Significant progress has been made in the treatment of multiple myeloma (MM), with the introduction of several new drugs with different mechanisms of action. The treatment of newly diagnosed MM has evolved dramatically with the development of highly effective combinations that include 1 or more of the new drugs. Despite the continuing improvement in the overall survival of patients with MM, nearly a quarter of the patients have significantly inferior survival, often driven by a combination of factors, including tumor genetics and host frailty. The focus of initial therapy remains rapid control of the disease with reversal of the symptoms and complications related to the disease with minimal toxicity and a reduction in early mortality. The selection of the specific regimen, to some extent, depends on the ability of the patient to tolerate the treatment and the underlying disease risk. It is typically guided by results of randomized clinical trials demonstrating improvements in progression-free and/or overall survival. While increasing risk calls for escalating the intensity of therapy by using quadruplet combinations that can provide the deepest possible response and the use of autologous stem cell transplant, increasing frailty calls for a reduction in the intensity and selective use of triplet or doublet regimens. The choice of subsequent consolidation treatments and maintenance approaches, including duration of treatment, also depends on these factors, particularly the underlying disease risk. The treatment approaches for newly diagnosed myeloma continue to evolve, with ongoing trials exploring bispecific antibodies as part of initial therapy and CAR T cells for consolidation.

Learning Objectives

  • Review disease characteristics that determine outcomes and contribute to the treatment decision-making process in newly diagnosed multiple myeloma.

  • Describe the efficacy and toxicity observed with various doublet, triplet, and quadruplet induction regimens.

  • Evaluate scenarios based on the initial response to treatment.

A 64-year-old man presented to his primary care physician with a 2-week history of back pain that started while he was playing golf. He has been relatively healthy, works full-time as an executive, and regularly exercises by walking at least 30 minutes daily. He has tried local measures, including local heat, lidocaine cream, and oral naproxen, with no relief. The plain film shows a compression fracture of the T8 vertebral body that was not evident on prior chest X-rays available in his medical records. Magnetic resonance imaging demonstrated compression of the T8 vertebral body with diffuse marrow enhancement involving multiple vertebral bodies, concerning for a pathological process such as myeloma. Additional workup showed a hemoglobin of 11.2 gm/dL (13.4  g/dL a year earlier), normal white blood count and platelets, and normal metabolic parameters, including normal calcium and serum creatinine. Serum protein electrophoresis with immunofixation showed a monoclonal protein peak of 1.4 gm/dL of immunoglobulin G kappa, serum free light chain assay showed kappa at 121  mg/dL and lambda at 0.3  mg/dL with a ratio of 401, and 24-hour urine protein electrophoresis demonstrated 340  mg of M spike—kappa light chain and immunoglobulin G kappa fragments. A whole-body, low-dose computed tomographic scan showed multiple small lytic lesions scattered throughout the skeleton, with no additional fractures. A bone marrow biopsy was performed, which showed diffuse marrow infiltration with kappa light chain restricted plasma cells constituting 40% of the cellularity. Fluorescence in situ hybridization studies on the plasma cells showed monosomy 13, 1q gain, and t(4;14).

Multiple myeloma is always preceded by a premalignant phase—monoclonal gammopathy of undetermined significance (MGUS)—characterized by the presence of a monoclonal protein as well as the presence of clonal plasma cells in the marrow, typically, but with no identifiable impact on the host and usually meriting no initiation of specific treatment.1,2 Additionally, many patients may have a discernable phase in between MGUS and active myeloma, termed smoldering multiple myeloma, in which concepts around early intervention continue to evolve, but considerable debate exists in terms of the timing and nature of treatment.3 Given this, an accurate diagnosis of active myeloma and distinction from MGUS and smoldering multiple myeloma remain a critical first step. The diagnostic criteria were most recently revised to include biomarkers that predicted the imminent development of end-organ damage as triggers for the initiation of treatment, a paradigm shift from the prior approach to this disease.2 The current diagnostic criteria and the essential elements of the initial workup are shown in Figure 1.

Figure 1.

Diagnostic criteria for symptomatic myeloma and suggested initial workup. BUN, blood urea nitrogen; FDG-PET/CT, 18F-fludeoxyglucose–positron emission tomography/computed tomography; FISH, fluorescence in situ hybridization; LC, light chain; LDH, lactate dehydrogenase; LLN, lower limit of normal; MRI, magnetic resonance imaging; ULN, upper limit of normal; WBMRI, whole-body magnetic resonance imaging.

Figure 1.

Diagnostic criteria for symptomatic myeloma and suggested initial workup. BUN, blood urea nitrogen; FDG-PET/CT, 18F-fludeoxyglucose–positron emission tomography/computed tomography; FISH, fluorescence in situ hybridization; LC, light chain; LDH, lactate dehydrogenase; LLN, lower limit of normal; MRI, magnetic resonance imaging; ULN, upper limit of normal; WBMRI, whole-body magnetic resonance imaging.

Close modal

In addition to establishing the diagnosis, the initial workup should also focus on characterizing the disease to understand the patient's prognosis better and individualize the initial treatment.4 MM is a heterogeneous disease with varying outcomes that are driven by a variety of factors including tumor characteristics, host factors, and interaction between the tumor clone and the host (Figure 2).5 This is clear from the current outcomes, in which the median survival of the disease has improved from 3 years to over 10 years, but with nearly a quarter of patients still living less than 3 to 4 years from diagnosis.6,7 Among the many baseline features, genetic abnormalities in the tumor cell, age, and functional status play an outsized role in determining outcomes and, in turn, have become the major determinants of therapy selection for patients with newly diagnosed MM.

Figure 2.

General approach to risk assessment in patients with newly diagnosed symptomatic MM. PC, plasma cell.

Figure 2.

General approach to risk assessment in patients with newly diagnosed symptomatic MM. PC, plasma cell.

