The development of new drugs and subsequent novel combinations for the treatment of newly diagnosed multiple myeloma (NDMM) has resulted in a plethora of treatment options that can make the choice of initial induction therapy a challenge. A greater understanding of both patient- and disease-specific factors can provide a personalized approach to help design a treatment course. Historically, the choice of an induction regimen has been tethered to an initial impression of transplant eligibility at the time of diagnosis. As more effective and better-tolerated induction regimens have emerged, there has been increasing overlap in the induction strategies used for all patients with NDMM, which increasingly provide the ultimate goal of deep and durable remissions. The current treatment options and strategies for the management of NDMM are evaluated using the best available data to provide a rationale for these decisions.

Learning Objectives

  • Define the treatment options available to patients with newly diagnosed multiple myeloma (NDMM)

  • Understand the strategy for choosing treatment for patients with NDMM

A 63-year-old previously healthy African American woman presented with progressive dyspnea on exertion and lower back pain. Her initial laboratory evaluation revealed a hemoglobin of 7.4  g/dL, a calcium level of 9.6  mg/dL, a creatinine level of 1.7  mg/dL, and an elevated lactate dehydrogenase level. X-ray of the patient's lumbar spine demonstrated multiple lytic lesions and a moderate compression fracture at L2. Due to the suspicion for multiple myeloma, further diagnostic testing was performed. A serum protein electrophoresis showed a monoclonal paraprotein level of 3.7  g/dL that was identified as IgG λ by serum immunofixation. Quantitative immunoglobulins demonstrated a markedly elevated IgG level at 6730  mg/dL and low IgM and IgA levels. Serum free λ level was elevated at 335  mg/L with a markedly abnormal κ/λ ratio of 0.04. Urine protein electrophoresis revealed a free λ level of 355  mg/24 hours. The β2-microglobulin concentration was 4.1  mg/L, and her albumin was 3.2  g/dL. A positron emission tomography/computed tomography scan demonstrated multiple hypermetabolic osseous lesions without extramedullary disease. A bone marrow aspirate and biopsy specimen showed a hypercellular marrow with plasma cells accounting for 80% of the cellularity with λ restriction by flow cytometry. Fluorescence in situ hybridization revealed monosomy 13 in 38 of 50 cells. She received 1 unit of packed red blood cell transfusion, which improved her dyspnea.

Multiple myeloma is a neoplastic plasma cell disorder, characterized by clonal proliferation of malignant plasma cells in the bone marrow and usually a monoclonal protein in the blood and/or urine. It is associated with end-organ damage consisting of anemia, renal insufficiency, bone lesions, and/or hypercalcemia, and the International Myeloma Working Group updated the definition to include validated biomarkers present in patients without end-organ damage but associated with an 80% risk of progression to active disease with the first 2 years since diagnosis (clonal bone marrow plasma cell percentage ≥60%, involved/uninvolved serum free light chain ratio ≥100, or >1 focal lesion on magnetic resonance imaging or positron emission tomography/computed tomography) (Table 1).1 

Table 1.

Multiple myeloma diagnostic criteria

Clonal bone marrow plasma cells ≥10% 
OR  
Biopsy-proven plasmacytoma  
AND  
“CRAB” Hypercalcemia: Serum calcium >11  mg/dL or >1  mg/dL above ULN 
 Renal insufficiency: Serum creatinine >2  mg/dL or CrCl <40  mL/min 
 Anemia: Hemoglobin <10  g/dL or >2  g/dL below LLN 
 Bone lesions: ≥1 osteolytic lesion (on WBLDCT or PET scan) 
OR  
“SLiM” criteria ≥60% clonal bone marrow plasma cells 
 Involved to uninvolved FLC ratio ≥100 and involved FLC ≥10  mg/dL 
 >1 focal lesion on MRI ≥5  mm in size 
Clonal bone marrow plasma cells ≥10% 
OR  
Biopsy-proven plasmacytoma  
AND  
“CRAB” Hypercalcemia: Serum calcium >11  mg/dL or >1  mg/dL above ULN 
 Renal insufficiency: Serum creatinine >2  mg/dL or CrCl <40  mL/min 
 Anemia: Hemoglobin <10  g/dL or >2  g/dL below LLN 
 Bone lesions: ≥1 osteolytic lesion (on WBLDCT or PET scan) 
OR  
“SLiM” criteria ≥60% clonal bone marrow plasma cells 
 Involved to uninvolved FLC ratio ≥100 and involved FLC ≥10  mg/dL 
 >1 focal lesion on MRI ≥5  mm in size 

FLC, free light chain; MRI, magnetic resonance imaging; PET, positron emission topography; WBLDCT, whole-body low-dose computed tomography.

The emergence of novel agents, drug combinations, and therapeutic strategies has significantly improved outcomes in the past decade. Historical approaches to the management of newly diagnosed multiple myeloma (NDMM) have been categorized with a distinction between those patients deemed to be eligible or ineligible for high-dose chemotherapy and autologous stem cell transplantation at the time of diagnosis. Induction regimens were subsequently chosen based on a variety of patient- and disease-specific details. With limited treatment options and strategies available, the historic approach to the transplant-ineligible patient has largely been one of a “less is more” strategy by choosing fewer combined agents or by administering 2 to 3 drugs at lower doses, potentially sacrificing efficacy for safety. For transplant-eligible patients, the preferred intensity of induction has ranged widely from doublet regimens and transplant with modest clinical responses to Total Therapy regimens with deep responses despite high potential toxicity.2  The introduction of multiple agents and therefore multiple combinations allows the treatment intensity pendulum to continue to swing as we deepen our understanding of what our goals of therapy are. The question remains whether the disease course is a marathon that we aspire to finish with a slow and steady course or perhaps the treatment approach be aggressive from the time of diagnosis and use our best tools from the outset.

More therapeutic options have led to dramatic improvements in overall response rates and the depth of response, making the choice of the induction regimen solely based on transplant eligibility less relevant. It is well established that the depth of response is one of the most important prognostic factors in multiple myeloma (MM) and that the achievement of deep remissions represents a therapeutic goal for a large percentage of patients with MM.3  Ultimately, the optimal treatment approach for both fit and unfit patients with NDMM should incorporate the same strategy: provide a good balance of safety and efficacy while preserving the goal of eradicating as much disease as possible to provide deep and durable remissions, if not eventual cure.

The choice of an induction regimen for a patient with NDMM must take patient-specific, disease-specific, and therapy-related factors into consideration (Table 2). Importantly, shared decision- making between the physician and patient is paramount to meet differing treatment preferences and needs. The general approach to the management of any patient with NDMM is to administer initial induction therapy over a period of 4 to 6 cycles prior to possible high dose chemotherapy and autologous stem cell transplant (HDT-ASCT) or, alternatively, store stem cells and defer transplant. Patients who do not pursue HDT-ASCT, either because of fitness or choice, ideally receive continuous therapy until disease progression with the currently available data. Patients who undergo HDT-ASCT ideally receive maintenance therapy until progression.

Table 2.

Factors to be considered when choosing induction therapy

Patient relatedDisease relatedTreatment related
Age/Fitness Bone marrow disease burden Access to standard-of-care therapies 
Caregiver support Extramedullary disease Costs and copays 
Comorbidities Molecular cytogenetics/genomics Route of administration 
Compliance CRAB symptoms Clinical trial availability 
Lifestyle preferences R-ISS/ISS Toxicity 
Performance status/Frailty   
Patient relatedDisease relatedTreatment related
Age/Fitness Bone marrow disease burden Access to standard-of-care therapies 
Caregiver support Extramedullary disease Costs and copays 
Comorbidities Molecular cytogenetics/genomics Route of administration 
Compliance CRAB symptoms Clinical trial availability 
Lifestyle preferences R-ISS/ISS Toxicity 
Performance status/Frailty   

ISS, International Staging System; R-ISS, Revised International Staging System.

The menu of choices available to incorporate into the optimal induction therapy for fit patients with NDMM continues to grow. Recent trials have incorporated 3- and 4-drug combinations, using an immunomodulatory agent (IMiD), a proteasome inhibitor (PI), and a corticosteroid as a backbone. Several studies also support the use of minimal residual disease (MRD) for monitoring the response in MM because of its prognostic value. A recent meta-analysis demonstrated that the achievement of undetectable MRD improved progression-free survival (PFS) (hazard ratio [HR], 0.33) and overall survival (OS) (HR, 0.45) in comparison with the presence of MRD. Its prognostic impact was seen even in those patients with high-risk features such as high-risk cytogenetic abnormalities, demonstrating the importance of a deep response to therapy.4 

Three-drug combinations

The combination of lenalidomide, bortezomib, and dexamethasone (VRd) has been a recent standard of care for initial treatment for NDMM. The SWOG S0777 study compared VRd with lenalidomide and dexamethasone, showing definitively that VRd was associated with an improved overall response rate, very good partial response (VGPR) or better rate, PFS, and OS.5  The combination of VRd with or without initial transplantation was also studied and confirmed the significant initial efficacy of this regimen in the IFM 2009 and DETERMINATION phase 3 studies.6,7 

As MRD appears to be increasingly important as a goal of therapy to improve long-term outcomes, multiple other strategies to achieve MRD negativity have been evaluated. Gay and colleagues have presented results of the FORTE study, whose initial objectives were to compare induction with carfilzomib, lenalidomide, and dexamethasone (KRd) vs carfilzomib, cyclophosphamide, and dexamethasone (KCd) and to evaluate whether HDT-ASCT remained important in patients who received KRd induction. A second randomization assigned patients to receive either lenalidomide maintenance or doublet carfilzomib and lenalidomide (KR) maintenance.8,9  After KRd induction followed by HDT-ASCT and KRd consolidation (KRd-ASCT), the VGPR rate was 89% and the MRD-negative rate was 58%. A third arm of the study evaluated KRd for 12 cycles without transplant (KRd12) and demonstrated a VGPR rate of 90% and an MRD- negative rate of 54%, similar to what was seen in the KRd-ASCT arm. Survival analyses after a median follow-up of nearly 4 years subsequently have shown a significant improvement in PFS in the KRd-ASCT arm when compared to the KRd12 and KCd-ASCT arms (not reached vs 57 vs 53 months; KRd-ASCT vs KCd-ASCT: HR 0.53, P  <  .001; KRd-ASCT vs KRd12: HR 0.64, P  =  .0023; KRd12 vs KCd-ASCT: HR 0.82, P  =  .262) and was seen in all subgroups. The 3-year OS was 90% in both the KRd-ASCT and KRd12 arms vs 83% with KCd. These data further solidify the strategy of IMiD-PI-corticosteroid combinations and demonstrate the importance of deep responses to translate into improved outcomes. We now have several prospective studies solidifying an advantage when incorporating a proteasome inhibitor and an IMiD together as part of an induction regimen. Prolonged induction paired with consolidation strategies and maintenance provide deep responses and a PFS benefit and should be considered in any fit patient with NDMM.

