The role of high-dose therapy followed by autologous stem cell transplantation (ASCT) in the treatment of multiple myeloma (MM) continues to evolve in the novel agent era. The choice of induction therapy has moved from conventional chemotherapy to newer regimens incorporating the immunomodulatory derivatives thalidomide or lenalidomide and the proteasome inhibitor bortezomib. These drugs combine well with traditional therapies and with one another to form various doublet, triplet, and quadruplet regimens. Up-front use of these induction treatments, in particular 3-drug combinations, has affected unprecedented rates of complete response that rival those previously seen with conventional chemotherapy and subsequent ASCT. Autotransplantation applied after novel-agent-based induction regimens provides further improvement in the depth of response, a gain that translates into extended progression-free survival and, potentially, overall survival. High activity shown by immunomodulatory derivatives and bortezomib before ASCT has recently led to their use as consolidation and maintenance therapies after autotransplantation. Novel agents and ASCT are complementary treatment strategies for MM. This article reviews the current literature and provides important perspectives and guidance on the major issues surrounding the optimal current management of younger, transplantation-eligible MM patients.

Multiple myeloma (MM) is a disease of the elderly. Overall, only 35% of the patients are younger than 65 years at the time of diagnosis, whereas the remaining two-thirds are older.1  Age is an independent prognostic factor in MM2  and, importantly, provides a major criterion by which patients can be considered eligible to tolerate high-dose therapy (HDT) with autologous hematopoietic stem cell transplantation (ASCT). Over the last decade, the survival of patients with newly diagnosed MM, particularly those younger than 60 years, has significantly improved.3  The widespread use of ASCT and the introduction into clinical practice of the novel agents bortezomib and the immunomodulatory derivatives (IMiDs) thalidomide and lenalidomide have significantly contributed to major advances in MM therapy and prognosis.4,5 

Thalidomide or bortezomib combined with melphalan and prednisone represent new standards of care for elderly, transplantation-ineligible MM patients.6-8  In this setting, lenalidomide in combination with low-dose dexamethasone is an alternative treatment option.9  In younger patients, the novel agents have been incorporated into the therapeutic algorithm along with ASCT to improve clinical outcomes.10-12  In particular, these drugs have been used as part of induction therapy before ASCT and as consolidation/maintenance after autotransplantation. This manuscript from the International Myeloma Working Group (IMWG) presents an overview of the most recent studies of novel agents combined with ASCT and focuses on the main areas of current debate, including the choice of induction regimen, the role of post-ASCT consolidation and maintenance therapies, the impact on prognosis of ASCT incorporating the new drugs, and the management and prevention of major toxicities related to the use of novel therapies.

There is no formal definition of a younger patient with MM, although this term is commonly used to identify a person for whom ASCT is planned as part of the treatment program. As many phase 3 studies of ASCT have enrolled patients with an upper age limit not exceeding 65 years,13-19  younger MM patients are often operatively defined as being 65 years of age and younger. However, this arbitrary cut-off does not exclude patients who are older than 65 years from ASCT. In particular, in selected patients up to the age of 70 to 75 years who are medically fit, ASCT is a treatment option that can be performed safely at most specialized transplantation or myeloma centers.20  Unlike in younger patients, benefits from ASCT have not been consistently demonstrated in the elderly.

When symptomatic, or active, MM is diagnosed based on the presence of organ damage related to the underlying malignant clone (eg, hypercalcemia, renal insufficiency, anemia, and bone disease),21  therapy is required immediately. By contrast, patients with asymptomatic or smoldering MM are closely observed without specific therapy until the disease progresses to a symptomatic phase. Clinical trials are currently underway to investigate whether novel agents can delay the risk of progression from smoldering to active MM and improve overall survival (OS).22  At present, the IMWG does not recommend treatment for smoldering MM but considers patients at high risk of progression to symptomatic disease as candidates for investigative clinical trials.

Over the last decade, ASCT has been considered the standard of care for younger patients with newly diagnosed MM23-25  based on the increased rate of complete response (CR) and prolonged OS compared with conventional chemotherapy in several randomized studies.13,14  However, not all the studies published so far have uniformly demonstrated the superiority of ASCT over chemotherapy at standard doses.16-18  A number of factors may account for these discrepancies, including treatment crossover for patients randomized to conventional treatment, possible bias in patient selection criteria, and differences between studies with respect to the intensity and duration of conventional therapy. A systematic review and meta-analysis of randomized studies has shown a significant benefit with single ASCT in terms of prolonged progression-free survival (PFS), but not of OS.26  However, these results should be cautiously interpreted because of methodologic limitations of the analysis and significant heterogeneity across different studies. An alternative to autotransplantation up-front is to delay HDT with ASCT at the time of relapse. Although in a pilot study the length of OS for patients receiving early or late ASCT after conventional induction chemotherapy was equivalent, early autotransplantation was associated with a longer event-free survival (EFS) and better quality of life.27  In the novel agent era, the issue of early versus late ASCT needs to be reevaluated in the context of large randomized clinical trials. Two of these studies are currently ongoing, one of them headed by the European Myeloma Network and the other performed by a consortium of centers in France and the United States. While final results of these studies are awaited, the IMWG recommends that ASCT should be offered at some point in the course of the treatment program for a patient eligible to receive HDT. Although favorable results with ASCT up-front are backed by phase 3 studies, increasing numbers of patients and physicians, particularly in United States, are currently opting to collect stem cells early and deferring transplantation at the time of relapse.

Five randomized trials directly addressed the question of single versus double, or tandem, ASCT as up-front therapy for MM.15,19,28-30  Results were conflicting because of differences between studies with respect to their structural and methodologic characteristics. In particular, although extended EFS with double ASCT was observed in most of the trials, an OS benefit was demonstrated in only 2 of them. A meta-analysis of data pooled from controlled clinical trials (one of which has been recently retracted) failed to show superior OS with double ASCT which, by the opposite, was associated with improved response rates and EFS.31  A number of concerns related to the methodology of the analysis and errors involving data extractions have been raised, suggesting that these caveats might have negatively influenced the conclusion.32,33  More recently, a report of long-term outcomes of several trials of autotransplantation(s) confirmed superior results offered by double ASCT compared with a single transplantation.34 

In 2 studies of double ASCT, post-hoc subgroup analyses showed that the second autotransplantation improved clinical outcomes in those patients who failed high-quality responses after the first ASCT.15,19  However, a major limitation of these studies was their lack of power to demonstrate the equivalence of 1 versus 2 transplants for patients with high-quality responses after the first course of HDT. With the recent availability of highly effective novel agents, the role of single versus double ASCT is being explored in the context of prospective, randomized clinical trials, such as that currently headed by the Bone Marrow Transplant Clinical Trials Network. In the meantime, the IMWG suggests considering timely second ASCT in those patients who fail to achieve a very good partial response (VGPR) or better after the first ASCT.

Attainment of CR after both induction therapy and ASCT is one of the strongest predictors of long-term outcomes35,36  and represents a major endpoint of current treatment strategies incorporating autotransplantation up-front. To more carefully identify high-quality responses occurring beyond the CR level, the IMWG has recently introduced the category of stringent CR, as defined by negative immunofixation, normal free-light chain ratio, and absence of clonal bone marrow plasma cells by immunohistochemistry.37  It is probable that incorporation of novel agents into ASCT results in increased rates of immunophenotypic and/or molecular remissions38  compared with that reported in the recent past.