Close modal

The genetic abnormalities in MM are well described and can be broadly categorized into structural and numerical abnormalities that can be identified through fluorescence in situ hybridization–based testing, which is currently the most commonly used and is readily available in the clinic, and mutations in individual genes that can be identified by genetic sequencing of the purified tumor cells, which is not commonly done in the current practice setting.3,5 The primary chromosomal abnormalities, those that have been identified in the earliest stages of MGUS, in turn, can be grouped into trisomies that typically include several odd-numbered chromosomes and translocations involving the Ig heavy chain locus on chromosome 14 with a common set of partner chromosomes (4, 6, 11, 16, and 20). Trisomies can be identified in approximately 40% of patients and translocations in approximately 45%, and in about 15% of patients, tumor cells with both abnormalities can be found.8 Other abnormalities, often termed secondary because they are identified later during the tumor cell evolution and are often subclonal, include deletions, gains, and amplifications as well as monosomies, and the common ones include chromosome 1q gain or amplification, 1p deletion, 13q deletion or monosomy 13, and 17p deletion or monosomy 17. Some of these are associated with a poor outcome, while others may ameliorate the negative impact of high-risk abnormalities (Figure 2). These abnormalities have been incorporated into the disease-staging systems currently in use.9,10 While these abnormalities individually do not dictate the choice of a specific drug, the presence of high-risk characteristics influence the overall treatment strategy. However, this will likely change as we get more targeted agents.

The next major determinant of treatment selection is age and functional status or frailty.11,12 Age remains an easily assessable metric but a poor indicator of the ability of an individual to withstand the intensity of a given myeloma therapy. Given the median age at diagnosis of the late sixties, many patients present with significant comorbidities as well as several social determinants of health that can influence their disease presentation as well as the ability to tolerate the treatments. Several different frailty scores are in use currently, and any of them can be used to identify frail patients. These characteristics and patient preferences determine the intensity of the initial regimen and the ability to subsequently utilize some effective therapies like autologous stem cell transplantation (ASCT).

The goal of initial treatment in MM is to achieve rapid control of the disease, which can, in turn, lead to a reversal of many of the symptoms and complications related to the disease at presentation and reduce the risk of early mortality associated with uncontrolled disease while minimizing treatment-related toxicity. This serves as the foundation for further consolidation therapy and long-term maintenance approaches designed to maximize the outcomes from the initial treatment strategy. Over the past 2 decades, the initial treatment approach has evolved but has primarily depended on effective agents such as proteasome inhibitors (PIs; bortezomib, carfilzomib, ixazomib), immunomodulatory drugs (IMiDs; thalidomide, lenalidomide), and, more recently, anti-CD38 monoclonal antibodies (MoAbs; daratumumab, isatuximab) used in various combinations.4,13 During this period the treatments have slowly evolved from the use of a doublet that included either a PI or an IMiD along with dexamethasone to triplets that combined both a PI and an IMiD and, more recently, triplets that include an anti-CD38 MoAb with lenalidomide and quadruplets that add an anti-CD38 MoAb to the combination of PI and IMiD. Other regimens have utilized these effective agents in combination with traditional drugs, including alkylating agents. The use of multidrug combinations has resulted in deeper and more rapid responses to the initial therapy. The results from these different doublet, triplet, and quadruplet regimens are shown in Tables 1, 2, and 3, respectively.

Table 1.

Doublet regimens in randomized trials of newly diagnosed multiple myelomaa

TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow up (months)
NCT0005756426
NDMM 
Td Continued until progression/ toxicity 235 63% 15.8% 7.7%
14.9 NR (75.7% at 18 mo) 18 
Placebo-dex Continued until progression/ toxicity 235 46% 43.8% 2.6%
6.5 NR (71.1% at 18 mo) 
IFM 2005-01
NCT0020068127
NDMM, age < = 65 years 
VAD + ASCT + DCEP vs no consolidation 4 cycles + ASCT +2 cycles consolidation vs no consolidation 242 77.1% after ASCT 37.2% after ASCT 8.7% after ASCT
29.7 NR (77.4% at 3 y) 32.2 
Bd + ASCT + DCEP vs no consolidation 240 80.3% after ASCT 54.3% after ASCT 16.1% after ASCT
36 NR (81.4% at 3 y) 
NCT0055192828
NDMM, age < = 65 years 
Rd + MPR + R/no maintenance Induction × 4 cycles + MPR × 6 cycles + maintenance until progression/ tolerated 132 90.9% 62.9% 18.2%
22.4 NR, 65.3% at 4 y 51.2 
Rd + ASCT + R/no maintenance Induction × 4 cycles + ASCT + maintenance till progression/ tolerated 141 92.9% 58.9% 23.4%
43 NR, 81.6% at 4 y 
RV-MM-EMN-441
NCT0109183129
NDMM, age < = 65 years 
Rd + CRd + R/PR maintenance Induction × 4 cycles + CRd × 6 cycles + maintenance until progression/ tolerated 129 89% 50% 12%
28.6 NR, 73% at 4 y 52 
Rd + ASCT + R/PR maintenance Induction × 4 cycles + ASCT + maintenance until progression/ tolerated 127 91% 54% 13%
43.3 NR, 86% at 4 y 
NCT0109319630
NDMM 
Rd + R vs PR maintenance 9 cycles + maintenance till progression/ tolerated 217 74% 34% 3% 21 NR, 58% at 4 y 39 
MPR/CPR + R vs PR maintenance 437 MPR = 71%, CPR = 68% MPR = 26%, CPR = 20.5% MPR = 3%, CPR = 0.5% 22 NR, 67% at 4 y 
FIRST
NCT0068993620
NDMM 
Rd until progression Until progression/ tolerated 535 81% 48% 22% 26 59.1 67 
Rd  ×  18 cycles 18 cycles 541 79% 47% 20% 21 62.3 
MPT 12 cycles 547 67% 30% 12% 21.9 49.1  
TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow up (months)
NCT0005756426
NDMM 
Td Continued until progression/ toxicity 235 63% 15.8% 7.7%
14.9 NR (75.7% at 18 mo) 18 
Placebo-dex Continued until progression/ toxicity 235 46% 43.8% 2.6%
6.5 NR (71.1% at 18 mo) 
IFM 2005-01
NCT0020068127
NDMM, age < = 65 years 
VAD + ASCT + DCEP vs no consolidation 4 cycles + ASCT +2 cycles consolidation vs no consolidation 242 77.1% after ASCT 37.2% after ASCT 8.7% after ASCT
29.7 NR (77.4% at 3 y) 32.2 
Bd + ASCT + DCEP vs no consolidation 240 80.3% after ASCT 54.3% after ASCT 16.1% after ASCT
36 NR (81.4% at 3 y) 
NCT0055192828
NDMM, age < = 65 years 
Rd + MPR + R/no maintenance Induction × 4 cycles + MPR × 6 cycles + maintenance until progression/ tolerated 132 90.9% 62.9% 18.2%
22.4 NR, 65.3% at 4 y 51.2 
Rd + ASCT + R/no maintenance Induction × 4 cycles + ASCT + maintenance till progression/ tolerated 141 92.9% 58.9% 23.4%
43 NR, 81.6% at 4 y 
RV-MM-EMN-441
NCT0109183129
NDMM, age < = 65 years 
Rd + CRd + R/PR maintenance Induction × 4 cycles + CRd × 6 cycles + maintenance until progression/ tolerated 129 89% 50% 12%
28.6 NR, 73% at 4 y 52 
Rd + ASCT + R/PR maintenance Induction × 4 cycles + ASCT + maintenance until progression/ tolerated 127 91% 54% 13%
43.3 NR, 86% at 4 y 
NCT0109319630
NDMM 
Rd + R vs PR maintenance 9 cycles + maintenance till progression/ tolerated 217 74% 34% 3% 21 NR, 58% at 4 y 39 
MPR/CPR + R vs PR maintenance 437 MPR = 71%, CPR = 68% MPR = 26%, CPR = 20.5% MPR = 3%, CPR = 0.5% 22 NR, 67% at 4 y 
FIRST
NCT0068993620
NDMM 
Rd until progression Until progression/ tolerated 535 81% 48% 22% 26 59.1 67 
Rd  ×  18 cycles 18 cycles 541 79% 47% 20% 21 62.3 
MPT 12 cycles 547 67% 30% 12% 21.9 49.1  
a