Four-drug regimens

The emergence of CD38 monoclonal antibodies into frontline therapy offers an opportunity to improve upon the successes of 3-drug combinations. CD38-targeting monoclonal antibodies have shown marked activity and a favorable toxicity profile when first evaluated as a single agent in heavily pretreated patients with MM. The early experience with quadruplet regimens in NDMM was evaluated in 2 randomized phase 3 trials: ALCYONE and CASSIOPEIA.10,11  The ALCYONE study evaluated bortezomib-melphalan-prednisone with or without daratumumab in transplant-ineligible patients with NDMM. The primary end point of improved PFS was met when patients treated in the daratumumab arm achieved a deeper response and a 3-year PFS of 50% vs 18% compared with patients who were treated with bortezomib-melphalan-prednisone alone. There was a 40% reduction in the risk of death for those patients who received the quadruplet regimen, the first time that an improvement in OS was seen with a daratumumab-based combination therapy.10  The CASSIOPEIA study demonstrated that the addition of daratumumab to bortezomib-thalidomide-dexamethasone before and after transplantation improved the depth or response, PFS, and OS. A second randomization evaluated daratumumab monotherapy vs observation in those patients who achieved at least a partial response. The primary end point of the study was stringent complete response (sCR) after consolidation, and secondary end points included MRD-negative rate, PFS, and OS.11  This study was the first to show that the addition of a CD38 targeting monoclonal antibody to induction and a consolidation regimen is highly effective without adding significant additional toxicity.

The GRIFFIN study was a randomized phase 2 study designed to evaluate the addition of daratumumab to RVd (D-RVd) in transplant-eligible patients. The primary end point was sCR by the end of consolidation with a secondary end point of PFS.12  The primary end point demonstrated improved sCR rates with D-RVd vs RVd (67.0% vs 48.0%; P  =  .0079) in the final analysis after a median follow-up of 49.6 months. MRD negativity rates were also higher in the D-RVd group vs control (10-5: 64.4% vs 30.1%, P  <  .0001; 10-6: 35.6% vs 15.5%, P  =  .0013). Sustained MRD negativity rates were higher in the D-RVd vs RVd groups after 12 months in all relevant subgroups, including high-risk patients, associated with improved PFS for all intention-to-treat patients.13  Estimated 48-month PFS rates were 87.2% for D-RVd and 70.0% for RVd. The phase 3 randomized PERSEUS trial that compared these same 2 arms and evaluated if daratumumab can be discontinued in patients with sustained MRD negativity is ongoing. The MASTER, MANHATTAN, and IFM2018-04 trials will all evaluate the role of daratumumab with KRd induction in patients with NDMM, the option of an MRD response-adapted approach, MRD negativity rates, and particularly the use of daratumumab with KRd induction in high-risk patients, the last of which is a glaring unmet need.14-16  Although no trial compares the strategy of CD38 antibody-based therapy at induction vs the time of first relapse, many favor the use of the best drugs, including daratumumab, for first-line therapy to induce the deepest response possible to translate into the best clinical outcomes. It is becoming increasingly clear that CD38 monoclonal antibodies play a significant role in achieving unprecedented responses in a patient with NDMM, and it is expected that 4-drug regimens will likely become a new standard of care (Figure 1, Table 3).

Figure 1.

Treatment algorithm for newly diagnosed multiple myeloma. ASCT, autologous stem cell transplant; d, dexamethasone; D, daratumumab; R, lenalidomide; V, bortezomib.

Figure 1.

Treatment algorithm for newly diagnosed multiple myeloma. ASCT, autologous stem cell transplant; d, dexamethasone; D, daratumumab; R, lenalidomide; V, bortezomib.

Close modal
Table 3.

Summary of select data from randomized trials for induction therapy for transplant-eligible NDMM

ReferencePatients: total/arms, No.Median follow-up, moMedian age, yRegimenPFSOS
GIMEMA62  236 vs 238 43 57 VTd vs Td 3-y PFS 68% vs 56% 3-y 86% vs 84% 
IFM2013-0463  169 vs 169 NR 59.5 VTd vs VCd NR NR 
EMNO2/HO9523  495 vs 702 60.3 58 VCd-ASCT vs
VCd-VMP
 
Median PFS 56.7 vs 41.9 mo (HR, 0.73, P  =  .0001) 5-y OS 75.1% vs 71.6% (HR, 0.90; 95% CI 0.71-1.13; P  =  .35) 
STAMINA24 
 
247 vs 254 vs 257 38 56 ASCT-R vs
ASCT-VRd-R vs
Tandem ASCT-R
 
38-mo PFS rate:
ASCT/ASCT-R: 58.5% (95% CI, 51.7%-64.6%)
ASCT-VRd-R: 57.8% (95% CI, 51.4%-63.7%)
ASCT-R: 53.9% (95% CI, 47.4%-60%) 
OS:
ASCT+ R: 81.8% (95% CI, 76.2%-86.2%)
ASCT-VRd-R: 85.4% (95% CI, 80.4%-89.3%)
ASCT-R: 83.7% (95% CI, 78.4%-87.8%)
 
IFM 200925 
 
350 vs 350 44 59.5 RVd-ASCT-R
RVd-R 
Median 50 vs 36 mo
(HR, 0.65; 95% CI, 0.53-0.80; P  <  .001) 
4 y: 81% vs 82%
(HR, 1.16; 95% CI, 0.80-1.68; P  =  .87) 
DETERMINATION7  365 vs 357 76 56 RVd-ASCT-R
RVd-R 
Median: 67.5 vs 46.2 mo (HR, 1.53; 95% CI, 1.23-1.91; P  <  .001) 5 y: 79% vs 81% (HR, 1.10; 95% CI, 0.73-1.65; P  >  .99) 
FORTE8,9 158 vs 157 vs 159 50.9 57 KRd-ASCT-KRd
KRd12
KCd-ASCT-KCd 
3 y: 68.8% vs 68.5% vs 67.2% (P  =  .86)
4 y: 69% (KRd-ASCT) vs 56% (KRd12) vs 51% (KCd-ASCT)
HR for KRd-ASCT vs KCd-ASCT, 0.54; 95% CI, 0.38-0.78; P  =  .0008
HR for KRd-ASCT vs KRd12, 0.61; 95% CI, 0.43-0.88; P  =  .0084 
4 y: 86% (KRd-ASCT) vs 85% (KRd12) vs 76% (KCd-ASCT) 
GRIFFIN12  104 vs 103 22.1
 
60 (27%> 65 y) D-RVd-ASCT-DR
RVd-ASCT-R 
3 y: 88.9% vs 81.2% (HR, 0.46; 95% CI, 0.21-1.01) 3 y: 92.6% vs 92.2% (HR, 0.90; 95% CI, 0.32-2.57) 
CASSIOPEIA11  543 vs 542 18.8 58.5 R1: D-VTd-ASCT-D-VTd
VTd-ASCT-VTd
R2: D maint vs obs 
Median: NR in either arm (HR, 0.47; 95% CI, 0.33-0.67; P  <  .0001) Median: NR in either arm (HR, 0.43; 95% CI, 0.23-0.80) 
GMMG-HD652 
 
139 vs 141 vs 137 vs 142
 
49.8 59 RVd-ASCT-R
RVd-ASCT-EloR
Elo-RVd-ASCT-R
Elo-RVd-ASCT-EloR 
3 y: 68.8% vs 68.5% vs 66.2% vs 67.2% (P  =  .86) 3 y: 89.4% vs 89.1% vs 92.5% vs 89.7% (P  =  .43) 
ReferencePatients: total/arms, No.Median follow-up, moMedian age, yRegimenPFSOS
GIMEMA62  236 vs 238 43 57 VTd vs Td 3-y PFS 68% vs 56% 3-y 86% vs 84% 
IFM2013-0463  169 vs 169 NR 59.5 VTd vs VCd NR NR 
EMNO2/HO9523  495 vs 702 60.3 58 VCd-ASCT vs
VCd-VMP
 
Median PFS 56.7 vs 41.9 mo (HR, 0.73, P  =  .0001) 5-y OS 75.1% vs 71.6% (HR, 0.90; 95% CI 0.71-1.13; P  =  .35) 
STAMINA24 
 
247 vs 254 vs 257 38 56 ASCT-R vs
ASCT-VRd-R vs
Tandem ASCT-R
 
38-mo PFS rate:
ASCT/ASCT-R: 58.5% (95% CI, 51.7%-64.6%)
ASCT-VRd-R: 57.8% (95% CI, 51.4%-63.7%)
ASCT-R: 53.9% (95% CI, 47.4%-60%) 
OS:
ASCT+ R: 81.8% (95% CI, 76.2%-86.2%)
ASCT-VRd-R: 85.4% (95% CI, 80.4%-89.3%)
ASCT-R: 83.7% (95% CI, 78.4%-87.8%)
 
IFM 200925 
 
350 vs 350 44 59.5 RVd-ASCT-R
RVd-R 
Median 50 vs 36 mo
(HR, 0.65; 95% CI, 0.53-0.80; P  <  .001) 
4 y: 81% vs 82%
(HR, 1.16; 95% CI, 0.80-1.68; P  =  .87) 
DETERMINATION7  365 vs 357 76 56 RVd-ASCT-R
RVd-R 
Median: 67.5 vs 46.2 mo (HR, 1.53; 95% CI, 1.23-1.91; P  <  .001) 5 y: 79% vs 81% (HR, 1.10; 95% CI, 0.73-1.65; P  >  .99) 
FORTE8,9 158 vs 157 vs 159 50.9 57 KRd-ASCT-KRd
KRd12
KCd-ASCT-KCd 
3 y: 68.8% vs 68.5% vs 67.2% (P  =  .86)
4 y: 69% (KRd-ASCT) vs 56% (KRd12) vs 51% (KCd-ASCT)
HR for KRd-ASCT vs KCd-ASCT, 0.54; 95% CI, 0.38-0.78; P  =  .0008
HR for KRd-ASCT vs KRd12, 0.61; 95% CI, 0.43-0.88; P  =  .0084 
4 y: 86% (KRd-ASCT) vs 85% (KRd12) vs 76% (KCd-ASCT) 
GRIFFIN12  104 vs 103 22.1
 
