM is based on the premise that lower-disease burden may be more responsive to treatment, and thus more curable, than symptomatic disease (ie, frank MM).10,26 So far, only one randomized trial in SMM has been powered to detect an improvement in overall survival; this trial is ongoing, with no results available yet (NCT03937635). For an asymptomatic condition such as SMM that has variable risk of progression to symptomatic disease (ie, some patients never progress to MM), it is unclear whether trial endpoints based on response or depth of response (eg, MRD negativity) are meaningful for patients. Notably, even in NDMM, the endpoints of ORR and PFS correlate poorly with OS.74,75 Furthermore, treatment is often accompanied by impactful side effects9 and is expensive, which can lead to financial toxicity for patients76; given that in the United States, lenalidomide can cost >$17 000 a month per patient and quadruplet regimens can range from $300 000 to $500 000/year, MM is one of the most expensive cancers to treat in terms of drug costs.18,77,78

We did not find convincing evidence that SMM is inherently more responsive to therapy than MM. Differences in study populations, dosing schema, and duration of therapy precluded formal statistical comparison. However, our findings of similar or lower response rates to therapies in SMM versus NDMM challenge existing dogma. One potential explanation could be that SMM clones, which can be slower growing, may be harder to eradicate than MM clones. This is consistent with emerging evidence positing that patients with monoclonal gammopathy of undetermined significance-like phenotype in SMM exhibit low disease progression rates with no difference in TTP between treatment versus observation.79 This is also congruent with real world evidence from the Australia and New Zealand Myeloma and Related Diseases Registry of 1818 patients with MM, which showed no difference in response rates between early- and advanced-stage MM.80

Universal treatment of high-risk SMM overtreats many who would do well with just observation alone. For example, even among patients with high-risk SMM in the observation arm of the E3A06 trial, at least 50% were free from progression at 3 years.14 Seminal work by Kyle et al defining the natural history of SMM over 15 years also showed that even at 15 years, 27% of patients remained without disease progression.81 Furthermore, there is a current lack of reporting of the breakdown of CRAB progression in the majority of SMM trials, making it difficult to ascertain what effects early treatment may avert. In a recent retrospective cohort study, most progression events were asymptomatic laboratory changes, rather than morbid events such as fracture or renal failure.82 To justify the treatment of these patients, a net benefit must therefore be demonstrated in a randomized trial powered to assess a clinically-meaningful endpoint.

Current risk stratification models poorly predict patients’ risk of progression and have poor concordance with each other.82,83 Evolving models that incorporate changes to lab markers over time and genomics may help improve prediction of which patients are more likely to progress and need therapy.84 While it is paramount to longitudinally collect biospecimens as part of SMM trials for future translational research, the benefits to an individual study participant for enrolling on a therapeutic trial may not outweigh the risks of the interventions being offered.

The lack of SMM studies with active surveillance as the control arm not only makes it difficult to know whether patients ultimately benefit from earlier treatment, but also to capture potential harms of early intervention, which is especially important for asymptomatic patients.9 This was highlighted by patient deaths in recent trials of intensive therapy for SMM: GEM-CESAR26 (7 deaths, 8% of patients; and 51% of patients with nonhematological grade ≥3 toxicities) and ASCENT13 (3 deaths, 3% of patients). The lack of a control arm in these studies makes it impossible to know whether these deaths were due to treatment, and whether these patients would have lived longer if they had instead been observed until progression. Similarly, while there is an emergence of targeted therapies suggesting promising activity, such as the recent Immuno-PRISM85 trial of teclistamab (n = 19, ORR = 100%, MRD negativity = 100% at 10−6 for the 8 evaluable patients), the lack of a control arm receiving no therapy presents 2 major unanswered counterfactuals. First, we do not know how these patients would have responded if treated at the time of progression to myeloma and second, due to the lack of a control arm receiving active surveillance, we do not know how patients would have done without therapy. Patients in the trial arm (n = 12) experienced grade 3 or higher toxicities such as neutropenia (n = 4, 33.3%), pancreatitis (n = 1, 8.3%), elevated ALT (n = 3, 25%). The duration of response is also unknown. In comparison, a prospective observational cohort study of 96 patients with SMM observed no non-myeloma-related deaths after 28 months median follow-up.86

As treatment options improve for MM, it becomes increasingly difficult to show an overall survival benefit in newly diagnosed MM, let alone SMM. Nevertheless, we believe that the bar for adopting early intervention in an asymptomatic population should be an improvement in overall survival or quality of life with the adoption of the intervention. As an example, in asymptomatic chronic lymphocytic leukemia, agents have shown a PFS benefit, but no OS benefit, and the field has not adopted early therapy despite these data.87 The neutral prior has been that precursor hematological conditions that are not causing morbidity in their current state should not be treated unless earlier treatment improves survival.