Only regimens with any 1 of the PIs, IMiDs, or anti-CD38 MoAbs are included.

CPR, cyclophosphamide, prednisone, lenalidomide; CR, complete response; CVD, cardiovascular disease; dex, dexamethasone; EFS, event-free survival; MPR, melphalan, prednisone, lenalidomide; MPT, melphalan; NDMM, newly diagnosed multiple myeloma; NR, not reached; ORR, overall response rate; PR, prednisone and lenalidomide; > = PR, partial response or better; R, lenalidomide; Rd, lenalidomide and dexamethasone; s/c, subcutaneous; V, bortezomib; VAD, vincristine, doxorubicin, dexamethasone; Vd, bortezomib and dexamethasone; VGPR, very good partial response.

Table 2.

Triplet regimens in randomized trials of newly diagnosed multiple myelomaa

TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow-up (months)
IFM 2009
NCT0119106014
NDMM, age <65 y 
VRd + ASCT + R maintenance 3 cycles +2 cycles post ASCT + R maintenance × 1 y 350 98% 88% 59% 47.3 NR, 60.2% at 8 y 93 
VRd + R maintenance 8 cycles + R maintenance × 1 y 350 97% 77% 48% 35.0 NR, 62.2% at 8 years 
DETERMINATION15
NCT01208662
NDMM, age <65 y 
VRd + ASCT + R maintenance 3 cycles +2 cycles post ASCT + R maintenance until progression 365 98% 83% 47% 67.5 NR, 80.7% at 5 y 76 
VRd + R maintenance 8 cycles + R maintenance until progression 357 95% 80% 42% 46.2 NR, 79.2% at 5 y 
GIMEMA-MMY-3006
NCT0113448431
NDMM, age <65 y 
VTd + double ASCT + VTd consolidation + dex maintenance 3 cycles +2 cycles consolidation post double ASCT + dexamethasone maintenance until progression/ tolerated 236 96% 89% 58% 60 NR 124.1 
Td + double ASCT + Td consolidation + dex maintenance 238 89% 74% 41% 41 110 
GEM05menos65
NCT0046174732
NDMM, age <65 y 
VTd + IFNα2b/T/ VT maintenance 24-week induction + ASCT + maintenance × 3 years 130 85% after induction 60% after induction 35% after induction 56.1 60% at 8 y 70.6 
Td + IFNα2b/T/ VT maintenance 127 62% after induction 29% after induction 14% after induction 29.2 
VBMCP/VBAD/ bortezomib + IFNα2b/T/VT maintenance 129 75% after induction 36% after induction 21% after induction 39.9 
NCT0091089733
NDMM, age <65 y 
VTd + ASCT 4 cycles + ASCT 100 89% after ASCT 74% after ASCT 29% after ASCT 26  32 
Vd + ASCT 99 86% after ASCT 58% after ASCT 31% after ASCT 30 
IFM2013-04
NCT0156453734
NDMM, age <65 y 
VCd 4 cycles 169 83.4% after induction 56.2% after induction 8.9% after induction 
VTd 4 cycles 169 92.3% after induction 66.3% after induction 13% after induction  
S0777
NCT0064422817
NDMM 
VRd + Rd maintenance 8 cycles + Rd maintenance until progression/ tolerated 225 90.2% 74.9% 24.2% 41 NR 84 
Rd + Rd maintenance 6 cycles + Rd maintenance until progression/ tolerated 235 78.8% 53.2% 12.1% 29 69 
UPFRONT
NCT0050741635
NDMM 
Vd + V maintenance 8 cycles induction +35 weeks maintenance 168 73% 37% 3% 14.7 49.8 42.7 
VTd + V maintenance 167 80% 51% 4% 15.4 51.5 
VMP + V maintenance 167 70% 41% 4% 17.3 53.1 
ENDURANCE
NCT0186355025
NDMM 
VRd + R maintenance 12 cycles + maintenance × 2 years vs until progression/ tolerated 542 84% after induction 65% after induction 15% after induction 34.4 mo NR, 84% at 3 y 26 
KRd + R maintenance 9 cycles + maintenance × 2 years vs until progression/ tolerated 545 87% after induction 74% after induction 18% after induction 34.6 mo NR, 86% at 3 y 
TOURMALINE-MM2
NCT0185052436
NDMM 
IRd 18 cycles + maintenance until progression/ tolerated 351 82.1% 63% 25.6% 35.3 NR ~58 
Placebo-Rd 
354 79.7% 47.7% 14.1% 21.8 NR 
MAIA
NCT0225217237
NDMM 
DRd Treatment continued until progression/ tolerated 368 92.9% 81% 51% NR NR 56.2 
Rd 
369 81.6% 57% 30% 34.4 NR 
TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow-up (months)
IFM 2009
NCT0119106014
NDMM, age <65 y 
VRd + ASCT + R maintenance 3 cycles +2 cycles post ASCT + R maintenance × 1 y 350 98% 88% 59% 47.3 NR, 60.2% at 8 y 93 