60 (27%> 65 y) D-RVd-ASCT-DR
RVd-ASCT-R 
3 y: 88.9% vs 81.2% (HR, 0.46; 95% CI, 0.21-1.01) 3 y: 92.6% vs 92.2% (HR, 0.90; 95% CI, 0.32-2.57) 
CASSIOPEIA11  543 vs 542 18.8 58.5 R1: D-VTd-ASCT-D-VTd
VTd-ASCT-VTd
R2: D maint vs obs 
Median: NR in either arm (HR, 0.47; 95% CI, 0.33-0.67; P  <  .0001) Median: NR in either arm (HR, 0.43; 95% CI, 0.23-0.80) 
GMMG-HD652 
 
139 vs 141 vs 137 vs 142
 
49.8 59 RVd-ASCT-R
RVd-ASCT-EloR
Elo-RVd-ASCT-R
Elo-RVd-ASCT-EloR 
3 y: 68.8% vs 68.5% vs 66.2% vs 67.2% (P  =  .86) 3 y: 89.4% vs 89.1% vs 92.5% vs 89.7% (P  =  .43) 

ASCT, autologous stem cell transplant; D, daratumumab; DR, daratumumab, lenalidomide; Elo, elotuzumab; maint, maintenance; NR, not reported; obs, observation; R, lenalidomide; Td, thalidomide, dexamethasone; V, bortezomib; VCd, bortezomib, cyclophosphamide, dexamethasone; VMP, bortezomib, melphalan, prednisone; VTd, bortezomib, thalidomide, dexamethasone; VRd, bortezomib, lenalidomide, dexamethasone.

High-risk patients

The definition of high-risk MM (HRMM) continues to evolve, and a number of clinical, laboratory, and genetic features can be used to identify patients with high-risk biology.17  Historical therapeutic approaches have recommended the use of a PI-based treatment, but it is expected that multidrug combinations that offer multiple mechanisms of targeting the active malignant clone will lead to the greatest depth of response. The FORTE study did include approximately one-third of patients who had at least 1 high-risk cytogenetic feature. Subgroup analyses showed that patients with high-risk disease as defined by Revised International Staging System III had improved outcomes with KRd alone or KRd with transplantation compared with KCd.18  This prompted several groups to start using KRd and transplantation in high-risk patients as a follow-up of the KRd-ASCT and KRd-12 groups showed similar results for high-risk (4-year PFS 62% and 45%) and standard-risk (4-year PFS 80% and 67%) patients. This benefit was also seen in patients with double-hit high-risk disease defined by the presence of ≥2 high-risk cytogenetic abnormalities (4-year PFS 55% and 31%).19  During the second randomization of this trial, more MRD+ patients turned negative in the KR arm vs the single-agent lenalidomide arm (46% vs 32%, P   =  .04) after a median follow-up of 31 months. Patients with high-risk disease have inferior outcomes overall and are a clear unmet need. FORTE offers a glimpse into the role of prolonged inductions with or without transplant, each achieving deep responses, although still demonstrating the benefit of ASCT.

The phase 3 ENDURANCE trial is the only randomized trial to prospectively compare KRd to VRd in NDMM.20  The trial was designed for standard-risk NDMM patients not intended for immediate transplant and specifically excluded high-risk patients. At a median follow-up of 9 months, the median PFS was 34.6 months in the KRd group and 34.4 months in the VRd group (P  =  .74; HR, 1.04). Median OS was not reached in either group. Of note, a composite of grade 3 to 5 treatment-related cardiac, pulmonary, and renal toxicity was higher in the KRd vs VRd group (16% vs 5%, P  <  .0001) largely caused by a higher frequency of dyspnea and heart failure in the KRd group. A subsequent retrospective study of KRd vs RVd followed by ASCT in high-risk NDMM showed similar overall response rate pre-ASCT (98% vs 93%, P   =  .659), 100 days post-ASCT (100% vs 94.8%, P  =  .51), and final best response (100% vs 94.8%, P  =  .79).21  MRD assessment was performed pre- and post-ASCT, and there was no significant difference between both groups (pre: 18% vs 12%; post: 41% vs 43%) at either time point (P  =  .45 and 1.00, respectively).

As discussed above, the GRIFFIN study showed a significant benefit when daratumumab was included as part of initial induction therapy, but the study included few high-risk patients. Ongoing interest remains as to whether daratumumab-based induction is beneficial in the high-risk population as well. A meta-analysis was performed to evaluate the role of daratumumab for the treatment of MM in patients with high-risk cytogenetic factors. While this analysis did include trials of patients with NDMM or HRMM, among patients with newly diagnosed HRMM, the addition of daratumumab to backbone regimens was associated with improved PFS (pooled HR, <0.67; 95% confidence interval [CI], 0.47-0.95; P  =  .02) with little evidence of heterogeneity (Cochran Q, P  =  .77; I2  = 0%).22  At this time, it is not clear that one combination is better than another specifically for high-risk NDMM, and we anxiously await ongoing trials designed to answer this question.

Role of transplant

The role of ASCT for NDMM continues to evolve. Transplant remains an option on the menu for fit patients to achieve a deep response. As mentioned above, multiple studies show the benefit of transplant to continually deepen the response and achieve higher rates of MRD negativity.6,7,23,24  The DETERMINATION and IFM-09 trials offer data from transplant trials in the era of novel agents.6,7  Although these trials by design are not intended to be a “transplant or no transplant” trial, as opposed to a “transplant now or later” trial, the primary end point of PFS supports early transplant in the fit patient. The median PFS was 46 months in the RVd-alone group and 67.5 months in the transplant group. An important note was that patients with high-risk features had a median duration of PFS of 55.5 months in the transplant group vs 17 months in the RVd-alone group. The OS, however, is similar among the 2 groups after a median follow-up of 76 months. The RVd + ASCT arm resulted in a higher percentage of patients in whom MRD was not detected (54% vs 40%), suggesting a strategy to drive deep and durable responses. Patients who achieved MRD negativity either before or after maintenance therapy had an improved 5-year PFS compared with those who remained MRD positive, regardless of whether they received transplantation. The IFM-09 had a nearly identical approach to clinical trial design but discontinued lenalidomide maintenance after 12 months. The median PFS in this trial, however, was 36 months in the RVd-alone group vs 50 months in the transplant arm, perhaps at least in part due to the limited exposure to lenalidomide maintenance. Importantly, PFS was particularly improved in patients who achieved MRD negativity, no matter which assigned treatment arm. This suggests that if deep response is a goal, the treatment selection from the menu should give patients the best chance of obtaining that goal, with or without transplant. These modern transplant trials demonstrate the feasibility of both pursuing or deferring transplant, the successes of continuous therapy to achieve deep responses, and the importance of the elimination of MRD.

Maintenance therapy

Post-ASCT maintenance strategies are a key component to delaying disease recurrence in NDMM and aim to prolong OS. Four randomized phase 3 studies have compared lenalidomide maintenance with observation or placebo post-ASCT.25-28  While there were inherent differences among the patient populations and study designs, each trial demonstrated a clear improvement in median time to progression/PFS in the lenalidomide arms. A meta-analysis demonstrated a median PFS of 52.8 months for the lenalidomide group compared to 23.5 months for the placebo/observation group (HR, 0.48; 95% CI, 0.41-0.55). Although none of the evaluated trials were powered to evaluate OS as a primary endpoint, the meta-analysis demonstrated a significant OS benefit (median not reached vs 86.0 months; HR, 0.75; 95% CI, 0.63-0.90; P  =  .001) for lenalidomide vs placebo/observation. A second meta-analysis, including data from the maintenance portion of the Myeloma XI trial, confirmed this PFS and OS benefit and specifically regardless of age, sex, International Staging System stage, response after ASCT, or whether patients received a lenalidomide-containing induction regimen.29  The optimal duration of maintenance therapy remains to be determined, but the recent IFM-09 and DETERMINATION studies showed the inherent difference in outcomes that resulted when maintenance was discontinued prior to disease progression. Based on the existing body of evidence available, lenalidomide maintenance should ideally continue until disease progression. Multiple other drugs and drug combinations have been or are currently being evaluated in the maintenance setting with the goal to tailor maintenance therapy based on an individual's disease characteristics, cytogenetic risk group, and tolerance, thereby maximizing disease response and survival outcomes while minimizing treatment burden (Table 4).30 

Table 4.