Other studies have also emphasized the adverse prognostic importance of residual focal lesions detected by magnetic resonance imaging.39  In contrast, sustained CR is predictive of favorable long-term outcomes.40  Therefore, not only attainment of CR per se, but maintenance of a durable CR, appears to be a major prognostic variable in MM. Interestingly, achievement of CR does not seem to be of critical prognostic relevance for several subgroups of patients, including those with low-risk disease or in whom active MM reverts to an indolent phase similar to that of monoclonal gammopathy of undetermined significance.41 

Patients who are eligible for early ASCT typically receive a limited number of cycles of induction therapy to reduce tumor cell mass and bone marrow plasma cell infiltration before collection of peripheral blood stem cells. Compared with conventional treatments used in the past, a number of novel agents are now available that affect increased rates of CR. Currently, these novel agents are incorporated into induction regimens to enhance the depth of response before ASCT and further improve post-ASCT outcomes.

The activity of thalidomide, especially when combined with dexamethasone (TD), in the relapsed/refractory setting has provided the rationale for the design of phase 2 and 3 trials investigating the role of this regimen in patients with newly diagnosed disease.42-44  In 2005, a retrospective case-matched study provided the first demonstration of superior rate and depth of response affected by TD compared with VAD as induction therapy in preparation for ASCT,42  a finding confirmed by a subsequent phase 3 study43  (Table 1). Based on the results of a randomized study showing a higher response rate with TD compared with high-dose dexamethasone44  (Table 1), the United States Food and Drug Administration granted accelerated approval for TD in patients with newly diagnosed MM. As a result, over the past years, TD has emerged as one of the most commonly used induction regimens in the United States and European countries (European Union).

Table 1

Phase 2 and 3 studies of thalidomide-dexamethasone and triplet thalidomide-based combinations in preparation for ASCT

RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR/ ≥ VGPR, %
TD vs 100 76 10/19 NR NR NR NR 42  
VAD (retrospective case-matched study) 100 52 8/14 NR NR NR NR  
TD vs 103 63 4/NR NR NR NR NR 44  
VAD 104 41 0/NR NR NR NR NR  
TD vs 100 66 NR/35 68 NR/44 NR NR 43  
Descamethasone 104 52 NR/13 62 NR/42 NR NR  
TAD vs 268 71 3/37 84 14/54 median, 34 mo median, 73 mo 47  
VAD 268 57 2/18 76 12/44 median, 22 mo median, 60 mo  
      P < .001 P = .77  
CTD vs NR 87 19/NR NR 51/NR NR NR 48  
CVAD NR 75 9/NR NR 40/NR NR NR  
TT2 + THAL vs 323 NR NR NR 62/NR 5-yr, 56% 5-yr, 65% 46  
TT2 without THAL 345 NR NR NR 43/NR 5-yr, 44% 5-yr, 65%  
      P = .01 P = .90  
Double ASCT + THAL vs 135 NR NR/30 NR NR/68 4-yr, 51% 5-yr, 69% 45  
Double ASCT without THAL (retrospective case-matched study) 135 NR NR/15 NR NR/49 4-yr, 31% 5-yr, 53%  
      P = .001 P = .07  
RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR/ ≥ VGPR, %
TD vs 100 76 10/19 NR NR NR NR 42  
VAD (retrospective case-matched study) 100 52 8/14 NR NR NR NR  
TD vs 103 63 4/NR NR NR NR NR 44  
VAD 104 41 0/NR NR NR NR NR  
TD vs 100 66 NR/35 68 NR/44 NR NR 43  
Descamethasone 104 52 NR/13 62 NR/42 NR NR  
TAD vs 268 71 3/37 84 14/54 median, 34 mo median, 73 mo 47  
VAD 268 57 2/18 76 12/44 median, 22 mo median, 60 mo  
      P < .001 P = .77  
CTD vs NR 87 19/NR NR 51/NR NR NR 48  
CVAD NR 75 9/NR NR 40/NR NR NR  
TT2 + THAL vs 323 NR NR NR 62/NR 5-yr, 56% 5-yr, 65% 46  
TT2 without THAL 345 NR NR NR 43/NR 5-yr, 44% 5-yr, 65%  
      P = .01 P = .90  
Double ASCT + THAL vs 135 NR NR/30 NR NR/68 4-yr, 51% 5-yr, 69% 45  
Double ASCT without THAL (retrospective case-matched study) 135 NR NR/15 NR NR/49 4-yr, 31% 5-yr, 53%  
      P = .001 P = .07  

Studies incorporating thalidomide-dexamethasone throughout double ASCT are also included.

CTD indicates cyclophosphamide, thalidomide, dexamethasone; CVAD, cyclophosphamide added to VAD (vincristine, doxorubicin, dexamethasone); NR, not reported; TAD, thalidomide, doxorubicin, dexamethasone; TD, thalidomide, dexamethasone; THAL, thalidomide; and TT2, Total Therapy 2.

In 2 additional studies in which TD was incorporated into double ASCT and given from the outset through the second ASCT45  or until relapse/progression,46  superior rates of CR or at least VGPR, EFS, and OS were seen with TD plus double ASCT compared with tandem transplantation not incorporating thalidomide (Table 1). However, the rate of adverse events, in particular peripheral neuropathy and venous thromboembolism, was consistently high with thalidomide maintenance therapy and led to drug discontinuation in 30% and 60% of patients after 2 and 4 years, respectively.46 

Two phase 3 trials explored the activity of induction regimens combining TD with doxorubicin or cyclophosphamide in transplantation candidates. In one study, TD and doxorubicin provided a significantly higher rate of VGPR or better compared with VAD (37% vs 18%), a gain that was maintained after the first ASCT (54% vs 44%, respectively).47  Median EFS for patients randomly assigned to TD and doxorubicin followed by post-ASCT thalidomide maintenance was 34 months versus 22 months for those assigned to VAD and subsequent maintenance with interferon.

In another study, superior rates of CR both before and after ASCT were seen with TD and cyclophosphamide compared with cyclophosphamide added to VAD (pre-ASCT, 19% vs 9%; and post-ASCT, 51% vs 40%, respectively).48 

The role of up-front standard-dose bortezomib (1.3 mg/m2) given twice weekly either as a single agent or with added dexamethasone in patients with suboptimal response to the first cycles of therapy was initially explored in patients who were either eligible or ineligible for ASCT (Table 2).49,50  In 2 additional phase 2 studies, bortezomib and high-dose dexamethasone (VD) were given either in combination51  or on an alternating basis52  before ASCT. The rate of at least VGPR was 31% with VD and 22.5% with the alternating schedule; the corresponding value after ASCT was 55% in each of the 2 studies. In a phase 3 study, VD was prospectively compared with VAD as induction therapy in preparation for single or double ASCT; in both arms, lenalidomide was given as post-ASCT consolidation and maintenance therapy.53  After 4 21-day cycles, the rates of at least VGPR, including CR and near CR (nCR), affected by VD were significantly higher than with VAD (≥ VGPR, 38% vs 15%; CR-nCR, 15% vs 6%, respectively), a gain maintained after both the first and second ASCT (≥ VGPR, 68% vs 47%; CR-nCR, 39.5% vs 22.5%, respectively). A borderline, albeit not statistically significant, PFS benefit was seen in the VD arm compared with the VAD arm (median, 36 vs 30 months, respectively).