VRd + R maintenance 8 cycles + R maintenance × 1 y 350 97% 77% 48% 35.0 NR, 62.2% at 8 years 
DETERMINATION15
NCT01208662
NDMM, age <65 y 
VRd + ASCT + R maintenance 3 cycles +2 cycles post ASCT + R maintenance until progression 365 98% 83% 47% 67.5 NR, 80.7% at 5 y 76 
VRd + R maintenance 8 cycles + R maintenance until progression 357 95% 80% 42% 46.2 NR, 79.2% at 5 y 
GIMEMA-MMY-3006
NCT0113448431
NDMM, age <65 y 
VTd + double ASCT + VTd consolidation + dex maintenance 3 cycles +2 cycles consolidation post double ASCT + dexamethasone maintenance until progression/ tolerated 236 96% 89% 58% 60 NR 124.1 
Td + double ASCT + Td consolidation + dex maintenance 238 89% 74% 41% 41 110 
GEM05menos65
NCT0046174732
NDMM, age <65 y 
VTd + IFNα2b/T/ VT maintenance 24-week induction + ASCT + maintenance × 3 years 130 85% after induction 60% after induction 35% after induction 56.1 60% at 8 y 70.6 
Td + IFNα2b/T/ VT maintenance 127 62% after induction 29% after induction 14% after induction 29.2 
VBMCP/VBAD/ bortezomib + IFNα2b/T/VT maintenance 129 75% after induction 36% after induction 21% after induction 39.9 
NCT0091089733
NDMM, age <65 y 
VTd + ASCT 4 cycles + ASCT 100 89% after ASCT 74% after ASCT 29% after ASCT 26  32 
Vd + ASCT 99 86% after ASCT 58% after ASCT 31% after ASCT 30 
IFM2013-04
NCT0156453734
NDMM, age <65 y 
VCd 4 cycles 169 83.4% after induction 56.2% after induction 8.9% after induction 
VTd 4 cycles 169 92.3% after induction 66.3% after induction 13% after induction  
S0777
NCT0064422817
NDMM 
VRd + Rd maintenance 8 cycles + Rd maintenance until progression/ tolerated 225 90.2% 74.9% 24.2% 41 NR 84 
Rd + Rd maintenance 6 cycles + Rd maintenance until progression/ tolerated 235 78.8% 53.2% 12.1% 29 69 
UPFRONT
NCT0050741635
NDMM 
Vd + V maintenance 8 cycles induction +35 weeks maintenance 168 73% 37% 3% 14.7 49.8 42.7 
VTd + V maintenance 167 80% 51% 4% 15.4 51.5 
VMP + V maintenance 167 70% 41% 4% 17.3 53.1 
ENDURANCE
NCT0186355025
NDMM 
VRd + R maintenance 12 cycles + maintenance × 2 years vs until progression/ tolerated 542 84% after induction 65% after induction 15% after induction 34.4 mo NR, 84% at 3 y 26 
KRd + R maintenance 9 cycles + maintenance × 2 years vs until progression/ tolerated 545 87% after induction 74% after induction 18% after induction 34.6 mo NR, 86% at 3 y 
TOURMALINE-MM2
NCT0185052436
NDMM 
IRd 18 cycles + maintenance until progression/ tolerated 351 82.1% 63% 25.6% 35.3 NR ~58 
Placebo-Rd 
354 79.7% 47.7% 14.1% 21.8 NR 
MAIA
NCT0225217237
NDMM 
DRd Treatment continued until progression/ tolerated 368 92.9% 81% 51% NR NR 56.2 
Rd 
369 81.6% 57% 30% 34.4 NR 
a

Only regimens with any 2 of the PIs, IMiDs, or anti-CD38 MoAbs are included.

CPR, cyclophosphamide, prednisone, lenalidomide; CR, complete response; CVD, cardiovascular disease; Dara, daratumumab; dex, dexamethasone; DRd, daratumumab, lenalidomide, dexamethasone; EFS, event-free survival; I, ixazomib; IRd, ixazomib, lenalidomide, dexamethasone; K, carfilzomib; KRd, carfilzomib, lenalidomide, dexamethasone; M, melphalan; MP, melphalan and prednisone; MPR, melphalan, prednisone, lenalidomide; MPT, melphalan, prednisone, thalidomide; NDMM, newly diagnosed multiple myeloma; NR, not reached; ORR, overall response rate; PR, prednisone and lenalidomide; > = PR, partial response or better; R, lenalidomide; Rd, lenalidomide and dexamethasone; s/c, subcutaneous; V, bortezomib; VAD, vincristine, doxorubicin, dexamethasone; Vd, bortezomib and dexamethasone; VGPR, very good partial response; VMP, bortezomib, melphalan, prednisone; VMPT, bortezomib, melphalan, prednisone, thalidomide; VRd, bortezomib, lenalidomide, dexamethasone; VT, bortezomib and thalidomide; VTd, bortezomib, thalidomide, dexamethasone.