Summary of select data from randomized trials for post-ASCT maintenance therapy

ReferencePatients: total/arms, No.Median follow-up, moRegimenDosing scheduleKey efficacy outcomesKey safety data
Myeloma IX42  245 vs 247 38 T vs obs T 50-100  mg/d continuous Median PFS: 30 vs 23 mo (HR, 1.42)
3-y OS: 75% vs 80% 
Discontinuation due to AEs: 52.2%; SAE: 8.5% 
 1098 vs 1333 NR Meta-analysis, T vs no T maint, including non-ASCT Various doses/durations OS: HR 0.75
7-y OS: 12.3% difference in rate, in favor of T maint 
NR 
HOVON-5043,44 268 vs 268 Initial analysis: 52
Follow-up: 129 
TAd-ASCT-T vs
VAd-ASCT-IFN 
T 50  mg/d until PD Median EFS: 34 vs 22 mo (HR, 0.60); HR 0.62 at follow-up
Median PFS: 34 vs 25 mo (HR, 0.67)
OS: HR 0.96
OS from relapse 20 vs 31 mo (HR, 1.50) 
T maint: discontinuation due to AEs: 33%; 42% at follow-up (vs 27% IFN)
Grade 3-4 PN: 10% 
NCIC-CTG
Myeloma 1045  
166 vs 166 4.1 y TP vs obs T 200  mg/d, P 50  mg q2d; up to 4 y 4-y PFS: 32% vs 14% (HR, 0.55)
4-y OS: 68% vs 60% (HR, 0.77)
Median OS postrelapse: 27.7 vs 34.1 mo 
Grade 3/4 thromboembolism: 7.3% vs 0
Grade 3/4 PN: 9.6% vs 1.2% 
IMWG meta-analysis46  1276 vs 1510 NR T vs no T maint including non-ASCT T various doses/durations PFS: HR 0.65
OS: HR 0.84 
NR 
IFM 2005-0225  307 vs 307 45 R vs placebo 10  mg continuous, increase up to 15  mg; until progression Median PFS: 41 vs 23 mo (HR, 0.50; P  <  .001)
4-y PFS: 43 vs 22% (P  <  .001)
4-y OS: 73% vs 75% (P  =  .7) 
Discontinued for AEs: 27% vs 15%; 2.4 times risk of SPMs with R 
CALGB 10010426,27 231 vs 229 Initial report: 34
Follow-up: 91 
R vs placebo post-ASCT 10  mg continuous, increase up to 15  mg; until progression Initial:
Median PFS/TTP 46 vs 27 mo (HR, 0.48)
3-y OS: 88% vs 80% (HR, 0.62)
Follow-up:
Median PFS/TTP: 57 vs 29 mo (HR, 0.57; P  <  .0001)
Median OS: 114 vs 84 mo (P  =  .0004) 
Discontinuation due to AEs: 10%
Grade 3/4 neutropenia: 32%/13% vs 12%/3%
Heme/solid SPMs: 8%/6% vs 1%/4% 
RV-MM-20928  162 vs 125 (67 vs 68 post-ASCT) 51.2 R vs obs post-MPR (n = 132) or ASCT (n = 141) consolidation 10  mg (21 out of 28 days); until progression ASCT-R vs ASCT:
Median PFS: 54.7 vs 37.4 mo
5-y OS: 78.4% vs 66.6%
R maint CR rate improvement: 15.7% to 35.7%
R vs no maint, post-MPR/ASCT
Median PFS: 41.9 vs 21.6 mo (HR, 0.47)
OS: HR 0.64 
R vs no maint, post-MPR/ASCT
Grade 3/4 AEs:
Neutropenia 23.3% vs 0%
Infections 6.0% vs 1.7%
Discontinuation due to AEs: 5.2% vs 0%
 
Phase 3 meta-analysis26  605 vs 603 79.5 R vs placebo/obs post-ASCT R doses varied Median PFS: 52.8 vs 23.5 mo (HR, 0.48)
Median PFS2: 73.3 vs 56.7 mo (HR, 0.72)
7-year OS: 62% vs 50% (HR, 0.75) 
Discontinuation due to AEs: 29.1% vs 12.2%
Heme/solid SPMs prior to PD: 5.3%/5.8% vs 0.8%/2.0%
 
Myeloma XI29  730 vs 518 31 R vs obs post-ASCT R 10  mg (21 out of 28 days); until progression Median PFS: 57 vs 30 mo (HR, 0.48; P  <  .0001)
3-y OS: 88% vs 80% (HR, 0.69; P  =  .014) 
Grade 3/4 neutropenia: 28%/5%
Discontinuations due to AEs: 28%
SPMs: 5.3% vs 3.1% 
NCT0109183131 
 
60 vs 57 41 vs 42.3 RP vs R alone post-ASCT RP (R 10  mg, days 1-21, 28-d cycles; P 50  mg, q2d); until progression Median PFS: 37.6 vs 31.5 mo
4-year OS: 77% vs 75%
 
Grade 3/4 AEs:
Neutropenia: 8% vs 13%
Infections: 8% vs 5%
Discontinuations due to AEs: 5% vs 8% 
GMMG-MM547  PAd-R → 2 y vs VCd-R → 2 y vs PAd-R → CR vs VCd-R → CR: 125 vs 126 vs 126 vs 125 60.1 R → 2 y vs R CR post-PAd/VCd + ASCT
 
R 10-15  mg/d continuous both arms Median PFS: 43.2 vs 40.9 vs 35.9 vs 35.7 mo
36-mo OS: 83% vs 85% vs 75% vs 77% 
AEs during maintenance (R → 2 y vs R → CR): 77.6% vs 58.2%
Grade ≥2 infections (R → 2 y vs R → CR): 52.7% vs 32.3%
 
HOVON-65/GMMG-HD48,49 413 vs 414
(270 vs 230 maint) 
Initial analysis: 41
Updated analysis: 96 
PAd-ASCT-V vs VAd-ASCT-T V 1.3  mg/m2 IV q2w, up to 2 y
T 50  mg/d, up to 2 y 
Initial:
Median PFS: 35 vs 28 mo (HR, 0.75)
Median PFS from last ASCT: 31 vs 26 mo
5-y OS: 61% vs 55% (HR, 0.81)
Updated:
Median PFS: 34 vs 28 mo (HR, 0.76)
Median OS: 91 vs 82 mo (HR, 0.89)
Median OS from relapse: 43 vs 40 mo (HR, 1.02)
 
SPMs: 7% in both arms
 
GEM05MENOS6550 
 
91 vs 88 vs 92 58.6 VT vs T vs IFN VT (V 1.3  mg/m2 IV days 1, 4, 8, 11, q3m; T 100  mg/d) vs T (T 100  mg/d) vs IFN (3 MU × 3 per wk) post-ASCT, up to 3 y Median PFS: 50.6 vs 40.3 vs 32.5 mo
5-year OS: 78% vs 72% vs 70%
 
Grade 2-3 PN: 48.8% vs 34.4% vs 1%
Discontinuation due to toxicity: 21.9% vs 39.7% vs 20%
 
Tourmaline-MM351  395 vs 261 31 Ixazomib vs placebo post-ASCT Ixazomib 3-4  mg, days 1, 8, 15, 28-d cycles; up to 2 y Median PFS: 26.5 vs 21.3 mo (HR, 0.72)
 
Grade ≥3 AEs: 42% vs 26%
Grade ≥3 infections/infestations: 15% vs 8%
Grade ≥3 GI disorders: 6% vs 1%
Discontinuation due to AEs: 7% vs 5% 
ReferencePatients: total/arms, No.Median follow-up, moRegimenDosing scheduleKey efficacy outcomesKey safety data
Myeloma IX42  245 vs 247 38 T vs obs T 50-100  mg/d continuous Median PFS: 30 vs 23 mo (HR, 1.42)
3-y OS: 75% vs 80% 
Discontinuation due to AEs: 52.2%; SAE: 8.5% 
 1098 vs 1333 NR Meta-analysis, T vs no T maint, including non-ASCT Various doses/durations OS: HR 0.75
7-y OS: 12.3% difference in rate, in favor of T maint 
NR 
HOVON-5043,44 268 vs 268 Initial analysis: 52
Follow-up: 129 
TAd-ASCT-T vs
VAd-ASCT-IFN 
T 50  mg/d until PD Median EFS: 34 vs 22 mo (HR, 0.60); HR 0.62 at follow-up
Median PFS: 34 vs 25 mo (HR, 0.67)
OS: HR 0.96
OS from relapse 20 vs 31 mo (HR, 1.50) 
T maint: discontinuation due to AEs: 33%; 42% at follow-up (vs 27% IFN)
Grade 3-4 PN: 10% 
NCIC-CTG
Myeloma 1045  
166 vs 166 4.1 y TP vs obs T 200  mg/d, P 50  mg q2d; up to 4 y 4-y PFS: 32% vs 14% (HR, 0.55)
4-y OS: 68% vs 60% (HR, 0.77)
Median OS postrelapse: 27.7 vs 34.1 mo 
Grade 3/4 thromboembolism: 7.3% vs 0
Grade 3/4 PN: 9.6% vs 1.2% 
IMWG meta-analysis46  1276 vs 1510 NR T vs no T maint including non-ASCT T various doses/durations PFS: HR 0.65
OS: HR 0.84 
NR 
IFM 2005-0225  307 vs 307 45 R vs placebo 10  mg continuous, increase up to 15  mg; until progression Median PFS: 41 vs 23 mo (HR, 0.50; P  <  .001)
4-y PFS: 43 vs 22% (P  <  .001)
4-y OS: 73% vs 75% (P  =  .7) 
Discontinued for AEs: 27% vs 15%; 2.4 times risk of SPMs with R 
CALGB 10010426,27 231 vs 229 Initial report: 34
Follow-up: 91 
R vs placebo post-ASCT 10  mg continuous, increase up to 15  mg; until progression Initial:
Median PFS/TTP 46 vs 27 mo (HR, 0.48)
3-y OS: 88% vs 80% (HR, 0.62)
Follow-up:
Median PFS/TTP: 57 vs 29 mo (HR, 0.57; P  <  .0001)
Median OS: 114 vs 84 mo (P  =  .0004) 
Discontinuation due to AEs: 10%
Grade 3/4 neutropenia: 32%/13% vs 12%/3%
Heme/solid SPMs: 8%/6% vs 1%/4% 
RV-MM-20928  162 vs 125 (67 vs 68 post-ASCT) 51.2 R vs obs post-MPR (n = 132) or ASCT (n = 141) consolidation 10  mg (21 out of 28 days); until progression ASCT-R vs ASCT:
Median PFS: 54.7 vs 37.4 mo
5-y OS: 78.4% vs 66.6%
R maint CR rate improvement: 15.7% to 35.7%
R vs no maint, post-MPR/ASCT
Median PFS: 41.9 vs 21.6 mo (HR, 0.47)
OS: HR 0.64 
R vs no maint, post-MPR/ASCT
Grade 3/4 AEs:
Neutropenia 23.3% vs 0%
Infections 6.0% vs 1.7%
Discontinuation due to AEs: 5.2% vs 0%
 
Phase 3 meta-analysis26  605 vs 603 79.5 R vs placebo/obs post-ASCT R doses varied Median PFS: 52.8 vs 23.5 mo (HR, 0.48)
Median PFS2: 73.3 vs 56.7 mo (HR, 0.72)
7-year OS: 62% vs 50% (HR, 0.75) 
Discontinuation due to AEs: 29.1% vs 12.2%
Heme/solid SPMs prior to PD: 5.3%/5.8% vs 0.8%/2.0%
 
