The introduction of proteasome inhibitor and immunomodulatory drugs has considerably changed the treatment paradigm of multiple myeloma. Autologous stem cell transplantation (ASCT) is superior to conventional chemotherapy and is considered the standard of care for patients younger than 65 years. Nevertheless, the favorable results shown by multidrug inductions, consolidations, and long-term maintenance approaches have challenged the role of ASCT. This article provides an overview of recent and ongoing clinical trials and aims to define the role of ASCT in the era of novel agents.

Multiple myeloma (MM) is a plasma cell malignancy, accounting for 1% of all cancers and 10% of hematologic neoplasms. Incidence increases greatly with age: the median age at diagnosis is 70 years, with 35% of patients younger than 65 years.1  In the Western countries, the annual age-adjusted incidence has reached 5.6 cases per 100 000 persons because of the aging of the general population. In the 1990s, treatment efficacy was improved by the use of autologous stem cell transplantation (ASCT) and subsequently by the introduction of proteasome inhibitors and immunomodulatory agents (IMiDs). In the last decade, survival of patients younger than 50 years with newly diagnosed MM has significantly improved, with 10-year survival rate increasing from 24.5% to 41.3%.2  However, MM is characterized by multiple relapses. Genomic instability and clonal heterogeneity cause the selection of more aggressive subclones responsible for drug resistance and finally dismal outcome.3  For these reasons, effective treatment should be concentrated at the early phases of disease, when clones are more drug sensitive, long-lasting remissions are more frequent, and serious adverse events are less prominent. This approach significantly improves quality of life and may ultimately prolong overall survival (OS).

Achievement of profound cytoreduction and durable response

The efficacy of treatment is mainly related to the achievement of a durable response. The achievement of a complete response (CR) is associated with prolongation of progression-free survival (PFS) and OS in both young and elderly patients.4,5  Before the introduction of bortezomib and IMiDs, an analysis of 4990 patients with MM receiving ASCT, including 2991 patients in prospective studies, showed that patients achieving CR after transplantation had longer event-free survival (EFS) and OS compared with patients achieving partial response (PR) only: median OS was 59-88.6 months in patients with CR compared with 39-68 months in patients with PR (P < .001).4  Of note, a recent study reported that 35% of patients attaining CR were alive at 12 years after a median follow-up of 153 months.6  In the elderly patients treated with either conventional chemotherapy or combinations, including thalidomide or bortezomib, the achievement of CR confirmed to be an independent predictor of longer outcome: the 3-year OS rose from 67% in patients in PR to 91% in patients in CR (P < .001), regardless of age, International Staging System stage, and treatment received.5  Conventional chemotherapy, including thalidomide or bortezomib, has shown similar results to high-dose therapy and ASCT. In MM, CR is defined by negative serum and urine immunofixation, disappearance of any soft tissue plasmacytoma, and < 5% plasma cell on bone marrow examination. This definition has low sensitivity and is somehow inappropriate, because residual tumor is always present despite the achievement of CR. More sensitive techniques are now showing better correlation between a deeper response level and an improved outcome.7,8  In patients with immunofixation-negative CR the 3-year PFS was 50% as opposed to 95% in patients with immunophenotypic CR (P = .02).7  However, even in patients with stringent, immunophenotypic, or molecular CR, late relapses occur, showing the need for adequate biomarkers to detect the eradication of the tumor clone in vivo. In a recent analysis, after a median follow-up of 65 months, the 6-year PFS was 46%, despite the previous achievement of a molecular CR. It is important not only to achieve CR but also to maintain a sustained CR. Patients with sustained CR for ≥ 3 years had a longer OS than patients who had unsustained CR (5-year OS, 82% vs 24%; P < .001).9  These data support a treatment strategy tailored to increase both CR rates and duration of response.

Early treatment discontinuation

Excessive toxicities are the main cause of early discontinuation that significantly affect cumulative dose intensity and efficacy. In the Total Therapy 3 (TT3) program, the premature discontinuation of bortezomib increased the risk of progression by 6 times (P < .001).10  In the exploratory analysis of 134 patients receiving twice-weekly bortezomib compared with 369 patients on the once-weekly schedule, the twice-weekly bortezomib schedule resulted in a higher rate of nonhematologic grade 3-4 adverse events (51% vs 36%; P = .003), mainly peripheral neuropathy (16% vs 3%; P = .001). The twice-weekly schedule had a higher rate of discontinuation (15% vs 5%; P < .001), consequently the cumulative delivered dose of bortezomib was similar (40.1 mg/m2 for twice-weekly schedule and 39.4 mg/m2 for once-weekly schedule) and so was the 3-year PFS rate (47% vs 50%; P value is not significant).11 

Low-dose dexamethasone in combination with lenalidomide (Ld) reduced the frequency of adverse events and improved treatment adherence compared with high-dose dexamethasone with lenalidomide (LD): 30% of patients in the Ld group remained on treatment for > 1 year compared with 14% in the LD group. The 1-year OS was 96% for the Ld group compared with 87% for the LD group (P < .001).12  In a randomized trial, the cumulative dose intensity of the combination melphalan-prednisone-lenalidomide followed by lenalidomide maintenance (MPL-L) was 88% in patients younger than 75 years but only 56% in those older than 75 years, with a negative effect on median PFS.13  These data clearly show the need to avoid excessive toxicities, to reduce discontinuation rate to < 20%, and to maintain cumulative dose intensity to ≥ 80%, delivering the appropriate treatment to the appropriate patient subgroups.