Table 3.

Quadruplet regimens in randomized trials of newly diagnosed multiple myelomaa

TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow up (months)
CASSIOPEIA
NCT0254138318
NDMM, age <65 y 
VTd + ASCT + VTd consolidation + Dara vs no maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance × 2 years 542 89.9% (100 days post ASCT) 78% (100 days post ASCT) 26% (100 days post ASCT) NR (85% at 18 months) NR 18.8 
Dara-VTd + ASCT + Dara-VTd consolidation + Dara vs no maintenance 543 92.6% (100 days post ASCT) 83% (100 days post ASCT) 39% (100 days post ASCT) NR (93% at 18 months) NR 
Myeloma XI+
ISRCTN4940785238
NDMM 
KRdc + ASCT + R/no maintenance Minimum 4 cycles induction + ASCT + maintenance until progression/ tolerated 526 97.7% 91.9% 31% NR 34.5 
Rdc/Tdc + ASCT + R/no maintenance 530 95.3% 79.3% 24% 36.2 
GMMG-HD7
NCT0361773139
NDMM, age <70 y 
VRd + ASCT 3 cycles + ASCT 329 83.6% after induction 60.5% after induction 21.6% after induction 
Isa-VRd + ASCT 331 90% after induction 77.3% after induction 24.2% after induction 
PERSEUS19
NCT03710603
NDMM, age <70 y 
VRd + ASCT + VRd consolidation + R maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance until progression/ tolerated 354 93.8% 89.3% 70.1% NR, 67.7% at 4 years NR 47.5 
Dara-VRd + ASCT +Dara-VRd consolidation+ Dara R maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance until progression/ tolerated (Dara stopped if MRD− at 2 years) 355 96.6% 95.2% 87.9% NR, 84.3% at 4 y NR 
IMROZ21
NCT03319667
NDMM ≤80 y 
VRd with R maintenance 4 (6-week) cycles of VRd followed by 4-week cycles of Rd until disease progression 181 92.3 82.9% 64.1% 45.2% at 5 y NR  
Isatuximab VRd with Isa-R maintenance 4 (6-week) cycles of Isa-VRd followed by 4-week cycles of Isa-Rd until disease progression 264 91.3 89.1% 74.7% 63.2% at 5 y NR 
BENEFIT22
NCT04751877
NDMM ≤80 y 
Isatuximab-Rd with Isa-R maintenance Isa-Rd
(cycle 1-12), Isa-R (cycle 13-18), Isa (cycle 19 to progression) 
135 78% (18 mo) 70% (18 mo) 31% (18 mo) 80% at 2 y NR  
Isatuximab VRd with Isa-R maintenance Isa-Rd
Bortezomib (cycle 1-12), Isa-R-bortezomib (cycle 13-18), Isa (cycle 19 to progression) 
135 85% (18 mo) 82% (18 mo) 58% (18 mo) 91.5% at 2 y NR 
TrialRegimenDuration of treatmentNORR> = VGPR> = CRMedian PFS (months)Median OS (months)Median follow up (months)
CASSIOPEIA
NCT0254138318
NDMM, age <65 y 
VTd + ASCT + VTd consolidation + Dara vs no maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance × 2 years 542 89.9% (100 days post ASCT) 78% (100 days post ASCT) 26% (100 days post ASCT) NR (85% at 18 months) NR 18.8 
Dara-VTd + ASCT + Dara-VTd consolidation + Dara vs no maintenance 543 92.6% (100 days post ASCT) 83% (100 days post ASCT) 39% (100 days post ASCT) NR (93% at 18 months) NR 
Myeloma XI+
ISRCTN4940785238
NDMM 
KRdc + ASCT + R/no maintenance Minimum 4 cycles induction + ASCT + maintenance until progression/ tolerated 526 97.7% 91.9% 31% NR 34.5 
Rdc/Tdc + ASCT + R/no maintenance 530 95.3% 79.3% 24% 36.2 
GMMG-HD7
NCT0361773139
NDMM, age <70 y 
VRd + ASCT 3 cycles + ASCT 329 83.6% after induction 60.5% after induction 21.6% after induction 
Isa-VRd + ASCT 331 90% after induction 77.3% after induction 24.2% after induction 
PERSEUS19
NCT03710603
NDMM, age <70 y 
VRd + ASCT + VRd consolidation + R maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance until progression/ tolerated 354 93.8% 89.3% 70.1% NR, 67.7% at 4 years NR 47.5 
Dara-VRd + ASCT +Dara-VRd consolidation+ Dara R maintenance 4 cycles induction + ASCT +2 cycles consolidation + maintenance until progression/ tolerated (Dara stopped if MRD− at 2 years) 355 96.6% 95.2% 87.9% NR, 84.3% at 4 y NR 
IMROZ21
NCT03319667
NDMM ≤80 y 
VRd with R maintenance 4 (6-week) cycles of VRd followed by 4-week cycles of Rd until disease progression 181 92.3 82.9% 64.1% 45.2% at 5 y NR  
Isatuximab VRd with Isa-R maintenance 4 (6-week) cycles of Isa-VRd followed by 4-week cycles of Isa-Rd until disease progression 264 91.3 89.1% 74.7% 63.2% at 5 y NR 
BENEFIT22
NCT04751877
NDMM ≤80 y 
Isatuximab-Rd with Isa-R maintenance Isa-Rd
(cycle 1-12), Isa-R (cycle 13-18), Isa (cycle 19 to progression) 
135 78% (18 mo) 70% (18 mo) 31% (18 mo) 80% at 2 y NR  
Isatuximab VRd with Isa-R maintenance Isa-Rd
Bortezomib (cycle 1-12), Isa-R-bortezomib (cycle 13-18), Isa (cycle 19 to progression) 
135 85% (18 mo) 82% (18 mo) 58% (18 mo) 91.5% at 2 y NR 
a

Only regimens with any 3 of the PIs, IMiDs, or anti-CD38 MoAbs are included.