Myeloma XI29  730 vs 518 31 R vs obs post-ASCT R 10  mg (21 out of 28 days); until progression Median PFS: 57 vs 30 mo (HR, 0.48; P  <  .0001)
3-y OS: 88% vs 80% (HR, 0.69; P  =  .014) 
Grade 3/4 neutropenia: 28%/5%
Discontinuations due to AEs: 28%
SPMs: 5.3% vs 3.1% 
NCT0109183131 
 
60 vs 57 41 vs 42.3 RP vs R alone post-ASCT RP (R 10  mg, days 1-21, 28-d cycles; P 50  mg, q2d); until progression Median PFS: 37.6 vs 31.5 mo
4-year OS: 77% vs 75%
 
Grade 3/4 AEs:
Neutropenia: 8% vs 13%
Infections: 8% vs 5%
Discontinuations due to AEs: 5% vs 8% 
GMMG-MM547  PAd-R → 2 y vs VCd-R → 2 y vs PAd-R → CR vs VCd-R → CR: 125 vs 126 vs 126 vs 125 60.1 R → 2 y vs R CR post-PAd/VCd + ASCT
 
R 10-15  mg/d continuous both arms Median PFS: 43.2 vs 40.9 vs 35.9 vs 35.7 mo
36-mo OS: 83% vs 85% vs 75% vs 77% 
AEs during maintenance (R → 2 y vs R → CR): 77.6% vs 58.2%
Grade ≥2 infections (R → 2 y vs R → CR): 52.7% vs 32.3%
 
HOVON-65/GMMG-HD48,49 413 vs 414
(270 vs 230 maint) 
Initial analysis: 41
Updated analysis: 96 
PAd-ASCT-V vs VAd-ASCT-T V 1.3  mg/m2 IV q2w, up to 2 y
T 50  mg/d, up to 2 y 
Initial:
Median PFS: 35 vs 28 mo (HR, 0.75)
Median PFS from last ASCT: 31 vs 26 mo
5-y OS: 61% vs 55% (HR, 0.81)
Updated:
Median PFS: 34 vs 28 mo (HR, 0.76)
Median OS: 91 vs 82 mo (HR, 0.89)
Median OS from relapse: 43 vs 40 mo (HR, 1.02)
 
SPMs: 7% in both arms
 
GEM05MENOS6550 
 
91 vs 88 vs 92 58.6 VT vs T vs IFN VT (V 1.3  mg/m2 IV days 1, 4, 8, 11, q3m; T 100  mg/d) vs T (T 100  mg/d) vs IFN (3 MU × 3 per wk) post-ASCT, up to 3 y Median PFS: 50.6 vs 40.3 vs 32.5 mo
5-year OS: 78% vs 72% vs 70%
 
Grade 2-3 PN: 48.8% vs 34.4% vs 1%
Discontinuation due to toxicity: 21.9% vs 39.7% vs 20%
 
Tourmaline-MM351  395 vs 261 31 Ixazomib vs placebo post-ASCT Ixazomib 3-4  mg, days 1, 8, 15, 28-d cycles; up to 2 y Median PFS: 26.5 vs 21.3 mo (HR, 0.72)
 
Grade ≥3 AEs: 42% vs 26%
Grade ≥3 infections/infestations: 15% vs 8%
Grade ≥3 GI disorders: 6% vs 1%
Discontinuation due to AEs: 7% vs 5% 

AE, adverse event; CR, complete response; EFS, event-free survival; GI, gastrointestinal; heme, hematologic; IMWG, International Myeloma Working Group; IFN, interferon; IV, intravenous; MPR, melphalan, prednisone, lenalidomide; MU, million units; P, prednisone; PAd, bortezomib, doxorubicin, dexamethasone; PD, progressive disease; PFS2, progression-free survival from start of treatment to progression on next line of treatment; PN, peripheral neuropathy; q2d every other day; q2w, every 2 weeks; q3m, every 3 months; R, lenalidomide; RP, lenalidomide, prednisone; SAE, serious adverse event; SPM, second primary malignancy; T, thalidomide; TAd, thalidomide, doxorubicin, dexamethasone; TP, thalidomide, prednisone; TTP, time to progression; V, bortezomib; VAd, vincristine, doxorubicin, dexamethasone; VT, bortezomib, thalidomide.

It is important to remember that fitness for transplant may be fluid around the time of diagnosis and after the initiation of induction, which can often significantly improve performance status due to rapid improvement in disease burden. Transplant consultation is an important component to determining eligibility, and all potentially eligible patients should be referred to a transplant center for evaluation early in the induction course. However, once transplant ineligibility has been determined, a tailored approach to ensure that the treatment benefit is balanced carefully with safety, while maintaining the goal of deep and durable responses, must be employed.

Although age has previously been used as an inclusion criterion for ASCT,28,31-33  this has become less relevant due to an aging, yet fit, population that may be otherwise excluded. Comorbidity scores have been validated and can help evaluate transplant eligibility and frailty, including a myeloma-specific comorbidity index score.34-36  Preventing and managing treatment side effects is of significant importance in the frail population that can allow for continued therapy that is tolerable and can provide deep responses.

Two- vs 3-drug regimens

We now have numerous randomized trials that confirm the benefit of 3-drug regimens in a transplant-ineligible or transplant-deferred population (Table 5). We have previously reviewed the SWOG 0777 phase 3 trial that confirmed the PFS and OS benefit of VRd when compared to Rd and solidified the importance of a triplet induction regimen.5  This was further substantiated in the MAIA trial, which added daratumumab to Rd (DRd) in this randomized controlled phase 3 trial.37  The primary end point was PFS with a secondary end point of OS. An interim analysis after a median follow-up of 56.2 months showed the median PFS was not reached (95% CI, 54.8 months to not reached) in the daratumumab group vs 34.4 months (95% CI, 29.6-39.2 months) in the control group (HR, 0.53; 95% CI, 0.43-0.66; P  <  .0001); estimated PFS at 60 months was 52.5% (95% CI, 46.7%-58.0%) vs 28.7% (95% CI, 23.1%-34.6%). Estimated 60-month overall survival was 66.3% (95% CI, 60.8%-71.3%) in the daratumumab group vs 53.1% (95% CI, 47.2%-58.6%) in the control group.38  MRD negativity rates were also significantly higher in the daratumumab group than in the control group after 48 months (31% vs 10%). The most common (>15%) grade 3 or higher treatment-emergent adverse events were neutropenia, pneumonia, anemia, and lymphopenia, all higher in the triplet arm, with no new safety concerns. We now have 2 large randomized, controlled clinical trials, ALCYONE and MAIA, that incorporate daratumumab for transplant- ineligible patients and demonstrated a significant improvement in OS. With longer follow up, DRd is expected to result in an unprecedented median PFS in this patient population and supports its frontline use as a new standard of care for induction in this transplant-ineligible population (Table 5).

Table 5.

Summary of select data from randomized trials for induction therapy for transplant-ineligible NDMM

ReferencePatients: total/arms, No.Median follow-up, moMedian age (range), yRegimenPFSOSPrimary end point/comments
PETHEMA53  260 patients; 130/130 32 73 (68-77)
73 (69-76) 
VMP vs VTP
Maint VT or VP 
31 mo, all patients 3-y OS: 70% (64%-76%), all patients ORR
VTP 81%, VMP 80%, VTP more serious AEs (40 [31% vs 20 [15%], P  =  .01) and drug discontinuations 
GIMEMA 030535,54 511 patients;
254/257 
23.2 71 VMPT-VT
vs
VMP 
3-y PFS:
VMPT-VT 56%; VMP 41%;
HR 0.67
(95% CI, 0.50-0.90; P  =  .008) 
5-y OS:
VMPT-VT 61%
VMP 51%
HR 0.70
(P  =  .01) 
PFS
VMPT-VT arm: more frequent grade 3/4 AEs: neutropenia (38%), thrombocytopenia (22%), peripheral neuropathy (11%), cardiac events (11%) 
MRC-IX (59)55 
 
859 patients; 423/426 44 73 MP vs CTd MP 12.4 mo; CTd 13 mo
HR 0.82 (95% CI, 0.70-0.96; P  =  .01) 
MP 30.6 mo; CTd 33.2 mo; HR 0.89 (95% CI, 0.74-1.08; P  =  .24) ORR/PFS/OS
ORR: MP 32.6%; CTd 63.8% (P  < . 0001)
CTDa arm: higher rates of thromboembolic events, constipation, infection, and neuropathy 
FIRST56  1623 patients; 535/541/547 67 73 (40-92) Rd vs Rd18 vs MPT Rd 25.5 mo; Rd18 20.7 mo; MPT 21.2 mo; HR 0.72, Rd vs MPT
HR 0.70, Rd vs Rd18; P  <  .001 for both. Rd was superior to MPT for all secondary efficacy end points, including OS. 
4-y OS: Rd 59%; Rd18 56%; MPT 51% PFS
Grade 3/4 AEs more frequent with Rd than MPT (70% vs 78%) 
UPFRONT57 
 
502 patients;
168/167/167 
42.7 74.5 (67-69) Vd + V
73 (66-77) VTd + V
72 (68-77) VMP + V
 
Vd 14.7 mo; VTd 15.4 mo; VMP 17.3 mo (P  =  NS) Vd 49.8 mo
VTd 51.5 mo
VMP 53.1 mo (P  =  NS) 
PFS
Peripheral neuropathy near 50% in all arms. Early drug discontinuation (29%-38%).
QoL scores decreased during induction and improved or stabilized thereafter. 
HOVON-8758  668 patients
318/319 
36 72 (60-91) MPT-T vs MPR-R MPT-T 20 mo; MPR-R 23 mo; HR 0.87 (95% CI, 0.72-1.04; P  =  .12) 4-y OS:
MPT-T 52% MPR-R 56% 
PFS
Grade 3/4 hematologic toxicity with MPR-R vs clinically significant neuropathy with MPT-T 
SWOG S07775,59
 
525 patients; 264/261 84 43% ≥ 65 VRd vs Rd VRd 41 mo
Rd 29 mo
HR 0.742 (95% CI 0.594-0.9028; 1-sided P  =  .003) 
VRd NR; Rd 69 mo; HR 0.709 (95% CI, 0.543-0.926; 2-sided P  =  .0114) PFS
VRd (23%) and Rd (10%) discontinued induction treatment due to AE 
MAIA38 
 