Table 2

Phase 2 studies of bortezomib-based regimens in preparation for ASCT

RegimenNAfter induction
After transplantation
PFS, medianOS, 30 moReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR / ≥ VGPR, %
V (single agent) 64 41 NR (9)*/17 NR NR 17 mo 30 mo, 79% 49  
V ± D 32 87.5 6 (25)*/NR NR NR NR NR 50  
VD 48 66 NR (21)*/31 90 NR (33)*/55 NR NR 51  
V alternated with D 40 65 12.5/22.5 88 33/55 NR NR 52  
PAD-1 21 95 24 (5)*/62 95 43 (14)*/81 median, 29 mo 2-yr, 95% 54  
PAD-2 20 89 11 (5)*/42 90 37 (5)*/53 median, 24 mo 2-yr, 73% 54  
VDD 50 78 NR (27)*/NR 93 27/NR NR NR 56  
VDD 40 85 NR (37.5)*/57.5 87 NR (57)*/77 2-yr, 80% 2-yr, 92% 57  
CyBorD 33 88 3 (39)*/61 NR NR (70)*/74 NR NR 58  
VCD 391 85.4 NR (15)*/37 NR NR NR NR 59  
VTD vs VTDC 49 vs 48 100 vs 96 (29)/69 vs (31)/69 100 vs 100 (50)/87 vs (44)/85 NR 1-yr, 94.1% vs 94.2% 66  
TT3 + VTD-PACE vs TT2 + THAL (retrospective comparison) 303 vs 323 NR vs NR NR vs NR NR vs NR 2-yr, 54/NR vs 51/NR 2-yr, 84% vs 77% (P = .008) 2-yr, 87% vs 83% (P = .12) 67  
RegimenNAfter induction
After transplantation
PFS, medianOS, 30 moReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR / ≥ VGPR, %
V (single agent) 64 41 NR (9)*/17 NR NR 17 mo 30 mo, 79% 49  
V ± D 32 87.5 6 (25)*/NR NR NR NR NR 50  
VD 48 66 NR (21)*/31 90 NR (33)*/55 NR NR 51  
V alternated with D 40 65 12.5/22.5 88 33/55 NR NR 52  
PAD-1 21 95 24 (5)*/62 95 43 (14)*/81 median, 29 mo 2-yr, 95% 54  
PAD-2 20 89 11 (5)*/42 90 37 (5)*/53 median, 24 mo 2-yr, 73% 54  
VDD 50 78 NR (27)*/NR 93 27/NR NR NR 56  
VDD 40 85 NR (37.5)*/57.5 87 NR (57)*/77 2-yr, 80% 2-yr, 92% 57  
CyBorD 33 88 3 (39)*/61 NR NR (70)*/74 NR NR 58  
VCD 391 85.4 NR (15)*/37 NR NR NR NR 59  
VTD vs VTDC 49 vs 48 100 vs 96 (29)/69 vs (31)/69 100 vs 100 (50)/87 vs (44)/85 NR 1-yr, 94.1% vs 94.2% 66  
TT3 + VTD-PACE vs TT2 + THAL (retrospective comparison) 303 vs 323 NR vs NR NR vs NR NR vs NR 2-yr, 54/NR vs 51/NR 2-yr, 84% vs 77% (P = .008) 2-yr, 87% vs 83% (P = .12) 67  

CyBorD indicates cyclophosphamide, bortezomib, dexamethasone; D, dexamethasone; NR, not reported; PACE, cisplatin, doxorubicin, cyclophosphamide, etoposide; PAD, bortezomib, doxorubicin, dexamethasone; PAD-2, reduced-dose bortezomib; THAL, thalidomide; TT3, Total Therapy 3; V, bortezomib; VCD, bortezomib, cyclophosphamide, dexamethasone; VDD, bortezomib, pegylated liposomal doxorubicin, dexamethasone; and VTD, bortezomib, thalidomide, dexamethasone.

*

At least near CR.

Cytotoxic drugs added to VD as part of a 3-drug regimen in preparation for ASCT have included doxorubicin or cyclophosphamide (Table 2). A combination of bortezomib, doxorubicin, and dexamethasone, referred to as PAD, was investigated in 2 small cohorts of patients who received either standard-dose or reduced-dose bortezomib (1.0 mg/m2) on a twice-weekly basis (Table 2).54  In a phase 3 study, the PAD regimen was compared with VAD as induction therapy before 1 or 2 autotransplantations.55  Superior CR-nCR rates were seen with PAD compared with VAD after both induction (11% vs 5%, respectively) and autotransplantation(s) (30% vs 15%). PAD induction followed by ASCT and subsequent bortezomib maintenance was associated with significantly longer PFS and OS compared with VAD induction and post-ASCT thalidomide maintenance therapy (Table 2). Two additional phase 2 studies confirmed the activity of a PAD-like induction regimen incorporating pegylated liposomal doxorubicin (Table 2).56,57 

In addition, cyclophosphamide has also demonstrated substantial activity when combined with VD (CyBorD or VCD) in preparation for ASCT.58,59  In 2 phase 2 studies, the rate of at least VGPR was between 37% and 61%, a range that reflected heterogeneities between studies with respect to the number of planned treatment cycles and the delivered cyclophosphamide dose.

Preclinical data suggesting that IMiDs increase bortezomib activity, provided the rationale for combining thalidomide with VD (VTD). Promising rates of high-quality responses reported with VTD in small cohorts of relapsed/refractory and newly diagnosed MM patients60  led to the design of a phase 3 study of VTD versus TD as induction therapy before, and consolidation therapy after, double ASCT.61  After three 21-day induction cycles, VTD was superior to TD with respect to all response categories, including CR, CR-nCR (31% vs 11%), and at least VGPR (62% vs 28%). Increased frequencies of high-quality responses in the VTD arm compared with the TD arm were also seen after double autotransplantation and subsequent consolidation therapy (CR-nCR, 62% vs 45%; ≥ VGPR, 85% vs 68%, respectively). The estimated 3-year PFS for the VTD group of patients was significantly longer than for those assigned to TD plus double ASCT (68% vs 56%, respectively). In two additional phase 3 studies comparing VTD with either TD62  or VD63  as induction therapy in preparation for a single ASCT, superior rates of high-quality responses, both before and after ASCT, and extended PFS62  were seen with the triplet regimen (Table 3). Remarkable activity of VTD was further confirmed by several phase 2 studies,64,65  including a prospective comparison of VTD with the same regimen combined with cyclophosphamide66  (Table 2).

Table 3

Phase 3 trials of bortezomib-based regimens in preparation for ASCT

RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR/ ≥ VGPR, %
VD vs 223 78.5 6 (15)*/38 80 16 (35)*/54 median, 36 mo 3-yr, 81% 53  
VAD 218 63 1 (6)*/15 77 9 (18)*/37 median, 30 mo 3-yr, 77%  
      (P = .06) (P = .5)  
VTD vs 236 93 19 (31)*/62 93 42 (55)*/82 3-yr, 68% 3-yr, 86% 61  
TD 238 79 5 (11)*/28 84 30 (41)*/64 3-yr, 56% 3-yr, 84%  
      (P = .005) (P = .3)  
VBMCP/VBAD + V vs 129 75 21/36 73 38/51 38 mo NR 62  
VTD vs 130 85 35/60 77 46/65 27 mo NR  
TD 127 62 14/29 58 24/40 Not reached NR  
      (P = .006)   
PAD vs 371 78 NR (11)*/42 88 NR (30)*/61 3-yr, 36% 3-yr, 78% 55  
VAD 373 55 NR (5)*/15 77 NR (15)*/36 3-yr, 27% 3-yr, 70%  
      (P = .01) (P = .02)  
VD vs 99 81 12 (22)*/35 84 33 (54)*/59 NR NR 63  
vTD 100 90 13 (32)*/51 90 30 (61)*/73 NR NR  
RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR, %CR/ ≥ VGPR, %CR + PR, %CR/ ≥ VGPR, %
VD vs 223 78.5 6 (15)*/38 80 16 (35)*/54 median, 36 mo 3-yr, 81% 53  
VAD 218 63 1 (6)*/15 77 9 (18)*/37 median, 30 mo 3-yr, 77%  
      (P = .06) (P = .5)  
VTD vs 236 93 19 (31)*/62 93 42 (55)*/82 3-yr, 68% 3-yr, 86% 61  
TD 238 79 5 (11)*/28 84 30 (41)*/64 3-yr, 56% 3-yr, 84%  
      (P = .005) (P = .3)  
VBMCP/VBAD + V vs 129 75 21/36 73 38/51 38 mo NR 62  
VTD vs 130 85 35/60 77 46/65 27 mo NR  
TD 127 62 14/29 58 24/40 Not reached NR  
      (P = .006)   
PAD vs 371 78 NR (11)*/42 88 NR (30)*/61 3-yr, 36% 3-yr, 78% 55  
VAD 373 55 NR (5)*/15 77 NR (15)*/36 3-yr, 27% 3-yr, 70%  
      (P = .01) (P = .02)  
VD vs 99 81 12 (22)*/35 84 33 (54)*/59 NR NR 63  
vTD 100 90 13 (32)*/51 90 30 (61)*/73 NR NR  