Before the introduction of bortezomib and IMiDs, a meta-analysis performed on 9 randomized trials confirmed a PFS benefit with upfront ASCT in comparison with conventional chemotherapy combinations.14  Three randomized studies showed that OS was similar whether ASCT was done early or as salvage therapy at relapse.15  Interestingly, in one trial early ASCT improved median EFS (39 vs 13 months), as well as the average time without symptoms (27.8 vs 22.3 months) compared with late ASCT, but OS was unchanged (64.6 vs 64 months). The early approach was also associated with lower rate of relapse, reduced treatment-related toxicities, and discontinuation. Another trial detected no significant PFS improvement with early ASCT (42 vs 33 months; P = .57) and suggested that the greatest benefit from early ASCT was among patients with disease refractory to induction therapy.15 

In elderly patients with newly diagnosed MM, the combination melphalan-prednisone-thalidomide (MPT) was compared with an intermediate-dose melphalan 100 mg/m2 (Mel 100) followed by ASCT.16  The MPT regimen significantly reduced the risk of progression by 46% (P < .001) and the risk of death by 31% (P = .027). This study questioned the role of ASCT in the era of bortezomib and IMiDs. In recent years the efficacy of both ASCT and combination chemotherapy has been improved by the introduction of more effective induction, consolidation, and maintenance schedules that prolong duration of response.

Induction treatments

Bortezomib-dexamethasone (BD) induction regimen significantly increased CR/near CR (nCR) rate after ASCT (39.5%) in comparison with dexamethasone-vincristine-adriamicyn (VAD; 39.5% vs 22.5%; P < .001; Table 1).17  However, the PFS improvement induced by BD was modest (36 vs 29.7 months; P = .064). Better results were obtained with 3-drug combinations.18,20,30,31  The addition of cytotoxic drugs such as doxorubicin or cyclophosphamide to both thalidomide (TAD, CTD)30,31  or bortezomib (PAD, BCD)18,20  improved response rate. The TAD regimen prolonged PFS in comparison with VAD (34 vs 22 months; P < .001), whereas the PFS induced by CTD was similar to cyclophosphamide-VAD (median, 27 vs 25 months; P = .59). The regimen of BCD led to 70% CR/nCR rate after ASCT.20  The combination of PAD significantly improved PFS compared with VAD (35 vs 28 months; P = .002).18  The combination of bortezomib and IMiDs has shown similar results.21,32  Bortezomib-thalidomide-dexamethasone (BTD) was superior to thalidomide-dexamethasone (TD).21  The CR/nCR rate was increased with BTD induction compared with TD (55% vs 41%; P = .002) and so was also the 3-year PFS (68% vs 56%; P < .006).21,32  Grade 3-4 neuropathy was higher with BTD (10% vs 2%; P < .001). The TT program consisted of induction, consolidation with combination chemotherapy plus thalidomide (TT2) or bortezomib (TT3), and double ASCT. In the TT3 program the cumulative frequency of CR increased with time and reached 56% at 2 years, with a 5-year EFS of 69%.10,22 