We recognize that each hematological malignancy is different. For instance, the discovery of a high-grade lymphoma, even if asymptomatic, represents a very different clinical scenario than the discovery of an asymptomatic low-grade follicular lymphoma. Given the long follow-up required to ascertain overall survival, using an endpoint that captures the nature of progression—such as distinguishing between asymptomatic lab changes and morbid end-organ damage—may be a useful intermediate. This approach is being explored in a prospective noninterventional SMM trial (NCT06212323).88

Limitations

Limitations of this study include that assessed studies may only have had limited follow-up and many were limited by sample size. Furthermore, although we do not make claims that one regimen is more effective than another, or more effective in one context than another, comparison across trials (between NDMM and SMM) should be considered descriptive only and therefore interpreted with caution. We analyzed all studies in SMM, including studies done prior to diagnostic reclassification and the use of advanced imaging, and these studies may not be relevant to modern day SMM.89 Prospective studies are needed to evaluate the natural history of contemporary SMM, such as the SPOTLIGHT study (NCT06212323).

Conclusions

Our study highlights the heterogeneity of clinical trials evaluating interventions in SMM and suggests there is a lack of evidence to suggest that treatment regimens are more effective in SMM than in NDMM. Randomized trials powered to assess clinically meaningful endpoints (especially overall survival), with active surveillance as their control arm, are needed to assess the risk-benefit relationship in contemporary patients with SMM.

Author Contributions

Apoorva Kakkilaya and Aaron Trando screened studies, extracted data, performed statistical analysis, and wrote the manuscript. Wade L. Smith and Muhammad Aziz conducted an initial search of studies and literature review. Edward R. Scheffer Cliff, Hira Mian, Samer Al Hadidi, Aaron M. Goodman, Ah-Reum Jeong, Amar H. Kelkar, David Russler-Germain, and Rajshekhar Chakraborty provided critical insight on the methodology, data analysis and preparation of the manuscript. Ghulam R. Mohyuddin conceptualized the question, screened studies, extracted data, analyzed data, and wrote the manuscript. All authors reviewed the manuscript and had final responsibility for the decision to submit for publication. Apoorva Kakkilaya and Ghulam R. Mohyuddin had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis

Funding

This work received no specific funding.

Conflict of Interest

The authors have no conflict of interests to declare other than the following: E.R.S.C. receives research funding from Arnold Ventures. A.H.K. receives research funding from CareDx. S.A.H. reported receiving consulting from Janssen and Sanofi. R.C. does consulting or participates in advisory boards for Janssen, Sanofi, and Adaptive Biotech and receives research funding from Genentech and AbbVie. H.M.: reports honoraria/consultancy from Celgene/BMS, Takeda, Sanofi, Amgen, Janssen, Pfizer, FORUS, and GSK; research funding from Janssen. G.R.M. has received honoraria for writing for MashupMD and Medscape, and his institution has received funds from Janssen for him being a site principal investigator on a trial.

Data Availability

All data included in this manuscript are publicly available from the original publications from which data were extracted.

References

1.

Thorsteinsdóttir
S
,
Gíslason
GK
,
Aspelund
T
, et al. .
Prevalence of smoldering multiple myeloma based on nationwide screening
.
Nat Med
.
2023
;
29
(
2
):
467
-
472
. https://doi.org/10.1038/s41591-022-02183-6

2.

Rajkumar
SV
,
Landgren
O
,
Mateos
MV.
Smoldering multiple myeloma
.
Blood
.
2015
;
125
(
20
):
3069
-
3075
. https://doi.org/10.1182/blood-2014-09-568899

3.

Chakraborty
R.
Survival in smouldering myeloma and symptomatic myeloma: is there an emerging Will Rogers phenomenon
?
Eur J Cancer
.
2022
;
172
:
234
-
236
. https://doi.org/10.1016/j.ejca.2022.05.041

4.

Kumar
S
,
Paiva
B
,
Anderson
KC
, et al. .
International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol
.
2016
;
17
(
8
):
e328
-
e346
. https://doi.org/10.1016/S1470-2045(16)30206-6

5.

Mohty
M
,
Avet-Loiseau
H
,
Harousseau
JL.
Requirements for operational cure in multiple myeloma
.
Blood
.
2021
;
138
(
16
):
1406
-
1411
. https://doi.org/10.1182/blood.2021012854

6.