PAD indicates bortezomib, doxorubicin, dexamethasone; V, bortezomib; TD, thalidomide-dexamethasone; VAD, vincristine, doxorubicin, dexamethasone; VBAD, vincristine, carmustine, doxorubicin, dexamethasone; VBMCP, vincristine, carmustine, melphalan, cyclophosphamide, prednisone; VD, bortezomib, dexamethasone; VTD, bortezomib (1.3 mg/m2), thalidomide, dexamethasone; vTD, bortezomib (1.0 mg/m2), thalidomide, dexamethasone; and NR, not reported.

*

At least near CR.

In Total Therapy 3, VTD combined with cisplatin, doxorubicin, cyclophosphamide, and etoposide was given as induction therapy before, and consolidation after, double ASCT, whereas VTD maintenance therapy was continued for 1 year after ASCT.67  Compared with Total Therapy 2 incorporating TD into double ASCT, Total Therapy 3 significantly improved 2-year EFS (77% vs 84%) and duration of CR.

Lenalidomide plus high-dose dexamethasone (480 mg total in a 28-day cycle; RD) was prospectively compared with lenalidomide and low-dose dexamethasone (160 mg total per cycle; Rd) as frontline therapy for MM.9  Patient enrollment into the study was not restricted by age or eligibility for ASCT. Despite the overall response rate, including VGPR or better within 4 cycles of therapy was significantly higher with RD compared with Rd (42% vs 24%, respectively), a substantially higher toxicity and early mortality was seen with RD, particularly in patients older than 65 years. On landmark analysis, the 3-year OS of patients who received ASCT after RD or Rd was 92%; the corresponding value for patients who continued on primary therapy and did not receive ASCT was 79%.

Lenalidomide and dexamethasone were combined with bortezomib to form a triplet regimen (RVD), which has been investigated in limited series of patients with newly diagnosed MM.68-70  In a phase 1/2 study, a total of 66 patients who were either transplantation-eligible or ineligible for ASCT received a maximum of 8 RVD cycles; in responders, RVD maintenance was allowed.69  After 4 cycles, the rate of at least nCR and VGPR was 6% and 11%, respectively. However, in approximately two-thirds of patients, the quality of response improved from cycle 4 through cycle 8, and a further improvement was also seen in the maintenance phase.

In addition to RVD, alternative lenalidomide-containing regimens have included a combination of lenalidomide-cyclophosphamide-dexamethasone and a quadruplet regimen in which cyclophosphamide was added to RVD71  (Table 4). A prospective comparison of RVD with VCD and cyclophosphamide combined with RVD given for up to 8 cycles has been recently reported; the rate of CR-nCR after 4 cycles was in the 7%, 3%, and 10% range, respectively.72  An additional quadruplet regimen incorporating lenalidomide, bortezomib, dexamethasone, and pegylated liposomal doxorubicin was explored.73  After a median of 4 cycles, the rates of CR-nCR and VGPR or better were 30% and 58%, respectively.

Table 4

Phase 2 and 3 trials of doublet and triplet lenalidomide-based induction treatments for transplant-eligible and transplant-ineligible patients

RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR (best response), %CR/ ≥ VGPR (best response), %CR + PR, %CR + nCR, %
RD vs 223 81 5/50 NR NR median, 19 mo median, not reached 9  
Rd 222 70 4/40 NR NR median, 25 mo median, not reached  
      (P = .02) (P = .4)  
RVD 66 100 29 (39)*/67 NR NR 18 mo, 75% 18 mo, 97% 69  
RVD vs 42 83 24 (40)*/50 NR NR NR NR 72  
VCD vs 32 75 22 (31)*/41      
RVCD 42 86 24 (33)*/57 NR NR NR NR 73  
RVDD 57 4 cycles, 96 4 cycles, NR (30)*/58 NR NR NR NR  
RegimenNAfter induction
After ASCT
PFSOSReference
CR + PR (best response), %CR/ ≥ VGPR (best response), %CR + PR, %CR + nCR, %
RD vs 223 81 5/50 NR NR median, 19 mo median, not reached 9  
Rd 222 70 4/40 NR NR median, 25 mo median, not reached  
      (P = .02) (P = .4)  
RVD 66 100 29 (39)*/67 NR NR 18 mo, 75% 18 mo, 97% 69  
RVD vs 42 83 24 (40)*/50 NR NR NR NR 72  
VCD vs 32 75 22 (31)*/41      
RVCD 42 86 24 (33)*/57 NR NR NR NR 73  
RVDD 57 4 cycles, 96 4 cycles, NR (30)*/58 NR NR NR NR  

CRD indicates cyclophosphamide, lenalidomide, dexamethasone; NR, not reported; RD, lenalidomide, high-dose dexamethasone; Rd, lenalidomide, low-dose dexamethasone; RVD, lenalidomide, bortezomib, dexamethasone; RVCD, lenalidomide, bortezomib, cyclophosphamide, dexamethasone; RVDD, lenalidomide, bortezomib, pegylated liposomal doxorubicin, dexamethasone; and VCD, bortezomib, cyclophosphamide, dexamethasone.

*

At least near CR.

Cytogenetic abnormalities

The prognostic value of major cytogenetic abnormalities and the impact of novel agents on clinical outcomes of patients carrying different cytogenetic changes have been recently reviewed by the IMWG.74  Detection at diagnosis of translocation t(4;14) and t(14;16) or deletion of chromosome 17, del(17p), by fluorescence in situ hybridization, as well as deletion/monosomy of del(13q) or hypodiploidy by metaphase cytogenetics define approximately one-fourth of patients75  who in the past years did not benefit from ASCT and had shortened remission duration and OS.76,77 

Recent reports have suggested that incorporation of novel agents into ASCT may overcome, at least in part, the poor prognosis imparted by high-risk cytogenetic profiles. In two phase 3 studies of VD78  and PAD55  induction therapy followed by lenalidomide and bortezomib maintenance therapy, respectively, t(4;14)-positive patients had better outcomes than the control groups who carried the same abnormality but received VAD induction followed by maintenance therapy with either lenalidomide78  or thalidomide.55  However, in both of these studies, t(4;14) partly retained its adverse influence on PFS and OS, even among patients treated with bortezomib-based induction regimens and subsequent maintenance with novel agents.55,78  In contrast, in a phase 3 study of VTD induction and consolidation therapy plus double ASCT, PFS curves were almost identical regardless of the presence or absence of t(4;14).61  In an additional study, incorporation of VTD into double ASCT as part of both induction and consolidation therapy and as post-ASCT maintenance therapy resulted in improved CR duration, PFS, and OS for the gene expression profile-defined high-risk subgroup of patients carrying the MMSET/FGFR3 hybrid transcript.67  The role of bortezomib-based regimens and ASCT for the treatment of del(17p)-positive patients needs to be carefully evaluated in larger sample sizes than those explored so far.78  In particular, areas of major interest include the ability of less or more intense treatments (eg, doublet vs triplet or quadruplet combinations) given for different time periods (eg, short-term vs long-term) to impact on the poor prognosis related to this high-risk cytogenetic profile.