Table 1

Efficacy of selected regimens

RegimenPatients, nScheduleCR, %PFS/EFS/TTP, %OS, %
Stem cell transplantation      
    BD 121 B: 1.3 mg/m2 days 1, 4, 8, 11; D: 40 mg days 1-4 (cycles 1-4), days 9-12 (cycles 1-2); for four 21-d cycles 50 at 36 mo 81 at 36 mo 
    Mel 20017  Mel 200: 200 mg/m2 (double if < VGPR) 39 ≥ nCR 
    PAD induction 413 P: 1.3 mg/m2 days 1, 4, 8, 11; A: 9 mg/m2 days 1-4; D: 40 mg days 1-4, 9-12, 17-20; for three 28-d cycles 50 at 35 mo 61 at 60 mo 
    Mel 200 Mel 200: 200 mg/m2 (single-double) 21 
    B maintenance18  B: 1.3 mg/m2 days 1, 15; for 2 y 36 
    PAD induction 102 P: 1.3 mg/m2 days 1, 4, 8, 11; A: 30 mg/m2 day 4; D: 40 mg days 1-4, 8-11, 15-18 (cycles 1-2), days 1-4 (cycles 2-4) 12 66 at 36 mo 85 at 36 mo 
    Mel 100 Mel 100: 100 mg/m2 (double) 33 
    LP consolidation L: 25 mg days 1-21; P: 50 mg qod; for four 28-d cycles 40 
    L maintenance19  L: 25 mg days 1-21 until disease progression 40 
    BCD induction 33 B: 1.3 mg/m2 days 1, 4, 8, 11; C: 300 mg/m2 days 1, 8, 15, 22; D: 40 mg days 1-4, 9-12, 17-20; for four 28-d cycles 46 ≥ nCR*   
    Mel 20020  Mel 200: 200 mg/m2 (single) 70 ≥ nCR*   
    BTD induction 236 B: 1.3 mg/m2 days 1, 4, 8, 11; T: 100-200 mg/d; D: 40 mg days 1, 2, 4, 5, 8, 9, 11, 12; for three 21-d cycles 19 68 at 36 mo 90 at 36 mo 
    Mel 200 Mel 200: 200 mg/m2 (double) 42 
    BTD consolidation21  B: 1.3 mg/m2 days 1, 8, 15, 22; T: 100 mg/d; D: 40 mg days 1, 2, 8, 9, 15, 16, 22, 23; for two 35-d cycles 61 
    BTD PACE induction 303 B: 1.0 mg/m2 days 1, 4, 8, 11; T: 200 mg days 1-4; D: 40 mg/m2 days 1-4; P: 10 mg/m2 days 1-4; A: 10 mg/m2 days 1-4; C: 400 mg/m2 days 1-4; E: 40 mg/m2 days 1-4; for two ≤ 56-d cycles  69 at 60 mo 72 at 60 mo 
    Mel 200 Mel 200: 200 mg/m2 (double)  
    BTD PACE consolidation22  BTD as induction; P: 7.5 mg/m2 days 1-4; A: 7.5 mg/m2 days 1-4; C: 300 mg/m2 days 1-4; E: 30 mg/m2 days 1-4; for two 56-d cycles 56 
Drug combinations      
    MPT induction23  1685 M: 0.18 or 0.25 mg/kg days 1-7 or 1-4; P: 2 mg/kg days 1-4; T: 100-200 mg/d; M (0.18 mg) T (100 mg) for six 28-d cycles; M (0.25 mg) T (200 mg) for twelve 42-d cycles 25 ≥ VGPR 43 at 24 mo 50 at 39 mo 
    BMP induction24  344 B: 1.3 mg/m2 days 1, 4, 8, 11, 22, 25, 29, 32 (cycles 1-4), days 1, 8, 22, 29 (cycles 5-9); M: 9 mg/m2 days 1-4; P: 60 mg/m2 days 1-4; for nine 42-d cycles 30 50 at 24 mo 68 at 36 mo 
    BMPT induction 254 B: 1.3 mg/m2 days 1, 8, 15, 22; M: 9 mg/m2 days 1-4; P: 60 mg/m2 days 1-4; T: 50 mg/d; for nine 35-d cycles 38 56 at 36 mo 89 at 36 mo 
    BT maintenance11  B: 1.3 mg/m2 every 14 d; T: 50 mg/d for 2 y 42 
    BLD induction25  35 B: 1.3 mg/m2 days 1, 4, 8, 11; L: 25 mg days 1-14; D: 20 mg days 1, 2, 4, 5, 8, 9, 11, 12 (or days 1, 8, 15); for eight 28-d cycles 37 75 at 18 mo 97 at 18 mo 
    LD induction12  223 L: 25 mg days 1-21; D: 40 mg days 1-4, 9-12, 17-20; for four 28-d cycles 50 at 19 mo 75 at 24 mo 
    Ld induction12  222 L: 25 mg days 1-21; d: 40 mg days 1, 8, 15, 22; for four 28-d cycles 50 at 25 mo 76 at 24 mo 
    MPL induction 152 M: 0.18 mg/kg d 1-4; P: 2 mg/kg d 1-4; L: 10 mg days 1-21; for nine 4-wk cycles 10 50 at 31 mo 70 at 36 mo 
    L maintenance13  L: 10 mg days 1-21 until disease progression 33 ≥ VGPR 
    CLd induction26  53 C: 36 mg/m2 days 1, 2, 8, 9, 15, 16; L: 25 mg days 1-21; d: 40-20 mg weekly (cycles 1-4/5-8) for eight 28-d cycles 79 ≥ nCR*   
Maintenance regimens after stem cell transplantation      
    L maintenance27  307 L: 10-15 mg days 1-21 until disease progression 29 50 at 41 mo 73 at 48 mo 
    L maintenance28  231 L: 10-15 mg days 1-21 until disease progression  50 at 46 mo 88 at 36 mo 
Maintenance regimens after drug combinations      
    BT maintenance29  91 B:1.3 mg/m2 twice weekly days 1, 4, 8, 11, every 3 mo; T: 50 mg/d up to 3 y 44 50 at 32 mo  
    BP maintenance29  87 B: 1.3 mg/m2 twice weekly; days 1, 4, 8, 11, every 3 mo; P: 50 mg qod up to 3 y 39 50 at 24 mo  
RegimenPatients, nScheduleCR, %PFS/EFS/TTP, %OS, %
Stem cell transplantation      
    BD 121 B: 1.3 mg/m2 days 1, 4, 8, 11; D: 40 mg days 1-4 (cycles 1-4), days 9-12 (cycles 1-2); for four 21-d cycles 50 at 36 mo 81 at 36 mo 
    Mel 20017  Mel 200: 200 mg/m2 (double if < VGPR) 39 ≥ nCR 
    PAD induction 413 P: 1.3 mg/m2 days 1, 4, 8, 11; A: 9 mg/m2 days 1-4; D: 40 mg days 1-4, 9-12, 17-20; for three 28-d cycles 50 at 35 mo 61 at 60 mo 
    Mel 200 Mel 200: 200 mg/m2 (single-double) 21 
    B maintenance18  B: 1.3 mg/m2 days 1, 15; for 2 y 36 
    PAD induction 102 P: 1.3 mg/m2 days 1, 4, 8, 11; A: 30 mg/m2 day 4; D: 40 mg days 1-4, 8-11, 15-18 (cycles 1-2), days 1-4 (cycles 2-4) 12 66 at 36 mo 85 at 36 mo 
    Mel 100 Mel 100: 100 mg/m2 (double) 33 
    LP consolidation L: 25 mg days 1-21; P: 50 mg qod; for four 28-d cycles 40 
    L maintenance19  L: 25 mg days 1-21 until disease progression 40 
    BCD induction 33 B: 1.3 mg/m2 days 1, 4, 8, 11; C: 300 mg/m2 days 1, 8, 15, 22; D: 40 mg days 1-4, 9-12, 17-20; for four 28-d cycles 46 ≥ nCR*   
    Mel 20020  Mel 200: 200 mg/m2 (single) 70 ≥ nCR*   
    BTD induction 236 B: 1.3 mg/m2 days 1, 4, 8, 11; T: 100-200 mg/d; D: 40 mg days 1, 2, 4, 5, 8, 9, 11, 12; for three 21-d cycles 19 68 at 36 mo 90 at 36 mo 
    Mel 200 Mel 200: 200 mg/m2 (double) 42 
    BTD consolidation21  B: 1.3 mg/m2 days 1, 8, 15, 22; T: 100 mg/d; D: 40 mg days 1, 2, 8, 9, 15, 16, 22, 23; for two 35-d cycles 61 
    BTD PACE induction 303 B: 1.0 mg/m2 days 1, 4, 8, 11; T: 200 mg days 1-4; D: 40 mg/m2 days 1-4; P: 10 mg/m2 days 1-4; A: 10 mg/m2 days 1-4; C: 400 mg/m2 days 1-4; E: 40 mg/m2 days 1-4; for two ≤ 56-d cycles  69 at 60 mo 72 at 60 mo 
    Mel 200 Mel 200: 200 mg/m2 (double)  
    BTD PACE consolidation22  BTD as induction; P: 7.5 mg/m2 days 1-4; A: 7.5 mg/m2 days 1-4; C: 300 mg/m2 days 1-4; E: 30 mg/m2 days 1-4; for two 56-d cycles 56 
Drug combinations      
    MPT induction23  1685 M: 0.18 or 0.25 mg/kg days 1-7 or 1-4; P: 2 mg/kg days 1-4; T: 100-200 mg/d; M (0.18 mg) T (100 mg) for six 28-d cycles; M (0.25 mg) T (200 mg) for twelve 42-d cycles 25 ≥ VGPR 43 at 24 mo 50 at 39 mo 
    BMP induction24  344 B: 1.3 mg/m2 days 1, 4, 8, 11, 22, 25, 29, 32 (cycles 1-4), days 1, 8, 22, 29 (cycles 5-9); M: 9 mg/m2 days 1-4; P: 60 mg/m2 days 1-4; for nine 42-d cycles 30 50 at 24 mo 68 at 36 mo 
    BMPT induction 254 B: 1.3 mg/m2 days 1, 8, 15, 22; M: 9 mg/m2 days 1-4; P: 60 mg/m2 days 1-4; T: 50 mg/d; for nine 35-d cycles 38 56 at 36 mo 89 at 36 mo 
    BT maintenance11  B: 1.3 mg/m2 every 14 d; T: 50 mg/d for 2 y 42 
    BLD induction25  35 B: 1.3 mg/m2 days 1, 4, 8, 11; L: 25 mg days 1-14; D: 20 mg days 1, 2, 4, 5, 8, 9, 11, 12 (or days 1, 8, 15); for eight 28-d cycles 37 75 at 18 mo 97 at 18 mo 
    LD induction12  223 L: 25 mg days 1-21; D: 40 mg days 1-4, 9-12, 17-20; for four 28-d cycles 50 at 19 mo 75 at 24 mo 
    Ld induction12  222 L: 25 mg days 1-21; d: 40 mg days 1, 8, 15, 22; for four 28-d cycles 50 at 25 mo 76 at 24 mo 
    MPL induction 152 M: 0.18 mg/kg d 1-4; P: 2 mg/kg d 1-4; L: 10 mg days 1-21; for nine 4-wk cycles 10 50 at 31 mo 70 at 36 mo 
    L maintenance13  L: 10 mg days 1-21 until disease progression 33 ≥ VGPR 
    CLd induction26  53 C: 36 mg/m2 days 1, 2, 8, 9, 15, 16; L: 25 mg days 1-21; d: 40-20 mg weekly (cycles 1-4/5-8) for eight 28-d cycles 79 ≥ nCR*   
Maintenance regimens after stem cell transplantation      
    L maintenance27  307 L: 10-15 mg days 1-21 until disease progression 29 50 at 41 mo 73 at 48 mo 
    L maintenance28  231 L: 10-15 mg days 1-21 until disease progression  50 at 46 mo 88 at 36 mo 
Maintenance regimens after drug combinations      
    BT maintenance29  91 B:1.3 mg/m2 twice weekly days 1, 4, 8, 11, every 3 mo; T: 50 mg/d up to 3 y 44 50 at 32 mo  
    BP maintenance29  87 B: 1.3 mg/m2 twice weekly; days 1, 4, 8, 11, every 3 mo; P: 50 mg qod up to 3 y 39 50 at 24 mo  