Barlogie
B
,
Mitchell
A
,
van Rhee
F
, et al. .
Curing myeloma at last: defining criteria and providing the evidence
.
Blood
.
2014
;
124
(
20
):
3043
-
3051
. https://doi.org/10.1182/blood-2014-07-552059

7.

Kanth
P
,
Inadomi
JM.
Screening and prevention of colorectal cancer
.
BMJ
.
2021
;
374
:
n1855
. https://doi.org/10.1136/bmj.n1855

8.

Bretthauer
M
,
Løberg
M
,
Wieszczy
P
, et al. ;
NordICC Study Group
.
Effect of colonoscopy screening on risks of colorectal cancer and related death
.
N Engl J Med
.
2022
;
387
(
17
):
1547
-
1556
. https://doi.org/10.1056/NEJMoa2208375

9.

Chakraborty
R
,
Al Hadidi
S
,
Cliff
ERS
,
Mohyuddin
GR.
Is aggressive treatment of smoldering myeloma the path to curing myeloma
?
Blood Adv
.
2023
;
7
(
15
):
3932
-
3935
. https://doi.org/10.1182/bloodadvances.2023009658

10.

Kumar
SK
,
Alsina
M
,
Laplant
B
, et al. .
Fixed duration therapy with daratumumab, carfilzomib, lenalidomide and dexamethasone for high risk smoldering multiple myeloma-results of the ascent trial
.
Blood
.
2022
;
140
(
Supplement 1
):
1830
-
1832
. https://doi.org/10.1182/blood-2022-168930

11.

Nadeem
O
,
Redd
R
,
Stampleman
LV
, et al. .
A phase II study of daratumumab in patients with high-risk MGUS and low-risk smoldering multiple myeloma: first report of efficacy and safety
.
Blood
.
2019
;
134
(
Supplement_1
):
1898
-
1898
. https://doi.org/10.1182/blood-2019-129103

12.

Lomas
OC
,
Ghobrial
IM.
Clinical controversies in the management of smoldering multiple myeloma
.
Am Soc Clin Oncol Educ Book
.
2020
;
40
:
314
-
319
. https://doi.org/10.1200/edbk_278911

13.

Mateos
MV
,
Hernández
MT
,
Giraldo
P
, et al. .
Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma
.
N Engl J Med
.
2013
;
369
(
5
):
438
-
447
. https://doi.org/10.1056/NEJMoa1300439

14.

Lonial
S
,
Jacobus
S
,
Fonseca
R
, et al. .
Randomized trial of lenalidomide versus observation in smoldering multiple myeloma
.
J Clin Oncol
.
2020
;
38
(
11
):
1126
-
1137
. https://doi.org/10.1200/JCO.19.01740

15.

Goodman
AM
,
Kim
MS
,
Prasad
V.
Persistent challenges with treating multiple myeloma early
.
Blood
.
2021
;
137
(
4
):
456
-
458
. https://doi.org/10.1182/blood.2020009752

16.

Thorsteinsdottir
S
,
Kristinsson
SY.
The consultant’s guide to smoldering multiple myeloma
.
Hematology Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
551
-
559
. https://doi.org/10.1182/hematology.2022000355

17.

Mohyuddin
GR
,
Chakraborty
R
,
Cliff
ERS
,
Derman
BA.
Clinician preferences on treatment of smoldering myeloma: a cross-sectional survey
.
eClinicalMedicine
.
2023
;
65
:
102272
. https://doi.org/10.1016/j.eclinm.2023.102272

18.

Mohyuddin
GR
,
Ouchveridze
E
,
Goodman
A
,
Prasad
V.
The landscape of trials for smoldering multiple myeloma: endpoints, trial design, and lessons learnt
.
Leuk Lymphoma
.
2021
;
62
(
11
):
2793
-
2795
. https://doi.org/10.1080/10428194.2021.1938032

19.

Soomro
MA
,
Hoffman
J
,
Goodman
AM
,
Sborov
DW
,
Mohyuddin
GR.
Heterogeneity of enrolment criteria for ongoing smouldering myeloma trials
.
Br J Haematol
.
2022
;
197
(
6
):
e86
-
e88
. https://doi.org/10.1111/bjh.18102

20.

Medline
,
Embase
,
PsycInfo
.
All Clinical Trials
. CADTH Search Filters Database.
CADTH
;
2022
. hscclA.

21.