In most studies incorporating thalidomide as part of induction therapy79  or as post-ASCT maintenance,80,81  the outcome of patients with del(13q), t(4;14), and/or del(17p) was inferior to that of patients who lacked these abnormalities. Conflicting results concerning the ability of lenalidomide to overcome the poor prognosis associated with del(13q) and t(4;14) were found in 2 retrospective studies of patients with relapsed/refractory MM.82,83  The adverse prognostic impact of del(17p) was emphasized in one of these studies.83  In a recent report on newly diagnosed MM patients who were either transplantation-eligible or ineligible for ASCT and received lenalidomide-dexamethasone up-front, both response duration and PFS, but not OS, were significantly worse when high-risk genetic abnormalities were present at baseline.84 

Renal failure

In patients with MM and renal failure, rapid reduction of myeloma cell mass and recovery of normal renal function are critical goals of both myeloma-specific therapy and supportive care measures.85  Neither thalidomide86  nor bortezomib87  is excreted through the kidneys, and dose adjustments are not required for patients with renal impairment. In contrast, it is mandatory to modify the dose and schedule of lenalidomide according to renal clearance.85  In general, bortezomib-based regimens are the preferred treatment option in this setting, as recently recommended by the IMWG.85 

Thalidomide and lenalidomide

For patients who receive thalidomide up-front, either as a single agent or in combination therapy, the most common toxicities include constipation, somnolence, and peripheral neuropathy (PN).88  Thalidomide-induced PN is more frequently sensory or sensorimotor, is dose-dependent (more prevalent with doses higher than 200 mg/day) and duration-dependent (more likely to occur after 6-12 months).89,90  Reduction of the dose or discontinuation of thalidomide according to the severity of PN are measures commonly used in clinical practice. Unlike thalidomide, lenalidomide induces myelosuppression, mainly neutropenia and thrombocytopenia, which can be managed via dose reductions and/or hematopoietic growth factor support.91  PN is uncommonly seen with lenalidomide. Another major challenge to be considered in patients who receive thalidomide or lenalidomide up-front is the increased risk of thromboembolic complications.92,93  Adequate guidelines on the most appropriate thromboprophylactic treatments have been provided by the IMWG.92  Finally, hypothyroidism is an additional important adverse event associated with long-term therapy incorporating thalidomide or lenalidomide. Long-term use of lenalidomide is also associated with severe diarrhea and cramps in a subset of patients.

The effect of newer induction regimens, in particular those incorporating lenalidomide, on peripheral blood stem cell mobilization and the optimal strategies to obtain adequate stem cell harvests have recently been reviewed by the IMWG.94 

Bortezomib

One of the most important nonhematologic toxicities of bortezomib is PN, which may lead to impaired quality of life. Bortezomib-induced PN is predominantly sensory, although in < 10% of cases motor neuropathy has been reported.95  Unlike neurologic toxicity associated with thalidomide, neuropathic pain, mainly located in the fingertips and toes, is a major problem with bortezomib. Major risk factors of bortezomib-induced PN include the cumulative dose of the drug and treatment schedule. Attempts to decrease the rate and severity of neurologic toxicity in transplantation candidates have included either dose reduction of bortezomib given on a twice-weekly basis63  or once-weekly administration of the drug at a higher dose to maintain activity.96  In elderly, transplantation-ineligible patients for whom treatment plan was composed of long-term exposure to melphalan and prednisone combined with bortezomib (given twice weekly for 4 cycles, followed by once-weekly administration for the next 5 cycles), the overall risk of PN was 47%, including 13% grade 3 or 4.8  In 2 recent studies of melphalan and prednisone combined with standard-dose bor-tezomib given on a weekly basis for 6 to 9 cycles, the incidence of grade 3 or 4 PN was reduced to 6% to 7%, whereas efficacy was retained.97,98  Whether these favorable results may be obtained in transplantation-eligible patients who usually receive a shorter induction therapy is an issue not yet addressed in clinical trials. Notably, compared with single-agent bortezomib short-term use of combined bortezomib and thalidomide was not associated with a major increase in the frequency of any grade and grade 3 or 4 PN.53,61,64,66  Besides symptomatic therapy, the optimal management of bortezomib-induced PN requires its early recognition and dose reduction or discontinuation of the drug using a validated algorithm; an alternative option may be to prolong the dosing schedule. Provided these procedures are promptly adopted, approximately 70% of patients have partial or complete reversibility of their neurologic symptoms. The issue of the management of treatment-emergent PN in MM has been recently addressed.99  Severe thrombocytopenia occurs in approximately 5% or less of patients in the frontline setting. An additional adverse effect commonly seen with bortezomib-based regimens is reactivation of varicella zoster virus,100  a complication that can be virtually abrogated with acyclovir prophylaxis.101 

Consolidation treatment is generally short-term and aims to improve responses after ASCT. Upgraded rates of CR and CR-nCR, in the range between 10% and 30%, have been recently reported with post-ASCT use of bortezomib or lenalidomide as single agents102,103  or with VTD.104  In several of these studies, consolidation therapy with VTD yielded molecular remissions in up to 60% of patients.38,105 

Maintenance treatment is given for a prolonged time period with the goal of extending the duration of response, PFS, and OS, while maintaining a good quality of life.106  Several randomized studies showed a PFS benefit with thalidomide as single agent or combined with prednisone as maintenance therapy after ASCT.46,47,80,81,107,108  In 2 of these studies, OS was extended in the thalidomide arm,80,107  a gain lost when thalidomide was also given as part of induction therapy before ASCT.46,47,81  Concerns exist about the use of thalidomide maintenance after ASCT, including the possible emergence of tumor-resistant clones in patients with prolonged exposure to this agents and its lack of efficacy in patients with adverse cytogenetic abnormalities.109  However, the major caveat that precludes a widespread use of thalidomide maintenance is the toxicity related to long-term administration of this agent, primarily PN. In several studies, thalidomide-induced PN led to discontinuation rates in the 60% range46,81  and impairment in patients' quality of life.108  Lenalidomide is an attractive alternative to thalidomide because of the lack of neurologic toxicity. Two independent randomized trials have recently shown a significantly longer PFS110,111  for patients randomized to lenalidomide maintenance (5-15 mg/day) compared with the placebo group after a single or double ASCT.105,106  An increased incidence of second primary malignancies, in the 7% range, has been recently reported. Although a concerted effort is needed to better define the underlying mechanisms and identify risk factors, the optimal role and duration of lenalidomide maintenance therapy need to be tested in future clinical trials.