CR, complete response; Dara, daratumumab; Dara-VTd, daratumumab, bortezomib, thalidomide, dexamethasone; dex, dexamethasone; Isa, isatuximab; Isa-VRd, isatuximab, bortezomib, lenalidomide, dexamethasone; NDMM, newly diagnosed multiple myeloma; NR, not reached; ORR, overall response rate; R, lenalidomide; s/c, subcutaneous; V, bortezomib; VGPR, very good partial response; VRd, bortezomib, lenalidomide, dexamethasone.

The traditional approach had been to classify patients as transplant eligible or ineligible, often based on age and the presence of comorbidities, before deciding on the initial treatment regimen. This was primarily driven by the potential impact of initial therapy on the ability to collect stem cells, a valid concern when alkylating agents were a part of the initial treatments. As a result of this approach, the development of initial therapy has evolved on 2 parallel tracks, 1 for those considered eligible for ASCT, in which case patients received 4 to 6 cycles of induction therapy followed by ASCT used as consolidation. For those patients who were not considered eligible for ASCT, the treatment regimens over successive phase 3 trials initially explored the addition of novel therapies to melphalan-based regimens to demonstrate benefit. With subsequent trials demonstrating the ability to eliminate melphalan as part of the initial therapy, there has been increasing convergence in the type of initial treatment irrespective of the transplant eligibility. This, along with randomized trials demonstrating equivalent benefits in terms of overall survival (OS) for early or delayed ASCT, has allowed us to use similar therapies for initial control of the disease before making a decision on transplant eligibility.14,15 This has allowed providers to make a more accurate assessment of the ability of the patient to undergo ASCT and for patients to make an informed decision about the timing of ASCT, if they choose to pursue it.

For those patients who can potentially undergo an ASCT, the approach has been to increase the intensity of initial treatment to achieve rapid and deep control of the disease before proceeding to transplant. For these patients, the treatment regimens have evolved from a doublet to a triplet and eventually to a quadruplet. Initial studies demonstrated the superiority of the lenalidomide and dexamethasone (Rd) combination as an effective initial therapy.16 Subsequently, bortezomib was added to Rd, and the triplet regimen was shown to improve progression-free survival (PFS) and OS when used as an initial therapy.17 The randomized trials that explored the use of ASCT as consolidation increasingly used the VRd combination as the induction therapy before transplant. More recently, several phase 2 and 3 trials demonstrated improved PFS outcomes associated with adding daratumumab.18,19 In contrast, in patients not considered fit for ASCT, Rd was compared with melphalan, prednisone, and thalidomide and was shown to have improved PFS.20 Phase 3 trials initially explored triplets in this patient population, given the concern with tolerability of more intense therapies in older patients. The addition of daratumumab to lenalidomide and dexamethasone led to improved PFS and OS with these 3-drug regimens, making this the current standard treatment approach for these patients.18,19 More recently, 2 large phase 3 trials demonstrated the feasibility of using quadruplet regimens in an older population until the age of 80 years; both the IMROZ and BENEFIT trials demonstrated improved response rates and deeper response with the use of quadruplets compared with triplets, with the IMROZ trial demonstrating an improved OS as well.21,22 

The choice between doublets, triplets, or quadruplets is increasingly made based on the patient's ability to tolerate the treatment while also considering the underlying genomic risk (Figure 3), fully acknowledging that these 2 parameters exist in a continuum.23,24 As a result, an accurate assessment of the patient's functional status at the time of diagnosis is key to making this decision. It is also important to keep in mind that in patients who are thought to be unable to tolerate intense therapy, control of the disease with a less intense regimen can often improve the functional status to a point where another drug can be added to the combination. Given the results from the phase 3 trials, most patients should receive at least a triplet consisting of 2 of these 3 classes of drugs plus dexamethasone, modified to the patient's tolerance. In patients in their late 80s and older, as well as those who are quite frail, a doublet can certainly be considered with the intent to add the third drug if their functional status improves with treatment. For most patients, the current standard approach is to use a quadruplet regimen containing an anti-CD38 MoAb, a PI, an IMiD, and dexamethasone for the initial treatment of MM. A triplet consisting of an anti-CD38 MoAbs and lenalidomide can be considered if the 4-drug combination cannot be tolerated or transplant is not considered an option for whatever reason. Recent phase 3 trials have demonstrated an equivalent benefit using isatuximab, another anti-CD 38 MoAb similar to daratumumab. Two randomized trials evaluated carfilzomib instead of bortezomib and did not demonstrate an improved survival outcome despite deeper responses, likely due to the increased toxicity associated with carfilzomib.25 However, phase 2 trials have utilized carfilzomib in combination with an anti-CD 38 MoAb, lenalidomide, and dexamethasone and have demonstrated deep responses and certainly remain an option if bortezomib cannot be used.

Figure 3.

Approach to initial treatment selection in patients with newly diagnosed symptomatic MM accounting for frailty and risk.

Figure 3.

Approach to initial treatment selection in patients with newly diagnosed symptomatic MM accounting for frailty and risk.