TTP indicates time to progression; VGPR, very good partial response or better; PAD, bortezomib-adriamicyn-dexamethasone (in this schedule, P stands for bortezomib); BCD, bortezomib-cyclophosphamide-dexamethasone; BTD, bortezomib-thalidomide-dexamethasone; BLD, bortezomib-lenalidomide-dexamethasone; BTD-PACE, bortezomib-thalidomide-dexamethasone and cisplatin-doxorubicin-cyclophosphamide-etoposide; LP, lenalidomide-prednisone; qod, every other day; L, lenalidomide; B, bortezomib; BMP, bortezomib-melphalan-prednisone; BMPT, bortezomib-melphalan-prednisone-thalidomide; BT, bortezomib-thalidomide; BP, bortezomib-prednisone; CLd, carfilzomib, lenolidomide, and low-dose dexamethasone; Mel, melphalan; CR, complete response; PFS, progression-free survival; EFS, event-free survival; OS, overall survival; BD, bortezomib-dexamethasone; LD, lenalidomide plus high-dose dexamethasone; Ld, lenalidomide plus low-dose dexamethasone; MPT, melphalan-prednisone-thalidomide; and MPL, melphalan-prednisone-lenalidomide.

*

In per-protocol population.

Percentage at 2 years.

Alternative BMP schedule11  was B 1.3 mg/m2 days 1, 8, 15, 22; M 9 mg/m2 days 1-4; P 50 mg/m2 days 1-4.

Consolidation treatments

In several studies, the double ASCT extended PFS in comparison with single ASCT, but OS benefit was found in only 2 of them.33  The role of the double ASCT is being questioned by the availability of novel effective combinations, although it is currently recommended in patients who fail to achieve VGPR or better after the first ASCT. In a recent study, tandem ASCT improved OS in comparison with single ASCT (5-year OS, 70% vs 55%; P = .03), the treatment regimen included PAD as induction and bortezomib or thalidomide as maintenance; unfortunately, the tandem versus single ASCT option was not randomized and only depended on insurance reimbursement.18  Consolidation with bortezomib and IMiDs is currently under evaluation. BTD consolidation increased CR from 15% to 49% in patients who achieved VGPR after double ASCT.8  Recently, the role of BTD versus TD consolidation after double ASCT was assessed. BTD consolidation increased the CR/nCR rate from 63% to 73%, but 3-year PFS was marginally improved (60% vs 48%; P = .042).32  Four cycles of lenalidomide-prednisone have been adopted as consolidation after double ASCT; this approach increased the CR rate from 38% to 66%.