In conclusion, incorporation of IMiDs and/or bortezomib into newer regimens given in preparation for ASCT has been extensively explored using a wide range of different combinations. Doublet therapies combining either an IMiD or bortezomib with dexamethasone (eg, TD or Rd or VD) affected higher overall response rates than traditional treatments,42,43,45,53  although the lowest rate of high-quality responses was seen with TD. Compared with doublets, such as TD and VD, triplet induction regimens, in particular, bortezomib plus thalidomide and dexamethasone (VTD), further increased the rate of CR and/or at least VGPR, both before and after autotransplantation.61-63  In the context of triplet regimens combining bortezomib with an IMiD, RVD is an attractive alternative to VTD,70  although favorable results reported so far are not backed by phase 3 clinical studies. Several newer induction treatments, such as VD, VTD, PAD, and Rd, have been included as a category 1 recommendation, which signifies a high-level of evidence and uniform consensus among panel members, in the United States National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology for Multiple Myeloma Version 1.2011.23 

Enhanced high-quality responses affected by newer induction regimens translated into even higher frequencies of CR or at least VGPR after single or double ASCT. Although extended PFS was reported in several of these studies,45-47,53,55,61  no OS benefit was generally seen, a finding that reflects the lack of adequate power and/or follow-up to detect survival differences. Furthermore, proving an OS benefit at this time is probably difficult because of the rapidly increasing availability of effective salvage therapies at the time of relapse.

Based on these considerations and the close relationship between maximal response to induction therapy and favorable prognosis after ASCT, it is likely that many investigators in the IMWG would recommend using one of the bortezomib-containing triplet regimens as up-front induction therapy in a transplantation-eligible MM patient. However, other IMWG investigators might feel that until OS differences emerge, low-risk patients may have the option of choosing either a doublet regimen with low morbidity, such as Rd, or a bortezomib-based triplet, provided that they are properly informed about the pros and cons, particularly the risk of early PN with bortezomib. Besides once-weekly administration of bortezomib, the introduction into the clinical practice of subcutaneous bortezomib that has recently shown a significantly lower risk of PN compared with intravenous bortezomib in patients with relapsed/refractory disease112  and carfilzomib, a second-generation irreversible proteasome inhibitor with significantly less neurotoxicity than bortezomib, may solve some of these issues in the near future.

In the absence of randomized studies comparing different induction regimens, it is difficult to recommend one induction regimen over another. However, particular patient and disease characteristics may guide the clinician to select the most appropriate therapy. For instance, preliminary data suggest that bortezomib-based regimens, such as VTD, VD, and PAD, can partially or completely abrogate the poor prognosis related to t(4;14),55,61,78  although more mature data about del(17p) are needed. In patients presenting with acute renal failure, both bortezomib- and thalidomide-based regimens can be safely given, whereas lenalidomide requires appropriate dose reductions and frequent monitoring of blood counts. In patients at high risk of thromboembolic complications, a bortezomib-based regimen may be preferable. In contrast, the presence of neuropathy at baseline might suggest excluding bortezomib-based or thalidomide-based treatments in favor of a regimen, such as Rd. In the studies reported so far, the dose of dexamethasone was variable. However, high-dose dexamethasone is needed in those patients in whom a prompt reduction in tumor cell mass is required. Finally, it is worth remembering that in many countries novel-agent-based induction therapies for younger, transplantation-eligible patients are not approved as yet. In these cases, the choice of induction regimen should be based on drug availability; furthermore, referral of patients to a specialized myeloma center with access to studies of novel agents is recommended.

The usual choice of giving 3 to 6 cycles of induction therapy to maximize the depth of response before early ASCT represents a reasonable balance between maximum benefit and minimum toxicity. However, an alternative choice that can be discussed with the patient, particularly if response to therapy is favorable and he/she is unwilling to proceed to early ASCT, is to continue induction for as long as maximal tumor reduction is achieved and then to maintain response until relapse or progression, at which time salvage ASCT can be performed. In this scenario, especially in patients treated with lenalidomide-based regimens, peripheral blood stem cells should be collected early, after 4 to 6 cycles of induction therapy. The best timing of ASCT in the novel agent era represents an area of active debate and major interest. Unless final results of ongoing clinical trials comparing early versus late ASCT plus novel agents will be available, ASCT up-front should continue to be considered the preferred approach for a patient who is eligible to tolerate HDT. More recently, the treatment paradigm for transplantation-eligible MM patients has continued to evolve with the introduction of the novel agents as consolidation and maintenance therapies. Mature results demonstrating the role, if any, of consolidation therapy in improving clinical outcomes and the impact of maintenance therapy on OS are needed before these strategies are widely adopted. In the meantime, the choice of using consolidation and/or maintenance therapy outside clinical trials is at the patient's and physician's discretion. If post-ASCT therapy with lenalidomide is planned, the IMWG recommends that the benefits of extended disease control versus potential risks of second malignancies with continued lenalidomide therapy be discussed with each patient. For many other important and still unaddressed questions, prospective randomized phase 3 studies are currently planned or underway.

The online version of this article contains a data supplement.

R.O. was supported by Celgene and Millennium (research funding). J.B. was supported by Jansen and Celgene. O.S. was supported by Janssen and Novartis (research funding). P.S. was supported by Janssen, Celgene, and Onyx (research support). R.N. was supported by Millenium and Celgene (research support).

Contribution: All authors developed the consensus, provided critical review and edits to the manuscript, gave approval to the final manuscript, and significantly participated in the development and writing of the manuscript.

Conflict-of-interest disclosure: M.C. received honoraria from Janssen, Celgene, and Millennium. A.P. received honoraria from Celgene, Janssen, Merck, and Amgen and is on the advisory boards for Celgene and Janssen. P.M. received honoraria from Celgene and Janssen. R.O. is on the advisory boards for Celgene, Millennium, Novartis, and Onyx. J.B. received honoraria for lectures from Janssen and Celgene. O.S. received honoraria from Amgen, Celgene, Janssen, and Novartis. H.L. received honoraria for advisory boards for Celgene and is on the speaker's bureau of Celgene and Ortho-Biotech. M.A.D. received honoraria from Ortho-Biotech, Celgene, and Millenium. P.S. is on the advisory boards for Janssen, Celgene, Millennium, and Onyx. M.B. received honoraria from Celgene and Janssen. K.C.A. is on the advisory boards for Celgene, Millennium, Onyx, Bristol-Myers Squibb, Merck, and Novartis and is the founder of Acetylon. P.G.R. is on the advisory boards for Celgene, Millennium, Johnson & Johnson, Bristol-Myers Squibb, and Novartis. W.B. is a consultant and speaker for Millenium and Celgene, is a consultant for Onyx and Genzyme, and performed clinical trials for Millenium, Celgene, Onyx, and Genzyme. H.E.J. is on the advisory board for and received honoraria from Janssen, Johnson & Johnson, and Amgen. G.G. is a consultant for Fujomoto Pharmaceutical Cooperation Japan. P.L.B. is a consultant for Celgene and Novartis. D.H.V. is on the advisory boards for Amgen, Celgene, and Onyx and on the speaker's bureau of Millennium and Celgene. R.N. is a consultant and speaker for Millenium and Celgene, received research support from Millenium and Celgene, and is a consultant for Onyx. B.G.M.D. is on the advisory boards for Millennium, Celgene, and Onyx. J.F.S.-M. is on advisory boards for Millenium, Celgene, and Janssen-Cilag. S.L. is a consultant for Millennium, Celgene, Bristol-Myers Squibb, Novartis, Merck, and Onyx. The remaining authors declare no competing financial interests.

Correspondence: Michele Cavo, Istituto di Ematologia Seràgnoli, Azienda Ospedaliero-Universitaria, Policlinico S. Orsola-Malpighi, via Massarenti 9, 40138, Bologna, Italy; e-mail: michele.cavo@unibo.it; and S. Vincent Rajkumar, Mayo Clinic, 200 First St SW, Rochester, MN 55905; e-mail: rajks@mayo.edu.