Liberati
A
,
Altman
DG
,
Tetzlaff
J
, et al. .
The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration
.
J Clin Epidemiol
.
2009
;
62
(
10
):
e1
-
e34
. https://doi.org/10.1016/j.jclinepi.2009.06.006

22.

U.S. Department of Health and Human Services Food and Drug Administration
.
2018
. Clinical-Trial-Endpoints-Approval-Cancer-Drugs-Biologics-final-guidance.pdf

23.

Higgins
JPT
,
Thomas
J
,
Chandler
J
,
Cumpston
M
,
Li
T
,
Page
MJ
,
Welch
VA
, editors.
Cochrane Handbook for Systematic Reviews of Interventions Version 6.2
.
Cochrane
; Updated
2021
. www.training.cochrane.org/handbook

24.

Higgins
JPT
,
Altman
DG
,
Gotzsche
PC
, et al. ;
Cochrane Bias Methods Group
.
The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials
.
BMJ
.
2011
;
343
(
oct18 2
):
d5928
. https://doi.org/10.1136/bmj.d5928

25.

Sterne
JA
,
Hernán
MA
,
Reeves
BC
, et al. .
ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions
.
BMJ
.
2016
;
355
:
i4919
. https://doi.org/10.1136/bmj.i4919

26.

Mateos
MV
,
Lopez
JM
,
Rodríguez-Otero
P
, et al. .
Curative strategy (GEM-CESAR) for high-risk smoldering myeloma (SMM): carfilzomib, lenalidomide and dexamethasone (KRD) as induction followed by HDT-ASCT, consolidation with KRD and maintenance with RD
.
Blood
.
2021
;
138
(
SUPPL 1
):
1829
. https://doi.org/10.1182/blood-2021-148423

27.

Brighton
TA
,
Khot
A
,
Harrison
SJ
, et al. .
Randomized, double-blind, placebo-controlled, multicenter study of siltuximab in high-risk smoldering multiple myeloma
.
Clin Cancer Res
.
2019
;
25
(
13
):
3772
-
3775
. https://doi.org/10.1158/1078-0432.CCR-18-3470

28.

Landgren
CO
,
Chari
A
,
Cohen
YC
, et al. .
Daratumumab monotherapy for patients with intermediate-risk or high-risk smoldering multiple myeloma: a randomized, open-label, multicenter, phase 2 study (CENTAURUS)
.
Leukemia
.
2020
;
34
(
7
):
1840
-
1852
. https://doi.org/10.1038/s41375-020-0718-z

29.

Witzig
TE
,
Laumann
KM
,
Lacy
MQ
, et al. .
A phase III randomized trial of thalidomide plus zoledronic acid versus zoledronic acid alone in patients with asymptomatic multiple myeloma
.
Leukemia
.
2013
;
27
(
1
):
220
-
225
. https://doi.org/10.1038/leu.2012.236

30.

D’Arena
G
,
Gobbi
PG
,
Broglia
C
, et al. ;
Gimema (Gruppo Italiano Malattie Ematologiche Dell'Adulto)
.
Pamidronate versus observation in asymptomatic myeloma: final results with long-term follow-up of a randomized study
.
Leuk Lymphoma
.
2011
;
52
(
5
):
771
-
775
. https://doi.org/10.3109/10428194.2011.553000

31.

Musto
P
,
Petrucci
MT
,
Bringhen
S
, et al. ;
GIMEMA (Italian Group for Adult Hematologic Diseases)/Multiple Myeloma Working Party and the Italian Myeloma Network
.
A multicenter, randomized clinical trial comparing zoledronic acid versus observation in patients with asymptomatic myeloma
.
Cancer
.
2008
;
113
(
7
):
1588
-
1595
. https://doi.org/10.1002/cncr.23783

32.

Horwitz
LJ
,
Faiman
B
,
Elson
P
, et al. .
A prospective, randomized, chemoprevention trial of celecoxib for high risk monoclonal gammopathy of undetermined significance and asymptomatic multiple myeloma
.
Blood
.
2012
;
120
(
21
):
5045
-
5045
. https://doi.org/10.1182/blood.v120.21.5045.5045

33.

Hjorth
M
,
Hellquist
L
,
Holmberg
E
, et al. .
Initial versus deferred melphalan-prednisone therapy for asymptomatic multiple myeloma stage I--a randomized study. Myeloma Group of Western Sweden
.
Eur J Haematol
.
1993
;
50
(
2
):
95
-
102
. https://doi.org/10.1111/j.1600-0609.1993.tb00148.x

34.