Maintenance treatments

After ASCT, thalidomide maintenance therapy improved the quality of response and increased PFS. In a recent meta-analysis, maintenance therapy with thalidomide reduced both the risk of progression (36%) and death (27%).34  However a significant rate of grade 3-4 polineuropathy (7%-19%) was reported, and the rate of discontinuation reached 52.2%.35  Median time on therapy was ∼ 1 year.36  The role of lenalidomide maintenance after ASCT was assessed.27,28  In one study, lenalidomide reduced the risk of progression by 50% (P < .001), whereas OS was similar to the placebo group (P = .29).27  In another study, lenalidomide reduced the risk of progressive disease by 52% (P < .001) and the risk of death by 38% (P < .03; Figure 1A-B).28  In both studies, the most frequent grade 3-4 adverse events were neutropenia (45%-51%) and infections (1%-13%), and a higher incidence of second primary cancers in the lenalidomide arm was detected (7.8%-8.5% at 3 years; Table 2). Despite the high toxicities associated with this approach, a PFS improvement was detected. Median time on treatment was ∼ 2 years. Data on single-agent bortezomib maintenance are available in one trial only. Patients who were randomly assigned to PAD or VAD induction followed by ASCT received bortezomib or thalidomide as maintenance therapy, respectively. Bortezomib maintenance significantly improved CR/nCR, from 31% to 49%, and reduced the risk of progression on a landmark analysis (P = .04). With bortezomib maintenance, grade 3-4 peripheral neuropathy was 5% and grade 3-4 infections were 24%. The median time on therapy was ∼ 2 years in the bortezomib group and 1 year in the thalidomide group.18 

Figure 1

Hazard ratios. (A) PFS with regimens, including stem cell transplantation and novel agents; (B) OS with regimens, including stem cell transplantation and novel agents; (C) PFS with drug combinations, including novel agents; and (D) OS with drug combinations, including novel agents. Len indicates lenalidomide; NA, not available; and MP, melphalan-prednisone.

Figure 1

Hazard ratios. (A) PFS with regimens, including stem cell transplantation and novel agents; (B) OS with regimens, including stem cell transplantation and novel agents; (C) PFS with drug combinations, including novel agents; and (D) OS with drug combinations, including novel agents. Len indicates lenalidomide; NA, not available; and MP, melphalan-prednisone.

Close modal
Table 2

Safety of selected regimens

RegimenGrade 3-4 hematologic toxicities
Grade 3-4 nonhematologic toxicities
Neutropenia, %Thrombocytopenia, %Anemia, %Thromboembolism, %Neuropathy, %Infection, %Second primary cancer, %
Stem cell transplantation        
    BD17  2.9 4.2 1.7 7.1 8.8  
    PAD induction 10 24 26  
    B maintenance18  24  
    PAD induction 10 17 16 17  
    LP consolidation        
    L maintenance19  16  
    BCD induction20  13 25 12   
    BTD induction    10  
    BTD consolidation21     0.6 0.6 1.2  
    BTD PACE induction    11 14   
    BTD PACE consolidation22     10   
Drug combinations        
    MPT induction 17-43 (any hematologic toxicity) 2-6 1-6 4-20  
    BMP induction24  40 37 19 13 10 6* 
    BMPT induction 38 22 10 13  
    BT maintenance11  2.6 0.6 5.3 1.3  
    BLD induction25  4.5 7.5  
    LD induction12  12 26 16  
    Ld induction12  20 12  
    MPL induction 35 11 3 11 7 
    L maintenance13  7 6 4 
    CLd induction26  12 10 18 10  
Maintenance regimens after stem cell transplantation        
    L maintenance27  51 14 8.5§ 
    L maintenance28  45 14    7.8§ 
Maintenance regimens after drug combinations        
    BT maintenance29   
    BP maintenance29   
RegimenGrade 3-4 hematologic toxicities
Grade 3-4 nonhematologic toxicities
Neutropenia, %Thrombocytopenia, %Anemia, %Thromboembolism, %Neuropathy, %Infection, %Second primary cancer, %
Stem cell transplantation        
    BD17  2.9 4.2 1.7 7.1 8.8  
    PAD induction 10 24 26  
    B maintenance18  24  
    PAD induction 10 17 16 17  
    LP consolidation        
    L maintenance19  16  
    BCD induction20  13 25 12   
    BTD induction    10  
    BTD consolidation21     0.6 0.6 1.2  
    BTD PACE induction    11 14   
    BTD PACE consolidation22     10   
Drug combinations        
    MPT induction 17-43 (any hematologic toxicity) 2-6 1-6 4-20  
    BMP induction24  40 37 19 13 10 6* 
    BMPT induction 38 22 10 13  
    BT maintenance11  2.6 0.6 5.3 1.3  
    BLD induction25  4.5 7.5  
    LD induction12  12 26 16  
    Ld induction12  20 12  
    MPL induction 35 11 3 11 7 
    L maintenance13  7 6 4 
    CLd induction26  12 10 18 10  
Maintenance regimens after stem cell transplantation        
    L maintenance27  51 14 8.5§ 
    L maintenance28  45 14    7.8§ 
Maintenance regimens after drug combinations        
    BT maintenance29   
    BP maintenance29   

Abbreviations are explained in Table 1.

*

Incidence in the BMP arm at 5 years.

Grade 4 only.

Incidence in the whole MPL-L arm at 3 years.

§

Incidence in the lenalidomide arm at 3 years.

The 3-drug combinations, such as BCD, BTD, or BRD inductions, improved response rate. These results might be further improved with the introduction of novel proteasome inhibitors as shown by the combination of carfilzomib-lenalidomide-dexamethasone. The role of the second ASCT has been questioned and is currently limited to patients who do not reach VGPR after a single ASCT. Future trials to assess the role of single versus double ASCT are warranted. Consolidation with BTD improved CR rates and PFS, but data are limited and randomized studies for consolidation regimens are lacking. Maintenance therapy with IMiDs reduced the risk of progression by ∼ 50%. In patients aged 65-75 years, PAD induction followed by tandem Mel 100, ASCT, lenalidomide-prednisone consolidation, and lenalidomide maintenance induced a CR rate of 40%, 3-year PFS of 66%, and 3-year OS of 85%.19  Overall, the available data show that effective induction, consolidation, and maintenance therapy have greatly improved the efficacy of high-dose therapy and ASCT. With these approaches the CR/nCR rates are ranging from 36% to 70%, the 5-year PFS from 62% to 69%, and the 5-year OS from 61% to 72%.

A meta-analysis of the 6 randomized controlled trials that compared MPT with MP showed that MPT improved both PFS and OS of ∼ 6 months.23  Peripheral neuropathy (6%-23%) and venous thromboembolism (3%-12%) were the main adverse events associated with thalidomide. The combination bortezomib-melphalan-prednisone (BMP) improved the median time to next therapy by 8 months (P < .001) and reduced the risk of death by 32% in comparison with MP.24  Grade 3-4 peripheral neuropathy was 13%.