Close modal

Several presenting characteristics might dictate an alternate approach or regimen for the initial treatment of MM. Renal insufficiency can be present in up to a third of the patients with newly diagnosed MM, with approximately 5% of patients requiring renal replacement therapy at least through the initial weeks after diagnosis. While lenalidomide can be used on patients with renal dysfunction, it can be difficult given their fluctuating renal function as well as the time it often takes to get this started in the inpatient setting. Given the importance of rapid disease control in patients with renal insufficiency, regimens such as daratumumab combined with bortezomib, cyclophosphamide, and dexamethasone have been used with good outcomes. The additive value of cyclophosphamide has not been specifically explored in this quadruplet regimen, though 1 phase 3 trial did not demonstrate the benefit of adding cyclophosphamide to bortezomib and dexamethasone. In these patients, cyclophosphamide should be replaced with lenalidomide after the initial cycle and once the renal function improves, as this regimen has been associated with better outcomes.

Another clinical scenario that can bring challenges includes patients who present with bulky extramedullary disease or plasma cell leukemia. Plasma cell leukemia, now defined as greater than or equal to 5% circulating plasma cells, can be associated with cytopenias that are often challenging when using current treatment regimens. For some of these patients, there may be a role for combinations of traditional cytotoxic chemotherapy drugs for rapid disease control; regimens such as VDTPACE (bortezomib, dexamethasone, thalidomide, cisplatin, adriamycin, cyclophosphamide, and etoposide) have been used in this setting. After the initial disease debulking, these patients can switch to a more contemporary quadruplet regimen, as described above. While cytotoxic therapy agents such as anthracyclines can significantly impact the tumor, leading to rapid response induction, it is unclear whether this approach significantly impacts the long run. Other approaches have been explored in clinical trials like the OPTIMUM trial from the United Kingdom, where a 5-drug combination of cyclophosphamide added to DRVd has been used for high-risk patients.

Another setting where the quadruplet with lenalidomide can pose challenges is in those patients with concomitant light chain amyloidosis. Particularly with cardiac involvement, the use of lenalidomide has been associated with increased complications. In this setting, a combination of daratumumab and bortezomib, cyclophosphamide, and dexamethasone can be used for initial disease control.

Once the disease is under good control, whether with the quadruplet, triplet, or doublet, maintenance therapy with or without additional consolidation represents the current approach. The ideal duration of therapy has not been well defined from randomized clinical trials, but ongoing trials explore either stopping treatment after a defined duration or using a response depth adapted duration of therapy to allow a sustained disease response. For those patients fit enough to undergo ASCT and who desire this procedure, 4 to 6 cycles of induction therapy can be followed by a single ASCT. Post transplant, patients are frequently placed on maintenance therapy, typically with the single agent lenalidomide, which is often continued until disease progression or until the patient is unable to tolerate the medication. Many of the recent phase 3 trials have explored adding daratumumab to lenalidomide, but the additional benefit remains undefined, and ongoing trials directly comparing DR with R will provide the definitive answer. In patients with high-risk disease, following ASCT a 2-drug maintenance regimen is typically used, most commonly the combination of bortezomib along with lenalidomide. For those patients who are not undergoing an ASCT, the triplet regimen is often continued for 9 to 12 months, at which time the dexamethasone is discontinued, and the dose and schedule of the remaining drugs are modified to allow for prolonged therapy.

The initial treatment approach to myeloma continues to evolve rapidly with the advent of novel therapies, including immunotherapies such as bispecific antibodies. Ongoing clinical trials all explore achieving the deepest possible response with manageable toxicity to allow for prolonged disease control and, possibly, a defined duration of therapy in the future. This is being explored with novel immunotherapy, such as adding bispecific antibodies to the initial therapy, either as a replacement for 1 of the current drugs in the quadruplet or in addition to a doublet. Other therapies, such as CAR T and bispecific antibodies, are also being explored as potential consolidation, either as a replacement for or in addition to a stem cell transplant, depending upon the ability of the patient to undergo specific therapy as well as the underlying risks.

Shaji K. Kumar: consultancy: AbbVie, Amgen, ArcellX, Beigene, Bristol Myers Squibb, Carsgen, Epizyme, Glycostem, GSK, Janssen, K36, Menarini, Moderna, Pfizer, Regeneron, Roche- Genentech, Sanofi, Takeda, Telogenomics, Trillium, Window Therapeutics, Antengene, Calyx, CVS Caremark, BD Biosciences; clinical trial support to institution: AbbVie, Amgen, AstraZeneca, Bristol Myers Squibb, Carsgen, GSK, Gracell Bio, Janssen, Oricell, Roche-Genentech, Sanofi, Takeda, Telogenomics.

Shaji K. Kumar: Nothing to disclose.