Musto
P
,
Bodenizza
CA
,
Falcone
A
, et al. .
Treatment with pamidronate in asymptomatic myeloma: results of a pair-matched analysis with historical controls
.
Blood
 
2000
;
96
(
11
):
290b
.

35.

Frigeri
F
,
De Renzo
A
,
Marceno
R
, et al. .
Effect of αIFN on unaggressive immunoproliferative disorders
.
Haematologica
.
1995
;
80
(
1
):
35
-
39
.

36.

Barlogie
B
,
Van Rhee
F
,
Shaughnessy Jr
JD
, et al. .
Seven-year median time to progression with thalidomide for smoldering myeloma: partial response identifies subset requiring earlier salvage therapy for symptomatic disease
.
Blood
.
2008
;
112
(
8
):
3122
-
3125
. https://doi.org/10.1182/blood-2008-06-164228

37.

Detweiler-Short
K
,
Hayman
S
,
Gertz
MA
, et al. .
Long-term results of single-agent thalidomide as initial therapy for asymptomatic (smoldering or indolent) myeloma
.
Am J Hematol
.
2010
;
85
(
10
):
737
-
740
. https://doi.org/10.1002/ajh.21821

38.

Lust
JA
,
Lacy
MQ
,
Zeldenrust
SR
, et al. .
Reduction in C-reactive protein indicates successful targeting of the IL-1/IL-6 axis resulting in improved survival in early stage multiple myeloma
.
Am J Hematol
.
2016
;
91
(
6
):
571
-
574
. https://doi.org/10.1002/ajh.24352

39.

Carlsten
M
,
Korde
N
,
Kotecha
R
, et al. .
Checkpoint inhibition of KIR2D with the monoclonal antibody IPH2101 induces contraction and hyporesponsiveness of NK cells in patients with myeloma
.
Clin Cancer Res
.
2016
;
22
(
21
):
5211
-
5222
. https://doi.org/10.1158/1078-0432.ccr-16-1108

40.

Munshi
NC
,
Abonour
R
,
Beck
JT
, et al. .
Early evidence of anabolic bone activity of BHQ880, a fully human anti-DKK1 neutralizing antibody: results of a phase 2 study in previously untreated patients with smoldering multiple myeloma at risk for progression
.
Blood
.
2012
;
120
(
21
):
331
-
331
. https://doi.org/10.1182/blood.v120.21.331.331

41.

Jagannath
S
,
Laubach
J
,
Wong
E
, et al. .
Elotuzumab monotherapy in patients with smouldering multiple myeloma: a phase 2 study
.
Br J Haematol
.
2018
;
182
(
4
):
495
-
503
. https://doi.org/10.1111/bjh.15384

42.

Nooka
AK
,
Wang
ML
,
Yee
AJ
, et al. .
Assessment of Safety and Immunogenicity of PVX-410 vaccine with or without lenalidomide in patients with smoldering multiple myeloma: a nonrandomized clinical trial
.
JAMA Oncol
.
2018
;
4
(
12
):
e183267
. https://doi.org/10.1001/jamaoncol.2018.3267

43.

Kazandjian
D
,
Hill
E
,
Dew
A
, et al. .
Carfilzomib, lenalidomide, and dexamethasone followed by lenalidomide maintenance for prevention of symptomatic multiple myeloma in patients with high-risk smoldering myeloma: a phase 2 nonrandomized controlled trial
.
JAMA Oncol
.
2021
;
7
(
11
):
1678
-
1685
. https://doi.org/10.1001/jamaoncol.2021.3971

44.

Wichert
S
,
Juliusson
G
,
Johansson
A
, et al. .
A single-arm, open-label, phase 2 clinical trial evaluating disease response following treatment with BI-505, a human anti-intercellular adhesion molecule-1 monoclonal antibody, in patients with smoldering multiple myeloma
.
PLoS ONE
2017
;
12
(
2
):
e0171205
. https://doi.org/10.1371/journal.pone.0171205

45.

Ghobrial
I
,
Caola
A
,
Henrick
P
, et al. .
Phase II trial of combination of elotuzumab, lenalidomide, and dexamethasone in high-risk smoldering multiple myeloma
.
Haematologica
.
2017
;
102
:
317
.

46.

Mailankody
S
,
Salcedo
M
,
Tavitian
E
, et al. .
Ixazomib and dexamethasone in high risk smoldering multiple myeloma: a clinical and correlative pilot study
.
Leuk Lymphoma
.
2022
;
63
(
11
):
2760
-
2761
. https://doi.org/10.1080/10428194.2022.2095626

47.