737 patients: 368/369 56.2 73 (50-90) D-Rd continuous
74 (45-89) Rd continuous
43%-44% ≥ 75 
D-Rd NR
Rd 34.4 mo
HR 0.53 (95% CI, 0.43-0.66; P  <  .0001) 
NR in either arm. HR 0.68 (95% CI, 0.53-0.86; P  =  .0013) PFS
Median duration on continuous treatment 47.5 mo (D-Rd) and 22.6 mo (Rd) 
HOVON-126/NMSG60 
 
143 patients
 
23.4
 
73 (64-90) ITd → maintenance ixazomib vs placebo
 
PFS-R:
ITd-I 9.5 mo; ITd- P 8.4 mo 
OS-R at 18 mo, all patients 96% (88%-99%) PFS
Early mortality 8% age >75, only 55% randomly assigned to maintenance therapy 
ALCYONE10 
 
706 patients; 350/356 40.1 71 (40-93)
71 (50-91) 
D-VMP-D
VMP 
D-VMP-D 36.4 mo; VMP 19.3 mo; HR 0.42 (95% CI, 0.34-0.51; P  <  .0001) 3-y OS: D-VMP-D 78%; VMP 67.9%; HR 0.60 (95% CI, 0.46-0.80; P  =  .0003) PFS
Common 
ECOG E1A1120 
 
1087 patients;
542/545 
65 (57-71)
64 (59-71) 
VRd-R
KRd-R 
VRd 34.4 mo
KRd 34.6 mo 
Median OS NR either arm PFS, OS; excluded high-risk disease; 17.3% discontinued VRd early due to AEs 
TOURMALINE-MM261  705 patients;
351;354 
IRd 53.3
Rd 55.8 
73 (48-90)
74 (48-88) 
IRd-IR
Rd-R 
IRd 35.3 mo
Rd 21.8 mo
(HR, 0.830; 95% CI, 0.676-1.018; P  =  .073) 
Median OS NR either arm at 58 mo. HR 0.998 (95% CI, 0.790-1.261; P  =  .988) PFS
PFS not improved in age ≥75 in IRd 
ReferencePatients: total/arms, No.Median follow-up, moMedian age (range), yRegimenPFSOSPrimary end point/comments
PETHEMA53  260 patients; 130/130 32 73 (68-77)
73 (69-76) 
VMP vs VTP
Maint VT or VP 
31 mo, all patients 3-y OS: 70% (64%-76%), all patients ORR
VTP 81%, VMP 80%, VTP more serious AEs (40 [31% vs 20 [15%], P  =  .01) and drug discontinuations 
GIMEMA 030535,54 511 patients;
254/257 
23.2 71 VMPT-VT
vs
VMP 
3-y PFS:
VMPT-VT 56%; VMP 41%;
HR 0.67
(95% CI, 0.50-0.90; P  =  .008) 
5-y OS:
VMPT-VT 61%
VMP 51%
HR 0.70
(P  =  .01) 
PFS
VMPT-VT arm: more frequent grade 3/4 AEs: neutropenia (38%), thrombocytopenia (22%), peripheral neuropathy (11%), cardiac events (11%) 
MRC-IX (59)55 
 
859 patients; 423/426 44 73 MP vs CTd MP 12.4 mo; CTd 13 mo
HR 0.82 (95% CI, 0.70-0.96; P  =  .01) 
MP 30.6 mo; CTd 33.2 mo; HR 0.89 (95% CI, 0.74-1.08; P  =  .24) ORR/PFS/OS
ORR: MP 32.6%; CTd 63.8% (P  < . 0001)
CTDa arm: higher rates of thromboembolic events, constipation, infection, and neuropathy 
FIRST56  1623 patients; 535/541/547 67 73 (40-92) Rd vs Rd18 vs MPT Rd 25.5 mo; Rd18 20.7 mo; MPT 21.2 mo; HR 0.72, Rd vs MPT
HR 0.70, Rd vs Rd18; P  <  .001 for both. Rd was superior to MPT for all secondary efficacy end points, including OS. 
4-y OS: Rd 59%; Rd18 56%; MPT 51% PFS
Grade 3/4 AEs more frequent with Rd than MPT (70% vs 78%) 
UPFRONT57 
 
502 patients;
168/167/167 
42.7 74.5 (67-69) Vd + V
73 (66-77) VTd + V
72 (68-77) VMP + V
 
Vd 14.7 mo; VTd 15.4 mo; VMP 17.3 mo (P  =  NS) Vd 49.8 mo
VTd 51.5 mo
VMP 53.1 mo (P  =  NS) 
PFS
Peripheral neuropathy near 50% in all arms. Early drug discontinuation (29%-38%).
QoL scores decreased during induction and improved or stabilized thereafter. 
HOVON-8758  668 patients
318/319 
36 72 (60-91) MPT-T vs MPR-R MPT-T 20 mo; MPR-R 23 mo; HR 0.87 (95% CI, 0.72-1.04; P  =  .12) 4-y OS:
MPT-T 52% MPR-R 56% 
PFS
Grade 3/4 hematologic toxicity with MPR-R vs clinically significant neuropathy with MPT-T 
SWOG S07775,59
 
525 patients; 264/261 84 43% ≥ 65 VRd vs Rd VRd 41 mo
Rd 29 mo
HR 0.742 (95% CI 0.594-0.9028; 1-sided P  =  .003) 
VRd NR; Rd 69 mo; HR 0.709 (95% CI, 0.543-0.926; 2-sided P  =  .0114) PFS
VRd (23%) and Rd (10%) discontinued induction treatment due to AE 
MAIA38 
 
737 patients: 368/369 56.2 73 (50-90) D-Rd continuous
74 (45-89) Rd continuous
43%-44% ≥ 75 
D-Rd NR
Rd 34.4 mo
HR 0.53 (95% CI, 0.43-0.66; P  <  .0001) 
NR in either arm. HR 0.68 (95% CI, 0.53-0.86; P  =  .0013) PFS
Median duration on continuous treatment 47.5 mo (D-Rd) and 22.6 mo (Rd) 
HOVON-126/NMSG60 
 
143 patients
 
23.4
 
73 (64-90) ITd → maintenance ixazomib vs placebo
 
PFS-R:
ITd-I 9.5 mo; ITd- P 8.4 mo 
OS-R at 18 mo, all patients 96% (88%-99%) PFS
Early mortality 8% age >75, only 55% randomly assigned to maintenance therapy 
ALCYONE10 
 
706 patients; 350/356 40.1 71 (40-93)
71 (50-91) 
D-VMP-D
VMP 
D-VMP-D 36.4 mo; VMP 19.3 mo; HR 0.42 (95% CI, 0.34-0.51; P  <  .0001) 3-y OS: D-VMP-D 78%; VMP 67.9%; HR 0.60 (95% CI, 0.46-0.80; P  =  .0003) PFS
Common 
ECOG E1A1120 
 
1087 patients;
542/545 
65 (57-71)
64 (59-71) 
VRd-R
KRd-R 
VRd 34.4 mo
KRd 34.6 mo 
Median OS NR either arm PFS, OS; excluded high-risk disease; 17.3% discontinued VRd early due to AEs 
TOURMALINE-MM261  705 patients;
351;354 
IRd 53.3
Rd 55.8 
73 (48-90)
74 (48-88) 
IRd-IR
Rd-R 
IRd 35.3 mo
Rd 21.8 mo
(HR, 0.830; 95% CI, 0.676-1.018; P  =  .073) 
Median OS NR either arm at 58 mo. HR 0.998 (95% CI, 0.790-1.261; P  =  .988) PFS
PFS not improved in age ≥75 in IRd 

CTd, cyclophosphamide, thalidomide, dexamethasone; IRd, ixazomib, lenalidomide, dexamethasone; IRd-IR, ixazomib, lenalidomide, dexamethasone followed by ixazomib, lenalidomide continuous therapy; ITd, ixazomib, thalidomide, dexamethasone; ITd-I, ixazomib, thalidomide, dexamethasone induction and ixazomib continuous therapy; ITd-P, ixazomib, thalidomide, dexamethasone and placebo continuous therapy; MP, melphalan, prednisone; MPR, melphalan, prednisone, lenalidomide; MPT, melphalan, prednisone, thalidomide; ORR, overall response rate; QoL, quality of life; Rd, lenalidomide, dexamethasone; Rd18, lenalidomide, dexamethasone for 18 months; Vd, bortezomib, dexamethasone; VMPT, bortezomib, melphalan, prednisone, thalidomide; VTd, bortezomib, thalidomide, dexamethasone.

Studies have shown the benefit of continuous therapy and that responses can deepen over time and lead to survival improvements.39,40  Continuous dexamethasone, however, can lead to excess toxicity in this population, and the opportunity to discontinue it should be considered as data suggest that maintenance therapy can be equally as effective without prolonged usage.41  The treatment of HRMM remains an unmet challenge for the unfit population, and the optimal treatment approach continues to evolve.

The patient was diagnosed with Revised International Staging System II disease without high-risk features and received 4 cycles of D-RVd followed by high-dose melphalan and autologous stem cell transplant. She achieved MRD+ sCR after transplant and started lenalidomide maintenance. She is now 2 years post-transplant and remains in sCR.

We now have multiple combinations available for the management of NDMM, with 4-drug combinations particularly showing deeper and more durable remissions than have previously been seen. The early successes of immunotherapeutic options in relapsed/refractory MM have led to ongoing studies looking to incorporate these therapies, including chimeric antigen receptor T-cell therapies and bispecific antibodies, into induction strategies for NDMM as a means to harness the potential of a less impaired immune system. Modern trials that have used overall response rate, PFS, and OS as primary end points are now seeing contemporary trials include MRD negativity as an end point and an opportunity for treatment-free intervals, a significant paradigm shift from the maintenance strategies we have become familiar with. Novel therapies, strategic combinations, and new end points for trials will continue to improve the already unprecedented achievements that have been seen in the management of NDMM to date.

Caitlin L. Costello: research funding: BMS, Janssen, Takeda; consultancy: BMS, Janssen, Takeda, Pfizer.

Caitlin L. Costello: nothing to disclose.