BMP plus thalidomide followed by bortezomib-thalidomide maintenance (BMPT-BT) was compared with BMP alone. BMPT-BT significantly increased the CR rate (38% vs 24%; P < .001) and prolonged the 3-year PFS (56% vs 41%; P = .008), but the 3-year OS was similar (89% vs 87%; P = .77).11  The median PFS for BMPT-BT was 37.2 months. Thalidomide did not increase the hematologic toxicity of BMP, and the weekly dose of bortezomib plus 50 mg/d thalidomide did not increase the risk of grade 3-4 peripheral neuropathy (4%). In another study BTP versus BMP inductions were followed by BT or bortezomib-prednisone (BP) maintenance. From start of maintenance, the median PFS was 32 months for patients receiving BT and 24 months for those receiving BP (P = .1).29 

In a randomized phase 2 study, bortezomib-lenalidomide-dexamethasone (BLD), bortezomib-cyclophosphamide-dexamethasone (BCD) or bortezomib-cyclophosphamide-lenalidomide-dexamethasone (BCLD) was tested. The regimens were equally effective; CR rate was 24% with BLD, 22% with BCD, and 25% with BCLD, and the corresponding 1-year PFS was 83%, 93%, and 86%, respectively. BCLD increased the frequency of hematologic toxicities and the discontinuation rate (21%); the excessive toxicities did probably reduce the efficacy of the 4-drug combination.37  In another study, BLD induced 100% at least PR, including 37% of CRs; the 1.5-year PFS rate was 75% and the 1.5-year OS rate was 97%; the frequency of grade 3-4 neuropathy was 3%.25 

MPL-L prolonged the median PFS of 17 months in comparison with MPL and MP alone (31 vs 14 vs 13 months; P < .001).13  The 3-year OS was similar (70% vs 62% vs 66%). The most common adverse events were hematologic, with grade 4 neutropenia reported in 35% and 32% patients in the MPL-L and MPL groups, respectively. The 3-year rate of second primary cancers was 7% with MPL-L.

The combination of the new proteasome inhibitor carfilzomib with lenalidomide and low-dose dexamethasone was effective and well tolerated. Responses were rapid, with 46 of 49 patients achieving PR after 1 cycle; after 4 cycles, the PR rate was 100%; after 12 cycles, the VGPR rate was 100%, including 79% CR/nCR. No overlapping toxicities were recorded, with no grade 3-4 peripheral neuropathy.26 

In conclusion, MPT or BMP has improved median PFS of ∼ 6-8 months in comparison with MP. The latest 3- or 4-drug combinations, and particularly maintenance therapy, have further prolonged PFS approximately by another 10-17 months. OS advantage will require longer follow-up to be clearly assessed. Drug combinations, including proteasome inhibitors and IMiDs, have led to at least PR of 100% and high rates of CRs (24%-38%). Maintenance therapy with bortezomib or lenalidomide induced a median PFS of ∼ 31-37 months and 3-year OS of ∼ 70%-89% (Figure 1C-D). Despite these improvements and the difficult comparison of studies conducted in patients younger than 65 years versus studies conducted in patients older than 65 years, ASCT still seems to add clinical value to current induction, consolidation, and maintenance regimens.

In a retrospective analysis, OS was evaluated in patients receiving lenalidomide-dexamethasone followed by ASCT or lenalidomide-dexamethasone as continuous treatment, the 3-year OS was 94% in patients receiving early ASCT and 78% in those receiving lenalidomide-dexamethasone. Although this trial was not designed to assess early ASCT versus drug combination, OS with early ASCT appeared to be superior.38  In a posthoc landmark analysis of 53 patients who received BLD induction at diagnosis, PFS was unchanged in patients treated with early ASCT or continuous treatment (P = .38).25  In a retrospective analysis of 290 patients who received early versus delayed ASCT after IMiD-based initial therapy, the 4-year OS rate was 73% in both groups (P = .03).39 

In the first prospective randomized trial, patients received Ld induction therapy and were then randomly assigned to tandem ASCT or MPL. After a median follow-up of 26 months, the CR rate was 25% for ASCT and 20% for MPL (P = .49). ASCT reduced the risk of progression by ∼ 50% (P < .001). The 2-year PFS was 73% for ASCT and 54% for MPL. The 2-year OS was comparable in the 2 groups (90% vs 87%; P = .19).40  In a similar study, 390 patients received Ld induction regimen and were then randomly assigned to receive tandem ASCT or cyclophosphamide-lenalidomide-dexamethasone, followed by lenalidomide maintenance therapy. Patient accrual was completed in May 2011, and preliminary data should be available soon (NCT01091831 at www.clinicaltrials.gov).

Two ongoing large studies are currently enrolling patients. The IFM/DFCI2009 study addresses the question of whether high-dose therapy and ASCT or conventional chemotherapy should be preferred. In this trial 1000 patients are receiving BLD induction and are then randomly assigned to continue BLD treatment or to receive ASCT plus BLD consolidation. Afterward, the role of fixed-duration maintenance therapy will be investigated, and patients will receive lenalidomide maintenance for 1 year (NCT01191060 and NCT01208662 at www.clinicaltrials.gov). The EMN02 study not only aims to compare ASCT with conventional chemotherapy, but it also investigates the role of second ASCT and the advantages associated with consolidation therapy after transplantation. In this study, 1500 patients are receiving BCD induction and are then randomly assigned to BMP or ASCT and further randomly assigned to receive consolidation or no consolidation with BLD. The benefits of maintenance therapy will be assessed as well, but, compared with the previous study, patients will receive lenalidomide maintenance until disease progression (NCT01208766 at www.clinicaltrials.gov).