Manasanch
EE
,
Han
G
,
Mathur
R
, et al. .
A pilot study of pembrolizumab in smoldering myeloma: report of the clinical, immune, and genomic analysis
.
Blood Adv
.
2019
;
3
(
15
):
2400
-
2408
. https://doi.org/10.1182/bloodadvances.2019000300

48.

Manasanch
EE
,
Jagannath
S
,
Lee
HC
, et al. .
A multicenter phase II single arm trial of isatuximab in patients with high risk smoldering multiple myeloma (HRSMM)
.
Blood
.
2019
;
134
(
Supplement_1
):
3116
-
3116
. https://doi.org/10.1182/blood-2019-123205

49.

Nadeem
O
,
Redd
RA
,
Prescott
J
, et al. .
A phase II trial of the combination of ixazomib, lenalidomide, and dexamethasone in high-risk smoldering multiple myeloma
.
Blood
.
2021
;
138
(
Supplement 1
):
2749
-
2749
. https://doi.org/10.1182/blood-2021-149787

50.

Dimopoulos
MA
,
Voorhees
PM
,
Goldschmidt
H
, et al. .
Subcutaneous daratumumab (DARA SC) versus active monitoring in patients (pts) with high-risk smoldering multiple myeloma (SMM): randomized, open-label, phase 3 AQUILA study
.
J Clin Oncol
.
2022
;
40
(
16
). https://doi.org/10.1200/JCO.2022.40.16_suppl.TPS8075

51.

NCT. Personalized vaccine in treating patients with smoldering multiple myeloma
.Trial registry record; Clinical trial protocol; 
2018
https://clinicaltrials.gov/study/NCT03631043 

52.

NCT
Carfilzomib, lenalidomide and dexamethasone versus lenalidomide and dexamethasone in high-risk SMM (HO147SMM). Trial registry record; Clinical trial protocol;
2018
https://www.clinicaltrials.gov/study/NCT03673826

53.

NCT
Denosumab for smoldering multiple myeloma
. Trial registry record; Clinical trial protocol;
2019
https://clinicaltrials.gov/study/NCT03839459

54.

NCT
.
Lenalidomide, and dexamethasone with or without daratumumab in treating patients with high-risk smoldering myeloma
. Trial registry record; Clinical trial protocol;
2019
. https://clinicaltrialsgov/show/NCT03937635

55.

Ludwig
H
,
Willenbacher
W.
Denosumab for high-risk SMM and slim-CRABpositive, early myeloma patients-a randomized, placebo-controlled, phase-II-trial “DEFENCE” (DEnosumab For the rEductioN of the smoldering myeloma transformatioN inCidence ratE)
.
Memo
.
2018
;
11
(
1
):
S24
. https://doi.org/10.1007/s12254-018-0401-5

56.

Ghobrial
I
,
Rodríguez-Otero
P
,
Koh
Y
, et al. .
P-137: ITHACA, a randomized multicenter phase 3 study of isatuximab in combination with lenalidomide and dexamethasone in high-risk smoldering multiple myeloma: safety run-in preliminary results
.
Clin Lymphoma Myeloma Leuk
.
2021
;
21
:
S109
-
S110
. https://doi.org/10.1016/s2152-2650(21)02264-3

57.

Nadeem
O
,
Redd
RA
,
Prescott
J
, et al. .
B-prism (precision intervention smoldering myeloma): a phase II trial of combination of daratumumab, bortezomib, lenalidomide and dexamethasone in high-risk smoldering multiple myeloma
.
Blood
.
2021
;
138
(
Supplement 1
):
4782
-
4782
. https://doi.org/10.1182/blood-2021-148023

58.

NCT
.
Investigation of metformin for the prevention of progression of precursor multiple myeloma
. Trial registry record; Clinical trial protocol;
2021
. https://clinicaltrialsgov/show/NCT04850846

59.

NCT
.
Iberdomide alone or in combination with dexamethasone for the treatment of intermediate- or high-risk smoldering multiple myeloma
. Trial registry record; Clinical trial protocol;
2021
. https://clinicaltrialsgov/show/NCT04776395

60.

NCT
Immunomodulatory drugs (lenalidomide with or without pomalidomide) in combination with a corticosteroid drug (dexamethasone) for the treatment of multiple myeloma
. Trial registry record; Clinical trial protocol;
2022
https://clinicaltrials.gov/study/NCT05288062

61.

NCT
Leflunomide for the treatment of high-risk smoldering multiple myeloma in African-American and European-American patients
. Trial registry record; Clinical trial protocol;
2022
https://clinicaltrials.gov/study/NCT05014646

62.