1.
Rajkumar
SV
,
Dimopoulos
MA
,
Palumbo
A
, et al.
International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma
.
Lancet Oncol
.
2014
;
15
(
12
):
e538
-
e548
.
doi:10.1016/S1470-2045(14)70442-5
.
2.
Barlogie
B
,
Jagannath
S
,
Desikan
KR
, et al.
Total therapy with tandem transplants for newly diagnosed multiple myeloma
.
Blood
.
1999
;
93
(
1
):
55
-
65
.
doi:10.1182/blood.V93.1.55
3.
Paiva
B
,
van Dongen
JJ
,
Orfao
A
.
New criteria for response assessment: role of minimal residual disease in multiple myeloma
.
Blood
.
2015
;
125
(
20
):
3059
-
3068
.
doi:10.1182/blood-2014-11-568907
.
4.
Munshi
NC
,
Avet-Loiseau
H
,
Anderson
KC
, et al.
A large meta-analysis establishes the role of MRD negativity in long-term survival outcomes in patients with multiple myeloma
.
Blood Adv
.
2020
;
4
(
23
):
5988
-
5999
.
doi:10.1182/bloodadvances.2020002827
.
5.
Durie
BGM
,
Hoering
A
,
Abidi
MH
, et al.
Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial
.
Lancet
.
2017
;
389
(
10068
):
519
-
527
.
doi:10.1016/S0140-6736(16)31594-X
.
6.
Attal
M
,
Lauwers-Cances
V
,
Hulin
C
, et al
; IFM 2009 Study.
Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma
.
N Engl J Med
.
2017
;
376
(
14
):
1311
-
1320
.
doi:10.1056/NEJMoa1611750
.
7.
Richardson
PG
,
Jacobus
SJ
,
Weller
EA
, et al
;
DETERMINATION Investigators
.
Triplet therapy, transplantation, and maintenance until progression in myeloma
.
N Engl J Med
.
2022
;
387
(
2
):
132
-
147
.
doi:10.1056/NEJMoa2204925
.
8.
Gay
F
,
Musto
P
,
Scalabrini
DR
, et al.
Survival analysis of newly diagnosed transplant-eligible multiple myeloma patients in the randomized Forte trial [abstract]
.
Blood
.
2020
;
136
(
suppl 1
):
35
-
37
.
doi:10.1182/blood-2020-136907
.
9.
Gay
F
,
Cerrato
C
,
Scalabrini
DR
, et al.
Carfilzomib-lenalidomide-dexamethasone (KRd) induction-autologous transplant (ASCT)-KRd consolidation vs KRD 12 cycles vs carfilzomib-cyclophosphamide-dexamethasone (KCd) induction-ASCT-KCd consolidation: analysis of the randomized FORTE Trial in newly diagnosed multiple myeloma (NDMM) [abstract]
.
Blood
.
2018
;
132
(
suppl 1
):
121
.
doi:10.1182/blood-2018-99-112093
10.
Mateos
M-V
,
Dimopoulos
M-A
,
Cavo
M
, et al
;
ALCYONE Trial Investigators
.
Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma
.
N Engl J Med
.
2018
;
378
(
6
):
518
-
528
.
doi:10.1056/NEJMoa1714678
.
11.
Moreau
P
,
Attal
M
,
Hulin
C
, et al.
Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study
.
Lancet
.
2019
;
394
(
10192
):
29
-
38
.
doi:10.1016/S0140-6736(19)31240-1
.
12.
Voorhees
PM
,
Kaufman
JL
,
Laubach
J
, et al.
Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial
.
Blood
.
2020
;
136
(
8
):
936
-
945
.
doi:10.1182/blood.2020005288
.
13.
Rodriguez
C
,
Kaufman
JL
,
Laubach
J
, et al.
Daratumumab + lenalidomide, bortezomib, and dexamethasone in transplant-eligible newly diagnosed multiple myeloma: a post hoc analysis of sustained minimal residual disease negativity from GRIFFIN [abstract]
.
J Clin Oncol
.
2022
;
40
(
suppl 16
):
8011
.
doi:10.1200/JCO.2022.40.16_suppl.8011
.
14.
Costa
LJ
,
Chhabra
S
,
Medvedova
E
, et al.
Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma
.
J Clin Oncol
.
2022
;
40
(
25
):
2901
-
2912
.
doi:10.1200/JCO.21.01935
.
15.
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: the MANHATTAN nonrandomized clinical trial
.
JAMA Oncol
.
2021
;
7
(
6
):
862
-
868
.
doi:10.1001/jamaoncol.2021.0611
.
16.
Touzeau
C
,
Perrot
A
,
Hulin
C
, et al.
Daratumumab carfilzomib lenalidomide and dexamethasone as induction therapy in high-risk transplant eligible newly diagnosed myeloma patients: results of the phase 2 study IFM 2018-04
. [abstract].
J Clin Oncol.
2022
;
40
(
suppl 16
):
8002
.
doi:10.1200/JCO.2022.40.16_suppl.8002
.
17.
Davies
FE
,
Pawlyn
C
,
Usmani
SZ
, et al.
Perspectives on the risk-stratified treatment of multiple myeloma
.
Blood Cancer Discov
.
2022
;
3
(
4
):
273
-
284
.
doi:10.1158/2643-3230.BCD-21-0205
.
18.
Gay
F
,
Cerrato
C
,
Petrucci
MT
, et al.
Efficacy of carfilzomib lenalidomide dexamethasone (Kid) with or without transplantation in newly diagnosed myeloma according to risk status: results from the FORTE trial [abstract]
.
J Clin Oncol
.
2019
;
37
(
15, suppl
):
8002
.
doi:10.1200/JCO.2019.37.15_suppl.8002
.
19.
Gay
F
,
Mina
R
,
Rota-Scalabrini
D
, et al.
Carfilzomib-based induction/consolidation with or without autologous transplant (ASCT) followed by lenalidomide ® or carfilzomib-lenalidomide (KR) maintenance: efficacy in high-risk patients
.
J Clin Oncol
.
2021
;
39
(
15, suppl
):
8002
.
doi:10.1200/jco.2021.39.15_suppl.8002
.
20.
Kumar
SK
,
Jacobus
SJ
,
Cohen
AD
, et al.
Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial
.
Lancet Oncol
.
2020
;
21
(
10
):
1317
-
1330
.
doi:10.1016/S1470-2045(20)30452-6
.
21.
Gaballa
MR
,
Ma
J
,
Rauf
M
, et al.
KRD vs. VRD as induction before autologous hematopoietic progenitor cell transplantation for high-risk multiple myeloma
.
Bone Marrow Transplant
.
2022
;
57
(
7
):
1142
-
1149
.
doi:10.1038/s41409-022-01697-4
.
22.
Giri
S
,
Grimshaw
A
,
Bal
S
, et al.
Evaluation of daratumumab for the treatment of multiple myeloma in patients with high-risk cytogenetic factors: a systematic review and meta-analysis
.
JAMA Oncol
.
2020
;
6
(
11
):
1759
-
1765
.
doi:10.1001/jamaoncol.2020.4338
.
23.
Cavo
M
,
Gay
F
,
Beksac
M
, et al.
Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a multicentre, randomised, open-label, phase 3 study
.
Lancet Haematol
.
2020
;
7
(
6
):
e456
-
e468
.
doi:10.1016/S2352-3026(20)30099-5
.
24.
Stadtmauer
EA
,
Pasquini
MC
,
Blackwell
B
, et al.
Autologous transplantation, consolidation, and maintenance therapy in multiple myeloma: results of the BMT CTN 0702 Trial
.
J Clin Oncol
.
2019
;
37
(
7
):
589
-
597
.
doi:10.1200/JCO.18.00685
.
25.
Attal
M
,
Lauwers-Cances
V
,
Marit
G
, et al
;
IFM Investigators
.
Lenalidomide maintenance after stem-cell transplantation for multiple myeloma
.
N Engl J Med
.
2012
;
366
(
19
):
1782
-
1791
.
doi:10.1056/NEJMoa1114138
.
26.
McCarthy
PL
,
Owzar
K
,
Hofmeister
CC
, et al.
Lenalidomide after stem-cell transplantation for multiple myeloma
.
N Engl J Med
.
2012
;
366
(
19
):
1770
-
1781
.
doi:10.1056/NEJMoa1114083
.
27.
Holstein
SA
,
Jung
S-H
,
Richardson
P-G
, et al.
Updated analysis of CALGB (Alliance) 100104 assessing lenalidomide versus placebo maintenance after single autologous stem-cell transplantation for multiple myeloma: a randomised, double-blind, phase 3 trial
.
Lancet Haematol
.
2017
;
4
(
9
):
e431
-
e442
.
doi:10.1016/S2352-3026(17)30140-0
.
28.
Palumbo
A
,
Cavallo
F
,
Gay
F
, et al.
Autologous transplantation and maintenance therapy in multiple myeloma
.
N Engl J Med
.
2014
;
371
(
10
):
895
-
905
.
doi:10.1056/NEJMoa1402888
.
29.
Jackson
GH
,
Davies
FE
,
Pawlyn
C
, et al
;
UK NCRI Haemato-oncology Clinical Studies Group
.
Lenalidomide maintenance versus observation for patients with newly diagnosed multiple myeloma (Myeloma XI): a multicentre, open-label, randomised, phase 3 trial
.
Lancet Oncol
.
2019
;
20
(
1
):
57
-
73
.
doi:10.1016/S1470-2045(18)30687-9
.
30.
Holstein
SA
,
Suman
VJ
,
Hillengass
J
,
McCarthy
PL
. Future directions in maintenance therapy in multiple myeloma.
J Clin Med
.
2021
;
10
(
11
).
doi:10.3390/jcm10112261
.
31.
Gay
F
,
Oliva
S
,
Petrucci
MT
, et al.
Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial
.
Lancet Oncol
.
2015
;
16
(
16
):
1617
-
1629
.
doi:10.1016/S1470-2045(15)00389-7
.
32.
Child
JA
,
Morgan
GJ
,
Davies
FE
, et al
;
Medical Research Council Adult Leukaemia Working Party
.
High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma
.
N Engl J Med
.
2003
;
348
(
19
):
1875
-
1883
.
doi:10.1056/NEJMoa022340
.
33.
Bethge
WA
,
Hegenbart
U
,
Stuart
MJ
, et al.
Adoptive immunotherapy with donor lymphocyte infusions after allogeneic hematopoietic cell transplantation following nonmyeloablative conditioning
.
Blood
.
2004
;
103
(
3
):
790
-
795
.