The introduction of effective regimens, including bortezomib and IMiDs, has greatly improved the outcome observed with ASCT in young patients with MM. The achievement of CR is an independent predictor of prolonged PFS and OS. Despite the improvement reported with conventional chemotherapy plus IMiDs, ASCT remains a necessary component of therapy to attain CR and consequently superior PFS. The data from prospective trials performed so far suggest that the best available strategy to achieve high CR rates and to prolong their duration includes induction with 3-drug bortezomib-based combinations followed by ASCT and consolidation/maintenance with IMiDs. This sequential approach seems the most appropriate strategy to upgrade response and prolong survival. Longer follow-up is needed to evaluate whether the timing of ASCT is relevant for OS. Results from ongoing large collaborative studies that compare effective drug combinations with stem cell transplantation are awaited and will shed further light on this important clinical question.

This article was selected by the Blood and Hematology 2012 American Society of Hematology Education Program editors for concurrent submission to Blood and Hematology 2012. It is reprinted in Hematology Am Soc Hematol Educ Program. 2012;2012:335-341.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the editorial assistant Giorgio Schirripa.

Contribution: A.P. and F.C. collected and analyzed the data and wrote the manuscript.

Conflict-of-interest disclosure: A.P. has received honoraria from Celgene, Janssen-Cilag, Bristol-Myers Squibb, Millennium, Merck, and Onyx and has served on the advisory boards of Celgene and Janssen-Cilag. F.C. has received honoraria from Celgene, Janssen-Cilag, and Onyx and has served on the advisory committee of Celgene.

Correspondence: Antonio Palumbo, Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Via Genova 3, 10126 Torino, Italy; e-mail: appalumbo@yahoo.com.

1
Altekruse
 
SF
Kosary
 
C
Krapcho
 
M
Neyman
 
N
Aminou
 
R
Waldron
 
W
SEER Cancer Statistics Review, 1975-2007
2008
Accessed April 14, 2012
Bethesda, MD
National Cancer Institute
 
2
Brenner
 
H
Gondos
 
A
Pulte
 
D
Recent major improvement in long-term survival of younger patients with multiple myeloma.
Blood
2008
, vol. 
111
 
5
(pg. 
2521
-
2526
)
3
Keats
 
JJ
Chesi
 
M
Egan
 
JB
, et al. 
Clonal competition with alternating dominance in multiple myeloma.
Blood
2012
, vol. 
120
 
5
(pg. 
1067
-
1076
)
4
van de Velde
 
HJ
Liu
 
X
Chen
 
G
Cakana
 
A
Deraedt
 
W
Bayssas
 
M
Complete response correlates with long-term survival and progression-free survival in high-dose therapy in multiple myeloma.
Haematologica
2007
, vol. 
92
 
10
(pg. 
1399
-
1406
)
5
Gay
 
F
Larocca
 
A
Wijermans
 
P
, et al. 
Complete response correlates with long-term progression-free and overall survival in elderly myeloma treated with novel agents: analysis of 1175 patients.
Blood
2011
, vol. 
117
 
11
(pg. 
3025
-
3031
)
6
Martinez-Lopez
 
J
Blade
 
J
Mateos
 
MV
, et al. 
Long-term prognostic significance of response in multiple myeloma after stem cell transplantation.
Blood
2011
, vol. 
118
 
3
(pg. 
529
-
534
)
7
Paiva
 
B
Martinez-Lopez
 
J
Vidriales
 
MB
, et al. 
Comparison of immunofixation, serum free light chain, and immunophenotyping for response evaluation and prognostication in multiple myeloma.
J Clin Oncol
2011
, vol. 
29
 
12
(pg. 
1627
-
1633
)
8
Ladetto
 
M
Pagliano
 
G
Ferrero
 
S
, et al. 
Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma.
J Clin Oncol
2010
, vol. 
28
 
12
(pg. 
2077
-
2084
)
9
Hoering
 
A
Crowley
 
J
Shaughnessy
 
JD
, et al. 
Complete remission in multiple myeloma examined as time-dependent variable in terms of both onset and duration in total therapy protocols.
Blood
2009
, vol. 
114
 
7
(pg. 
1299
-
1305
)
10
van Rhee
 
F
Szymonifka
 
J
Anaissie
 
E
, et al. 
Total Therapy 3 for multiple myeloma: prognostic implications of cumulative dosing and premature discontinuation of VTD maintenance components, bortezomib, thalidomide, and dexamethasone, relevant to all phases of therapy.
Blood
2010
, vol. 
116
 
8
(pg. 
1220
-
1227
)
11
Palumbo
 
A
Bringhen
 
S
Rossi
 
D
, et al. 
Bortezomib-melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma: a randomized controlled trial.
J Clin Oncol
2010
, vol. 
28
 
34
(pg. 
5101
-
5109
)
12
Rajkumar
 
SV
Jacobus
 
S
Callander
 
NS
, et al. 
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
, vol. 
11
 
1
(pg. 
29
-
37
)
13
Palumbo
 
A
Hajek
 
R
Delforge
 
M
, et al. 
Continuous lenalidomide treatment for newly diagnosed multiple myeloma.
N Engl J Med
2012
, vol. 
366
 
19
(pg. 
1759
-
1769
)
14
Koreth
 
J
Cutler
 
CS
Djulbegovic
 
B
, et al. 
High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: a systematic review and meta-analysis of randomized controlled trials.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
2
(pg. 
183
-
196
)
15
Giralt
 
S
Stem cell transplantation for multiple myeloma: current and future status.
Hematology Am Soc Hematol Educ Program
2011
, vol. 
2011
 (pg. 
191
-
196
)
16
Facon
 
T
Mary
 
JY
Hulin
 
C
, et al. 
Melphalan and prednisone plus thalidomide versus melphalan and prednisone alone or reduced-intensity autologous stem cell transplantation in elderly patients with multiple myeloma (IFM 99-06): a randomised trial.
Lancet
2007
, vol. 
370
 
9594
(pg. 
1209
-
1218
)
17
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 stemcell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial.
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4621
-
4629
)
18
Sonneveld
 
P
Schmidt-Wolf
 
IGH
van der Holt
 
B
, et al. 
HOVON-65/GMMG-HD4 randomized phase III trial comparing bortezomib, doxorubicin, dexamethasone (PAD) vs VAD followed by high-dose melphalan (HDM) and maintenance with bortezomib or thalidomide in patients with newly diagnosed multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
 