NCT
Subcutaneous daratumumab, once weekly carfilzomib, and dexamethasone (DKd) in patients with high-risk smoldering multiple myeloma
. Trial registry record; Clinical trial protocol;
2022
https://clinicaltrials.gov/study/NCT04933539

63.

NCT
A phase 1 with extension cohort, single arm, single center, open label trial of belantamab mafodotin for the treatment of high-risk smoldering multiple myeloma
. Trial registry record; Clinical trial protocol;
2022
https://clinicaltrials.gov/study/NCT05055063

64.

NCT
.
Immuno-PRISM (PRecision Intervention Smoldering Myeloma)
. Trial registry record; Clinical trial protocol;
2022
. https://clinicaltrialsgov/show/NCT05469893

65.

NCT
.
Study on the anti-tumor activity, safety and pharmacology of IPH2101 in patients with smoldering multiple myeloma
. Trial registry record; Clinical trial protocol;
2010
. https://clinicaltrialsgov/show/NCT01222286

66.

Martín
A
,
García-Sanz
R
,
Hernández
J
, et al. .
Pamidronate induces bone formation in patients with smouldering or indolent myeloma, with no significant anti-tumour effect
.
Br J Haematol
.
2002
;
118
(
1
):
239
-
242
. https://doi.org/10.1046/j.1365-2141.2002.03549.x

67.

Zonder
JA
,
Crowley
J
,
Hussein
MA
, et al. .
Lenalidomide and high-dose dexamethasone compared with dexamethasone as initial therapy for multiple myeloma: a randomized Southwest Oncology Group trial (S0232)
.
Blood
 
2010
;
116
(
26
):
5838
-
5841
. https://doi.org/10.1182/blood-2010-08-303487

68.

Gay
F
,
Musto
P
,
Rota-Scalabrini
D
, et al. .
Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): a randomised, open-label, phase 2 trial
.
Lancet Oncol.
2021
;
22
(
12
):
1705
-
1720
https://doi.org/10.1016/S1470-2045(21)00535-0

69.

Dimopoulos
MA
,
Richardson
PG
,
Bahlis
NJ
, et al. .
Addition of elotuzumab to lenalidomide and dexamethasone for patients with newly diagnosed, transplantation ineligible multiple myeloma (ELOQUENT-1): an open-label, multicentre, randomised, phase 3 trial
.
Lancet Haematol.
2022
;
9
(
6
):
e403
-
e414
. https://doi.org/10.1016/S2352-3026(22)00103-X

70.

Facon
T
,
Venner
CP
,
Bahlis
NJ
, et al. .
Oral ixazomib, lenalidomide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma
.
Blood
2021
;
137
(
26
):
3616
-
3628
. https://doi.org/10.1182/blood.2020008787

71.

Landgren
O
,
Hultcrantz
M
,
Diamond
B
, et al. .
Safety and effectiveness of weekly carfilzomib, lenalidomide, dexamethasone, and daratumumab combination therapy for patients with newly diagnosed multiple myeloma
.
JAMA Oncol.
 
2021
;
7
(
6
):
862
. https://doi.org/10.1001/jamaoncol.2021.0611

72.

Nadeem
O
,
Redd
R
,
Mo
CC
, et al. .
B-PRISM (precision intervention smoldering myeloma): a phase II trial of combination of daratumumab, bortezomib, lenalidomide, and dexamethasone in high-risk smoldering multiple myeloma
.
J Clin Oncol.
 
2022
;
40
(
16_suppl
):
8040
. https://doi.org/10.1200/JCO.2022.40.16_suppl.8040

73.

Van Oekelen
O
,
Birrer
N
,
Wesson
W
, et al. .
Characteristics of measurable residual disease assessment in myeloma: a review of clinical trials from 2015–2020
.
Blood Cancer J
.
2022
;
12
(
11
):
155
. https://doi.org/10.1038/s41408-022-00750-1

74.

Etekal
T
,
Koehn
K
,
Sborov
DW
, et al. .
Time‐to‐event surrogate end‐points in multiple myeloma randomised trials from 2005 to 2019: a surrogacy analysis
.
Br J Haematol
.
2023
;
200
(
5
):
587
-
594
. https://doi.org/10.1111/bjh.18568

75.

Mainou
M
,
Madenidou
AV
,
Liakos
A
, et al. .
Association between response rates and survival outcomes in patients with newly diagnosed multiple myeloma. A systematic review and meta-regression analysis
.
Eur J Haematol
.
2017
;
98
(
6
):
563
-
568
. https://doi.org/10.1111/ejh.12868

76.