doi:10.1182/blood-2003-07-2344
.
34.
Engelhardt
M
,
Domm
A-S
,
Dold
S-M
, et al.
A concise revised Myeloma Comorbidity Index as a valid prognostic instrument in a large cohort of 801 multiple myeloma patients
.
Haematologica
.
2017
;
102
(
5
):
910
-
921
.
doi:10.3324/haematol.2016.162693
.
35.
Charlson
ME
,
Pompei
P
,
Ales
KL
,
MacKenzie
CR
.
A new method of classifying prognostic comorbidity in longitudinal studies: development and validation
.
J Chronic Dis
.
1987
;
40
(
5
):
373
-
383
.
doi:10.1016/0021-9681(87)90171-8
.
36.
Sorror
ML
,
Maris
MB
,
Storb
R
, et al.
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT
.
Blood
.
2005
;
106
(
8
):
2912
-
2919
.
doi:10.1182/blood-2005-05-2004
.
37.
Facon
T
,
Kumar
S
,
Plesner
T
, et al
;
MAIA Trial Investigators
.
Daratumumab plus lenalidomide and dexamethasone for untreated myeloma
.
N Engl J Med
.
2019
;
380
(
22
):
2104
-
2115
.
doi:10.1056/NEJMoa1817249
.
38.
Facon
T
,
Kumar
SK
,
Plesner
T
, et al.
Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): overall survival results from a randomised, open-label, phase 3 trial
.
Lancet Oncol
.
2021
;
22
(
11
):
1582
-
1596
.
doi:10.1016/S1470-2045(21)00466-6
.
39.
Palumbo
A
,
Bringhen
S
,
Caravita
T
,
et al; Italian Multiple Myeloma Network
GIMEMA
.
Oral melphalan and prednisone chemotherapy plus thalidomide compared with melphalan and prednisone alone in elderly patients with multiple myeloma: randomised controlled trial
.
Lancet
.
2006
;
367
(
9513
):
825
-
831
.
doi:10.1016/S0140-6736(06)68338-4
.
40.
Facon
T
,
Dimopoulos
MA
,
Dispenzieri
A
, et al.
Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma
.
Blood
.
2018
;
131
(
3
):
301
-
310
.
doi:10.1182/blood-2017-07-795047
.
41.
Larocca
A
,
Salvini
M
,
De Paoli
L
, et al.
Efficacy and feasibility of dose/schedule-adjusted RD-R vs continuous Rd in elderly and intermediate-fit newly diagnosed multiple myeloma (NDMM) patients: RV-MM-PI-0752 phase III randomized study
.
Blood
.
2018
;
32
(
suppl 1
):
305
.
doi:10.1182/blood-2018-99-111796
.
42.
Morgan
GJ
,
Gregory
WM
,
Davies
FE
, et al
;
National Cancer Research Institute Haematological Oncology Clinical Studies Group
.
The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis
.
Blood
.
2012
;
119
(
1
):
7
-
15
.
doi:10.1182/blood-2011-06-357038
.
43.
Lokhorst
HM
,
van der Holt
B
,
Zweegman
S
, et al
; Dutch-Belgian Hemato-Oncology Group (HOVON).
A randomized phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma
.
Blood
.
2010
;
115
(
6
):
1113
-
1120
.
doi:10.1182/blood-2009-05-222539
.
44.
van de Donk
NW
,
van der Holt
B
,
Minnema
MC
, et al.
Thalidomide before and after autologous stem cell transplantation in recently diagnosed multiple myeloma (HOVON-50): long-term results from the phase 3, randomised controlled trial
.
Lancet Haematol
.
2018
;
5
(
10
):
e479
-
e492
.
doi:10.1016/S2352-3026(18)30149-2
.
45.
Stewart
AK
,
Trudel
S
,
Bahlis
NJ
, et al.
A randomized phase 3 trial of thalidomide and prednisone as maintenance therapy after ASCT in patients with MM with a quality-of-life assessment: the National Cancer Institute of Canada Clinicals Trials Group Myeloma 10 Trial
.
Blood
.
2013
;
121
(
9
):
1517
-
1523
.
doi:10.1182/blood-2012-09-451872
.
46.
Ludwig
H
,
Durie
BG
,
McCarthy
P
, et al
;
International Myeloma Working Group
.
IMWG consensus on maintenance therapy in multiple myeloma
.
Blood
.
2012
;
119
(
13
):
3003
-
3015
.
doi:10.1182/blood-2011-11-374249
.
47.
Goldschmidt
H
,
Mai
EK
,
Durig
J
, et al.
Response-adapted lenalidomide maintenance in newly diagnosed, transplant-eligible multiple myeloma: results from the multicenter phase III GMMG-MM5 trial
.
Leukemia
.
2020
;
34
(
7
):
185301865
.
doi:10.1038/s41375-020-0724-1
.
48.
Goldschmidt
H
,
Lokhorst
HM
,
Mai
EK
, et al.
Bortezomib before and after high-dose therapy in myeloma: long-term results from the phase III HOVON-65/GMMG-HD4 trial
.
Leukemia
.
2018
;
32
(
2
):
383
-
390
.
doi:10.1038/leu.2017.211
.
49.
Sonneveld
P
,
Schmidt-Wolf
IG
,
van der Holt
B
, et al.
Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/GMMG-HD4 trial
.
J Clin Oncol
.
2012
;
30
(
24
):
2946
-
2955
.
doi:10.1200/JCO.2011.39.6820
.
50.
Rosiñol
L
,
Oriol
A
,
Teruel
AI
, et al.
Bortezomib and thalidomide maintenance after stem cell transplantation for multiple myeloma: a PETHEMA/GEM trial
.
Leukemia
.
2017
;
31
(
9
):
1922
-
1927
.
doi:10.1038/leu.2017.35
.
51.
Dimopoulos
MA
,
Gay
F
,
Schjesvold
F
, et al
;
TOURMALINE-MM3 study group
.
Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): a double-blind, randomised, placebo-controlled phase 3 trial
.
Lancet
.
2019
;
393
(
10168
):
253
-
264
.
doi:10.1016/S0140-6736(18)33003-4
.
52.
Goldschmidt
HME
,
Bertsch
U
,
Besemer
B
, et al.
Elotuzumab in combination with lenalidomide, bortezomib, dexamethasone and autologous transplantation for newly-diagnosed multiple myeloma: results from the randomized phase III GMMG-HD6 trial
.
Blood
.
2021
;
130
(
suppl 1
):
486
.
doi:10.1182/blood-2021-147323
.
53.
Mateos
MV
,
Oriol
A
,
Martínez-López
J
, et al.
Bortezomib, melphalan, and prednisoneversus bortezomib, thalidomide, and prednisone as induction therapy followed by maintenance treatment with bortezomib and thalidomide versus bortezomib and prednisone in elderly patients with untreated multiple myeloma: a randomised trial
.
Lancet Oncol
.
2010
;
11
(
10
):
934
-
941
.
doi:10.1016/S1470-2045(10)70187-X
54.
Palumbo
A
,
Bringhen
S
,
Larocca
A
, et al.
Bortezomib-melphalan-prednisone- thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: updated follow-up and improved survival
.
J Clin Oncol
.
2014
;
32
(
7
):
634
-
640
.
doi:10.1200/JCO.2013.52.0023
.
55.
Morgan
GJ
,
Davies
FE
,
Gregory
WM
, et al
;
NCRI Haematological Oncology Study Group
.
Cyclophosphamide, thalidomide, and dexamethasone (CTD) as initial therapy for patients with multiple myeloma unsuitable for autologous transplantation
.
Blood
.
2011
;
118
(
5
):
1231
-
1238
.
doi:10.1182/blood-2011-02-338665
.
56.
Benboubker
L
,
Dimopoulos
MA
,
Dispenzieri
A
, et al
;
FIRST Trial Team
.
Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma
.
N Engl J Med
.
2014
;
371
(
10
):
906
-
917
.
doi:10.1056/NEJMoa1402551
.
57.
Niesvizky
R
,
Flinn
IW
,
Rifkin
R
, et al.
Community-based phase IIIB trial of three UPFRONT bortezomib-based myeloma regimens
.
J Clin Oncol
.
2015
;
33
(
33
):
3921
-
3929
.
doi:10.1200/JCO.2014.58.7618
.
58.
Zweegman
S
,
van der Holt
B
,
Mellqvist
U-H
, et al.
Melphalan, prednisone, and lenalidomide versus melphalan, prednisone, and thalidomide in untreated multiple myeloma
.
Blood
.
2016
;
127
(
9
):
1109
-
1116
.
doi:10.1182/blood-2015-11-679415
.
59.
Durie
BGM
,
Hoering
A
,
Sexton
R
, et al.
Longer term follow-up of the randomized phase III trial SWOG S0777: bortezomib, lenalidomide and dexamethasone vs. lenalidomide and dexamethasone in patients (pts) with previously untreated multiple myeloma without an intent for immediate autologous stem cell transplant (ASCT)
.
Blood Cancer J
.
2020
;
10
(
5
):
53
.
doi:10.1038/s41408-020-0311-8
.
60.
Zweegman
S
,
Stege
CAM
,
Haukas
E
, et al.
Ixazomib-thalidomide-low dose dexamethasone induction followed by maintenance therapy with ixazomib or placebo in newly diagnosed multiple myeloma patients not eligible for autologous stem cell transplantation: results from the randomized phase II HOVON-126/NMSG 21.13 trial
.
Haematologica
.
2020
;
105
(
12
):
2879
-
2882
.
doi:10.3324/haematol.2019.240374
.
61.
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
.
doi:10.1182/blood.2020008787
.
62.
Cavo
M
,
Tacchetti
P
,
Patriarca
F
, et al
;
GIMEMA Italian Myeloma Network
.
Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study
.
Lancet
.
2010
;
376
(
9758
):
2075
-
2085
.
doi:10.1016/S0140-6736(10)61424-9
.
63.
Moreau
P
,
Hulin
C
,
Macro
M
, et al.
VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial
.
Blood
.
2016
;
127
(
21
):
2569
-
2574
.
doi:10.1182/blood-2016-01-693580
.