1
Altekruse
 
SF
Kosary
 
C
Krapcho
 
M
Neyman
 
N
Aminou
 
R
Waldron
 
W
SEER Cancer Statistics Review
1975–2007
Accessed March 10, 2011
Bethesda, MD
National Cancer Institute
 
2
Ludwig
 
H
Durie
 
BG
Bolejack
 
V
, et al. 
Myeloma in patients younger than age 50 years presents with more favorable features and shows better survival: an analysis of 10 549 patients from the International Myeloma Working Group.
Blood
2008
, vol. 
111
 
8
(pg. 
4039
-
4047
)
3
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
)
4
Cavo
 
M
Baccarani
 
M
The changing landscape of myeloma therapy.
N Engl J Med
2006
, vol. 
354
 
10
(pg. 
1076
-
1078
)
5
Kumar
 
SK
Rajkumar
 
SV
Dispenzieri
 
A
, et al. 
Improved survival in multiple myeloma and the impact of novel therapies.
Blood
2008
, vol. 
111
 
5
(pg. 
2516
-
2520
)
6
Palumbo
 
A
Bringhen
 
S
Liberati
 
AM
, et al. 
Oral melphalan, prednisone, and thalidomide in elderly patients with multiple myeloma: updated results of a randomized, controlled trial.
Blood
2008
, vol. 
112
 
8
(pg. 
3107
-
3114
)
7
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
)
8
San Miguel
 
JF
Schlag
 
R
Khuageva
 
NK
, et al. 
Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma.
N Engl J Med
2008
, vol. 
359
 
9
(pg. 
906
-
917
)
9
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
)
10
Bensinger
 
W
Stem-cell transplantation for multiple myeloma in the era of novel drugs.
J Clin Oncol
2008
, vol. 
26
 
3
(pg. 
480
-
492
)
11
Lonial
 
S
Cavenagh
 
J
Emerging combination treatment strategies containing novel agents in newly diagnosed multiple myeloma.
Br J Haematol
2009
, vol. 
145
 
6
(pg. 
681
-
708
)
12
Stewart
 
AK
Richardson
 
PG
San-Miguel
 
JF
How I treat multiple myeloma in younger patients.
Blood
2009
, vol. 
114
 
27
(pg. 
5436
-
5443
)
13
Attal
 
M
Harousseau
 
JL
Stoppa
 
AM
, et al. 
A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma: Intergroupe Francais du Myelome.
N Engl J Med
1996
, vol. 
335
 
2
(pg. 
91
-
97
)
14
Child
 
JA
Morgan
 
GJ
Davies
 
FE
, et al. 
High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma.
N Engl J Med
2003
, vol. 
348
 
19
(pg. 
1875
-
1883
)
15
Attal
 
M
Harousseau
 
JL
Facon
 
T
, et al. 
Single versus double autologous stem-cell transplantation for multiple myeloma.
N Engl J Med
2003
, vol. 
349
 
26
(pg. 
2495
-
2502
)
16
Blade
 
J
Rosinol
 
L
Sureda
 
A
, et al. 
High-dose therapy intensification compared with continued standard chemotherapy in multiple myeloma patients responding to the initial chemotherapy: long-term results from a prospective randomized trial from the Spanish cooperative group PETHEMA.
Blood
2005
, vol. 
106
 
12
(pg. 
3755
-
3759
)
17
Fermand
 
JP
Katsahian
 
S
Divine
 
M
, et al. 
High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe.
J Clin Oncol
2005
, vol. 
23
 
36
(pg. 
9227
-
9233
)
18
Barlogie
 
B
Kyle
 
RA
Anderson
 
KC
, et al. 
Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321.
J Clin Oncol
2006
, vol. 
24
 
6
(pg. 
929
-
936
)
19
Cavo
 
M
Tosi
 
P
Zamagni
 
E
, et al. 
Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study.
J Clin Oncol
2007
, vol. 
25
 
17
(pg. 
2434
-
2441
)
20
Kumar
 
SK
Dingli
 
D
Lacy
 
MQ
, et al. 
Autologous stem cell transplantation in patients of 70 years and older with multiple myeloma: results from a matched pair analysis.
Am J Hematol
2008
, vol. 
83
 
8
(pg. 
614
-
617
)
21
International Myeloma Working Group
Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group.
Br J Haematol
2003
, vol. 
121
 
5
(pg. 
749
-
757
)
22
Mateos
 
MV
Hernández
 
T
de la Rubia
 
J
, et al. 
Multicenter, randomized, open-label, phase III trial of lenalidomide-dexamethasone vs therapeutic abstention in smoldering multiple myeloma at high risk of progression to symptomatic MM: results of the first interim analysis [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 614
23
National Cancer Comprehensive Network
NCCN Clinical practice guidelines in oncology: multiple myeloma.
Accessed March 10, 2011 
24
Barosi
 
G
Boccadoro
 
M
Cavo
 
M
, et al. 
Management of multiple myeloma and related-disorders: guidelines from the Italian Society of Hematology (SIE), Italian Society of Experimental Hematology (SIES) and Italian Group for Bone Marrow Transplantation (GITMO).
Haematologica
2004
, vol. 
89
 
6
(pg. 
717
-
741
)
25
Smith
 
A
Wisloff
 
F
Samson
 
D
Guidelines on the diagnosis and management of multiple myeloma 2005.
Br J Haematol
2006
, vol. 
132
 
4
(pg. 
410
-
451
)
26
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
)
27
Fermand
 
JP
Ravaud
 
P
Chevret
 
S
, et al. 
High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial.
Blood
1998
, vol. 
92
 
9
(pg. 
3131
-
3136
)
28
Segeren
 
CM
Sonneveld
 
P
van der Holt
 
B
, et al. 
Overall and event-free survival are not improved by the use of myeloablative therapy following intensified chemotherapy in previously untreated patients with multiple myeloma: a prospective randomized phase 3 study.
Blood
2003
, vol. 
101
 
6
(pg. 
2144
-
2151
)
29
Fermand
 
J
High dose therapy supported with autologous blood stem cell transplantation in multiple myeloma: long term follow-up of the prospective studies of the MAG group [abstract].
Haematologica
2005
, vol. 
90
 
suppl 1
 
Abstract 40
30
Goldschmidt
 
H
Single vs. double high-dose therapy in multiple myeloma: second analysis of the GMMG-HD2 trial [abstract].
Haematologica
2005
, vol. 
90
 
suppl 1
 
Abstract 38
31
Kumar
 
A
Kharfan-Dabaja
 
MA
Glasmacher
 
A
Djulbegovic
 
B
Tandem versus single autologous hematopoietic cell transplantation for the treatment of multiple myeloma: a systematic review and meta-analysis.
J Natl Cancer Inst
2009
, vol. 
101
 
2
(pg. 
100
-
106
)
32
Giralt
 
S
Vesole
 
DH
Somlo
 
G
, et al. 
Re: Tandem vs single autologous hematopoietic cell transplantation for the treatment of multiple myeloma: a systematic review and meta-analysis.
J Natl Cancer Inst
2009
, vol. 
101
 
13
pg. 
964
  
author reply 6–7
33
Mehta
 
J
Re: Tandem vs single autologous hematopoietic cell transplantation for the treatment of multiple myeloma: a systematic review and meta-analysis.
J Natl Cancer Inst
2009
, vol. 
101
 
20
(pg. 
1430
-
1431
author reply 1–3
34
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 the University of Arkansas for Medical Sciences.
J Clin Oncol
2010
, vol. 
28
 
7
(pg. 
1209
-
1214
)
35
Harousseau
 
JL
Attal
 
M
Avet-Loiseau
 
H
The role of complete response in multiple myeloma.
Blood
2009
, vol. 
114
 
15
(pg. 
3139
-
3146
)
36
Chanan-Khan Giralt
 
S
Importance of achieving a complete response in multiple myeloma, and the impact of novel agents.
J Clin Oncol
2010
, vol. 
28
 