1.
Kumar
SK
,
Rajkumar
V
,
Kyle
RA
, et al.
Multiple myeloma
.
Nat Rev Dis Primers
.
2017
;
3
:17046.
2.
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
.
3.
Mateos
MV
,
Kumar
S
,
Dimopoulos
MA
, et al.
International Myeloma Working Group risk stratification model for smoldering multiple myeloma (SMM)
.
Blood Cancer J
.
2020
;
10
(
10
):
102
.
4.
Kumar
SK
,
Callander
NS
,
Adekola
K
, et al.
Multiple myeloma, Version 2.2024, NCCN Clinical Practice Guidelines in Oncology
.
J Natl Compr Canc Netw
.
2023
;
21
(
12
):
1281
-
1301
.
5.
Kumar
SK
,
Rajkumar
SV
.
The multiple myelomas—current concepts in cytogenetic classification and therapy
.
Nat Rev Clin Oncol
.
2018
;
15
(
7
):
409
-
421
.
6.
Binder
M
,
Nandakumar
B
,
Rajkumar
SV
, et al.
Mortality trends in multiple myeloma after the introduction of novel therapies in the United States
.
Leukemia
.
2022
;
36
(
3
):
801
-
808
.
7.
Corre
J
,
Perrot
A
,
Hulin
C
, et al
;
Intergroupe Francophone du Myélome
.
Improved survival in multiple myeloma during the 2005-2009 and 2010-2014 periods
.
Leukemia
.
2021
;
35
(
12
):
3600
-
3603
.
8.
Kumar
S
,
Fonseca
R
,
Ketterling
RP
, et al.
Trisomies in multiple myeloma: impact on survival in patients with high-risk cytogenetics
.
Blood
.
2012
;
119
(
9
):
2100
-
2105
.
9.
D'Agostino
M
,
Cairns
DA
,
Lahuerta
JJ
, et al.
Second revision of the International Staging System (R2-ISS) for overall survival in multiple myeloma: a European Myeloma Network (EMN) report within the HARMONY project
.
J Clin Oncol
.
2022
;
40
(
29
):
3406
-
3418
.
10.
Abdallah
NH
,
Binder
M
,
Rajkumar
SV
, et al.
A simple additive staging system for newly diagnosed multiple myeloma
.
Blood Cancer J
.
2022
;
12
(
1
):
21
.
11.
Pawlyn
C
,
Khan
AM
,
Freeman
CL
.
Fitness and frailty in myeloma
.
Hematology Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
337
-
348
.
12.
Mian
H
,
Wildes
TM
,
Vij
R
,
Pianko
MJ
,
Major
A
,
Fiala
MA
.
Dynamic frailty risk assessment among older adults with multiple myeloma: a population-based cohort study
.
Blood Cancer J
.
2023
;
13
(
1
):
76
.
13.
Dimopoulos
MA
,
Moreau
P
,
Terpos
E
, et al.
Multiple myeloma: EHA-ESMO clinical practice guidelines for diagnosis, treatment and follow-up
.
Hemasphere
.
2021
;
5
(
2
):
e528
.
14.
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
.
15.
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
.
16.
Rajkumar
SV
,
Jacobus
S
,
Callander
NS
, et al
;
Eastern Cooperative Oncology Group
.
Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial
.
Lancet Oncol
.
2010
;
11
(
1
):
29
-
37
.
17.
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
.
18.
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
.
19.
Sonneveld
P
,
Dimopoulos
MA
,
Boccadoro
M
, et al
;
PERSEUS Trial Investigators
.
Daratumumab, bortezomib, lenalidomide, and dexamethasone for multiple myeloma
.
N Engl J Med
.
2024
;
390
(
4
):
301
-
313
.
20.
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
.
21.
Facon
T
,
Dimopoulos
MA
,
Leleu
XP
, et al.
Isatuximab, bortezomib, lenalidomide, and dexamethasone for multiple myeloma [published online 3 June 2024]
.
N Engl J Med
.
22.
Leleu
X
,
Hulin
C
,
Lambert
J
, et al.
Isatuximab, lenalidomide, dexamethasone and bortezomib in transplant-ineligible multiple myeloma: the randomized phase 3 BENEFIT trial
.
Nat Med
.
2024
;
30
(
8
):
2235
-
2241
.
23.
Goldman-Mazur
S
,
Kumar
SK
.
Current approaches to management of high-risk multiple myeloma
.
Am J Hematol
.
2021
;
96
(
7
):
854
-
871
.
24.
Coulson
AB
,
Royle
KL
,
Pawlyn
C
, et al.
Frailty-adjusted therapy in transplant non-eligible patients with newly diagnosed multiple myeloma (FiTNEss (UK-MRA Myeloma XIV Trial)): a study protocol for a randomised phase III trial
.
BMJ Open
.
2022
;
12
(
6
):
e056147
.
25.
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
.
26.
Rajkumar
SV
,
Rosiñol
L
,
Hussein
M
, et al.
Multicenter, randomized, double-blind, placebo-controlled study of thalidomide plus dexamethasone compared with dexamethasone as initial therapy for newly diagnosed multiple myeloma
.
J Clin Oncol
.
2008
;
26
(
13
):
2171
-
2177
.
27.
Harousseau
JL
,
Attal
M
,
Avet-Loiseau
H
, et al.
Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial
.
J Clin Oncol
.
2010
;
28
(
30
):
4621
-
4629
.
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
.
29.
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
.
30.
Magarotto
V
,
Bringhen
S
,
Offidani
M
, et al.
Triplet vs doublet lenalidomide-containing regimens for the treatment of elderly patients with newly diagnosed multiple myeloma
.
Blood
.
2016
;
127
(
9
):
1102
-
1108
.
31.
Tacchetti
P
,
Pantani
L
,
Patriarca
F
, et al
;
Gruppo Italiano Malattie Ematologiche dell'Adulto Italian Myeloma Network
.
Bortezomib, thalidomide, and dexamethasone followed by double autologous haematopoietic stem-cell transplantation for newly diagnosed multiple myeloma (GIMEMA-MMY-3006): long-term follow-up analysis of a randomised phase 3, open-label study
.
Lancet Haematol
.
2020
;
7
(
12
):
e861
-
e873
.
32.
Rosiñol
L
,
Oriol
A
,
Teruel
AI
, et al.
;
Programa para el Estudio y la Terapéutica de las Hemopatías Malignas/Grupo Español de Mieloma (PETHEMA/GEM) Group
.
Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study
.
Blood
.
2012
;
120
(
8
):
1589
-
1596
.
33.
Moreau
P
,
Avet-Loiseau
H
,
Facon
T
, et al.
Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma
.
Blood
.
2011
;
118
(
22
):
5752
-
5758
; quiz 5982.
34.
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
.
35.
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
.
36.
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
.
37.
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
.
38.
Jackson
GH
,
Pawlyn
C
,
Cairns
DA
, et al.
;
UK NCRI Haemato-Oncology Clinical Studies Group
.
Carfilzomib, lenalidomide, dexamethasone, and cyclophosphamide (KRdc) as induction therapy for transplant-eligible, newly diagnosed multiple myeloma patients (Myeloma XI+): interim analysis of an open-label randomised controlled trial
.
PLoS Med
.
2021
;
18
(
1
):
e1003454
.
39.
Goldschmidt
H
,
Mai
EK
,
Bertsch
U
, et al
;
German-Speaking Myeloma Multicenter Group (GMMG) HD7 Investigators
.
Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active- controlled, phase 3 trial
.
Lancet Haematol
.
2022
;
9
(
11
):
e810
-
e821
.