Ouchveridze
E
,
Banerjee
R
,
Desai
A
, et al. .
Financial toxicity in hematological malignancies: a systematic review
.
Blood Cancer J
.
2022
;
12
(
4
):
74
. https://doi.org/10.1038/s41408-022-00671-z

77.

Beechinor
RJ
,
Mohyuddin
GR
,
Mitchell
DE
,
Aaron
D
,
Mahmoudjafari
Z.
The story of the development of generic lenalidomide: how one company thwarted the Waxman-Hatch Act to generate billions of dollars in revenue
.
J Cancer Policy
.
2023
;
38
:
100446
. https://doi.org/10.1016/j.jcpo.2023.100446

78.

Rajkumar
SV.
Value and cost of myeloma therapy
.
Am Soc Clin Oncol Educ Book
.
2018
;
38
:
662
-
666
. https://doi.org/10.1200/EDBK_200867

79.

Burgos
L
,
Tamariz-Amador
LE
,
Puig
N
, et al. ; on behalf of the PETHEMA/GEM Cooperative Group.
Definition and clinical significance of the monoclonal gammopathy of undetermined significance–like phenotype in patients with monoclonal gammopathies
.
J Clin Oncol
.
2023
;
41
(
16
):
3019
-
3031
. https://doi.org/10.1200/jco.22.01916

80.

Ho
PJ
,
Moore
EM
,
Wellard
C
, et al. .
The impact of S-Li-M criteria in myeloma in a real-life population: patient & disease characteristics, treatment and outcomes from the Australian and New Zealand Myeloma and Related Diseases Registry (MRDR)
.
Blood
.
2020
;
136
(
Supplement 1
):
30
-
31
. https://doi.org/10.1182/blood-2020-141511

81.

Kyle
RA
,
Remstein
ED
,
Therneau
TM
, et al. .
Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma
.
N Engl J Med
.
2007
;
356
(
25
):
2582
-
2590
. https://doi.org/10.1056/NEJMoa070389

82.

Mellgard
G
,
Gilligan
M
,
Cliff
ERS
, et al. .
Risk stratification models overestimate progression risk in contemporary patients with smoldering multiple myeloma
.
HemaSphere
.
2024
;
8
(
3
):
e61
. https://doi.org/10.1002/hem3.61

83.

Hill
E
,
Dew
A
,
Morrison
C
, et al. .
Assessment of discordance among smoldering multiple myeloma risk models
.
JAMA Oncol
.
2021
;
7
(
1
):
132
-
134
. https://doi.org/10.1001/jamaoncol.2020.5585

84.

Bustoros
M
,
Sklavenitis-Pistofidis
R
,
Park
J
, et al. .
Genomic profiling of smoldering multiple myeloma identifies patients at a high risk of disease progression
.
J Clin Oncol
.
2020
;
38
(
21
):
2380
-
2389
. https://doi.org/10.1200/JCO.20.00437

85.

Nadeem
O
,
Magidson
S
,
Midha
S
, et al. .
Immuno-PRISM: a randomized phase II platform study of bispecific antibodies in high-risk smoldering myeloma
.
Blood
.
2023
;
142
(
Supplement 1
):
206
-
206
. https://doi.org/10.1182/blood-2023-177954

86.

Wennmann
M
,
Goldschmidt
H
,
Mosebach
J
, et al. .
Whole‐body magnetic resonance imaging plus serological follow‐up for early identification of progression in smouldering myeloma patients to prevent development of end‐organ damage
.
Br J Haematol
.
2022
;
199
(
1
):
65
-
75
. https://doi.org/10.1111/bjh.18232

87.

Langerbeins
P
,
Zhang
C
,
Robrecht
S
, et al. .
The CLL12 trial: ibrutinib vs placebo in treatment-naïve, early-stage chronic lymphocytic leukemia
.
Blood
.
2022
;
139
(
2
):
177
-
187
. https://doi.org/10.1182/blood.2021010845

88.

Cliff
ERS
,
Mohyuddin
GR.
Overall survival as a primary end point in multiple myeloma trials
.
Nat Rev Clin Oncol
.
2022
;
19
(
9
):
565
-
566
. https://doi.org/10.1038/s41571-022-00665-7

89.

Chakraborty
R.
Survival in smouldering myeloma and symptomatic myeloma: is there an emerging Will Rogers phenomenon
?
Eur J Cancer
.
2022
;
172
:
234
-
236
. https://doi.org/10.1016/j.ejca.2022.05.041

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.