Abstract 40
19
Palumbo
 
A
Gay
 
F
Falco
 
P
, et al. 
Bortezomib as induction before autologous transplantation, followed by lenalidomide as consolidation-maintenance in untreated multiple myeloma patients.
J Clin Oncol
2010
, vol. 
28
 
5
(pg. 
800
-
807
)
20
Reeder
 
CB
Reece
 
DE
Kukreti
 
V
, et al. 
Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial.
Leukemia
2009
, vol. 
23
 
7
(pg. 
1337
-
1341
)
21
Cavo
 
M
Tacchetti
 
P
Patriarca
 
F
, et al. 
Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study.
Lancet
2010
, vol. 
376
 
9758
(pg. 
2075
-
2085
)
22
Barlogie
 
B
Anaissie
 
E
van Rhee
 
F
, et al. 
Incorporating bortezomib into upfront treatment for multiple myeloma: early results of total therapy 3.
Br J Haematol
2007
, vol. 
138
 
2
(pg. 
176
-
185
)
23
Fayers
 
PM
Palumbo
 
A
Hulin
 
C
, et al. 
Thalidomide for previously untreated elderly patients with multiple myeloma: meta-analysis of 1685 individual patient data from 6 randomized clinical trials.
Blood
2011
, vol. 
118
 
5
(pg. 
1239
-
1247
)
24
Mateos
 
MV
Richardson
 
PG
Schlag
 
R
, et al. 
Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow-up and impact of subsequent therapy in the phase III VISTA trial.
J Clin Oncol
2010
, vol. 
28
 
13
(pg. 
2259
-
2266
)
25
Richardson
 
PG
Weller
 
E
Lonial
 
S
, et al. 
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma.
Blood
2010
, vol. 
116
 
5
(pg. 
679
-
686
)
26
Jakubowiak
 
AJ
Dytfeld
 
D
Jagannath
 
S
, et al. 
Final results of a frontline phase 1/2 study of carfilzomib, lenalidomide, and low-dose dexamethasone (CRd) in multiple myeloma (MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
 
Abstract 631
27
Attal
 
M
Lauwers-Cances
 
V
Marit
 
G
, et al. 
Lenalidomide maintenance after stem-cell transplantation for multiple myeloma.
N Engl J Med
2012
, vol. 
366
 
19
(pg. 
1782
-
1791
)
28
McCarthy
 
PL
Owzar
 
K
Hofmeister
 
CC
, et al. 
Lenalidomide after stem-cell transplantation for multiple myeloma.
N Engl J Med
2012
, vol. 
366
 
19
(pg. 
1770
-
1781
)
29
Mateos
 
MV
Oriol
 
A
Martínez-López
 
J
, et al. 
Bortezomib, melphalan, and prednisone versus 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
, vol. 
11
 
10
(pg. 
934
-
941
)
30
Lokhorst
 
HM
Schmidt-Wolf
 
I
Sonneveld
 
P
, et al. 
Thalidomide in induction treatment increases the very good partial response rate before and after high-dose therapy in previously untreated multiple myeloma.
Haematologica
2008
, vol. 
93
 
1
(pg. 
124
-
127
)
31
Morgan
 
GJ
Davies
 
FE
Gregory
 
WM
, et al. 
Cyclophosphamide, thalidomide, and dexamethasone (CTD) as initial therapy for patients with multiple myeloma unsuitable for autologous transplantation.
Blood
2011
, vol. 
118
 
5
(pg. 
1231
-
1238
)
32
Cavo
 
M
Pantani
 
L
Petrucci
 
MT
, et al. 
Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy following autologous hematopoietic stem-cell transplantation in patients with newly diagnosed multiple myeloma.
Blood
2012
, vol. 
120
 
1
(pg. 
9
-
19
)
33
Barlogie
 
B
Attal
 
M
Crowley
 
J
, et al. 
Long-term follow-up of autotransplantation trials for multiple myeloma: update of protocols conducted by the Intergroupe Francophone du Myelome, Southwest Oncology Group, and University of Arkansas for Medical Sciences.
J Clin Oncol
2010
, vol. 
28
 
7
(pg. 
1209
-
1214
)
34
Hahn-Ast
 
C
von Lilienfeld-Toal
 
M
van Heteren
 
P
, et al. 
Improved progression-free survival and overall survival with thalidomide maintenance therapy in multiple myeloma: a meta-analyis of randomized trials in 2274 patients [abstract].
Haematologica
2010
, vol. 
95
 
2
 
Abstract 0942
35
Morgan
 
GJ
Gregory
 
WM
Davies
 
FE
, et al. 
The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis.
Blood
2012
, vol. 
119
 
1
(pg. 
7
-
15
)
36
Attal
 
M
Harousseau
 
JL
Leyvraz
 
S
, et al. 
Maintenance therapy with thalidomide improves survival in patients with multiple myeloma.
Blood
2006
, vol. 
108
 
10
(pg. 
3289
-
3894
)
37
Kumar
 
S
Flinn
 
I
Richardson
 
PG
, et al. 
Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma.
Blood
2012
, vol. 
119
 
19
(pg. 
4375
-
4382
)
38
Siegel
 
DS
Jacobus
 
S
Rajkumar
 
SV
, et al. 
Outcome with lenalidomide plus dexamethasone followed by early autologous stem cell transplantation in the ECOG E4A03 randomized clinical trial [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
 
Abstract 38
39
Kumar
 
SK
Lacy
 
MQ
Dispenzieri
 
A
, et al. 
Early versus delayed autologous transplantation after immunomodulatory agents-based induction therapy in patients with newly diagnosed multiple myeloma.
Cancer
2012
, vol. 
118
 
6
(pg. 
1585
-
1592
)
40
Palumbo
 
A
Cavallo
 
F
Hardan
 
I
, et al. 
A phase III study to compare melphalan, prednisone, lenalidomide (MPR) versus melphalan 200 mg/m2 and autologous transplantation (MEL200) in newly diagnosed multiple myeloma patients [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
 
Abstract 3573
Sign in via your Institution