15
(pg. 
2612
-
2624
)
37
Durie
 
BG
Harousseau
 
JL
Miguel
 
JS
, et al. 
International uniform response criteria for multiple myeloma.
Leukemia
2006
, vol. 
20
 
9
(pg. 
1467
-
1473
)
38
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
)
39
Walker
 
R
Barlogie
 
B
Haessler
 
J
, et al. 
Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications.
J Clin Oncol
2007
, vol. 
25
 
9
(pg. 
1121
-
1128
)
40
Barlogie
 
B
Anaissie
 
E
Haessler
 
J
van Rhee
 
F
, et al. 
Complete remission sustained 3 years from treatment initiation is a powerful surrogate for extended survival in multiple myeloma.
Cancer
2008
, vol. 
113
 
2
(pg. 
355
-
359
)
41
Pineda-Roman
 
M
Bolejack
 
V
Arzoumanian
 
V
, et al. 
Complete response in myeloma extends survival without, but not with history of prior monoclonal gammopathy of undetermined significance or smouldering disease.
Br J Haematol
2007
, vol. 
136
 
3
(pg. 
393
-
399
)
42
Cavo
 
M
Zamagni
 
E
Tosi
 
P
, et al. 
Superiority of thalidomide and dexamethasone over vincristine-doxorubicin dexamethasone (VAD) as primary therapy in preparation for autologous transplantation for multiple myeloma.
Blood
2005
, vol. 
106
 
1
(pg. 
35
-
39
)
43
Macro
 
M
Divine
 
M
Uzunhan
 
Y
, et al. 
Dexamethasone + thalidomide (Dex/Thal) compared to VAD as a pre-transplant treatment in newly diagnosed multiple myeloma (MM): a randomized trial [abstract].
Blood
2006
, vol. 
108
 
11
 
Abstract 57
44
Rajkumar
 
SV
Blood
 
E
Vesole
 
D
Fonseca
 
R
Greipp
 
PR
Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group.
J Clin Oncol
2006
, vol. 
24
 
3
(pg. 
431
-
436
)
45
Cavo
 
M
Di Raimondo
 
F
Zamagni
 
E
, et al. 
Short-term thalidomide incorporated into double autologous stem-cell transplantation improves outcomes compared with double autotransplantation for multiple myeloma.
J Clin Oncol
2009
, vol. 
27
 
30
(pg. 
5001
-
5007
)
46
Barlogie
 
B
Tricot
 
G
Anaissie
 
E
, et al. 
Thalidomide and hematopoietic-cell transplantation for multiple myeloma.
N Engl J Med
2006
, vol. 
354
 
10
(pg. 
1021
-
1030
)
47
Lokhorst
 
HM
van der Holt
 
B
Zweegman
 
S
, et al. 
A randomized phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma.
Blood
2010
, vol. 
115
 
6
(pg. 
1113
-
1120
)
48
Owen
 
RG
Jackson
 
GH
, et al. 
MRC Myeloma IX: preliminary results from the intensive pathway study [abstract].
Clin Lymphoma Myeloma
2009
, vol. 
9
 
1
 
Abstract 546
49
Richardson
 
PG
Xie
 
W
, et al. 
Single-agent bortezomib in previously untreated multiple myeloma: efficacy, characterization of peripheral neuropathy, and molecular correlations with response and neuropathy.
J Clin Oncol
2009
, vol. 
27
 
21
(pg. 
3518
-
3525
)
50
Jagannath
 
S
Durie
 
BG
Wolf
 
J
, et al. 
Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma.
Br J Haematol
2005
, vol. 
129
 
6
(pg. 
776
-
783
)
51
Harousseau
 
JL
Attal
 
M
Leleu
 
X
, et al. 
Bortezomib plus dexamethasone as induction treatment prior to autologous stem cell transplantation in patients with newly diagnosed multiple myeloma: results of an IFM phase II study.
Haematologica
2006
, vol. 
91
 
11
(pg. 
1498
-
1505
)
52
Rosinol
 
L
Oriol
 
A
Mateos
 
MV
, et al. 
Phase II PETHEMA trial of alternating bortezomib and dexamethasone as induction regimen before autologous stem-cell transplantation in younger patients with multiple myeloma: efficacy and clinical implications of tumor response kinetics.
J Clin Oncol
2007
, vol. 
25
 
28
(pg. 
4452
-
4458
)
53
Harousseau
 
J-L
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
, vol. 
28
 
30
(pg. 
4621
-
4629
)
54
Popat
 
R
Oakervee
 
HE
Hallam
 
S
, et al. 
Bortezomib, doxorubicin and dexamethasone (PAD) front-line treatment of multiple myeloma: updated results after long-term follow-up.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
512
-
516
)
55
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 and maintenance with bortezomib or thalidomide in patients with newly diagnosed multiple myeloma [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 40
56
Belch
 
A
Reece
 
DE
Bahlis
 
NJ
, et al. 
Bortezomib [VELCADE], pegylated liposomal doxorubicin [DOXIL/CAELYX] and dexamethasone in the treatment of previously untreated multiple myeloma patients: impact on quality-of-life [abstract].
Blood
2007
, vol. 
110
 
11
 
Abstract 1058
57
Jakubowiak
 
AJ
Kendall
 
T
Al-Zoubi
 
A
, et al. 
Phase II trial of combination therapy with bortezomib, pegylated liposomal doxorubicin, and dexamethasone in patients with newly diagnosed myeloma.
J Clin Oncol
2009
, vol. 
27
 
30
(pg. 
5015
-
5022
)
58
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
)
59
Einsele
 
H
Liebisch
 
P
Langer
 
C
, et al. 
Velcade, intravenous cyclophosphamide and dexamethasone (VCD) induction for previously untreated multiple myeloma (German DSMM XIa Trial) [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 131
60
Wang
 
M
Giralt
 
S
Delasalle
 
K
Handy
 
B
Alexanian
 
R
Bortezomib in combination with thalidomide-dexamethasone for previously untreated multiple myeloma.
Hematology
2007
, vol. 
12
 
3
(pg. 
235
-
239
)
61
Cavo
 
M
Tacchetti
 
P
Patriarca
 
F
, et al. 
Bortezomib, thalidomide and dexamethasone compared with thalidomide and dexamethasone as induction before and consolidation therapy after double autologous stem cell transplantation in newly diagnosed multiple myeloma: results from a randomized phase III study.
Lancet
2010
, vol. 
379
 
9758
(pg. 
2075
-
2085
)
62
Rosiñol
 
L
Cibeira
 
MT
Mateos
 
MV
, et al. 
A phase III PETHEMA/GEM study of induction therapy prior autologous stem cell transplantation in multiple myeloma: superiority of VTD (bortezomib/thalidomide/dexamethasone) over TD and VBMCP/VBAD plus bortezomib [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 307
63
Harousseau
 
J-L
Avet Loiseau
 
H
Facon
 
T
, et al. 
Bortezomib plus dexamethasone (VD) versus reduced-dose bortezomib plus thalidomide plus dexamethasone (vTD) as induction treatment prior to autologous stem-cell transplantation (ASCT) in newly diagnosed multiple myeloma (MM) [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 354
64
Kaufman
 
JL
Nooka
 
A
Vrana
 
M
, et al. 
Bortezomib, thalidomide, and dexamethasone as induction therapy for patients with symptomatic multiple myeloma.
Cancer
2010
, vol. 
116
 
13
(pg. 
3143
-
3151
)
65
Bensinger
 
W
Jagannath
 
S
Vescio
 
R
, et al. 
Phase 2 study of two sequential three-drug combinations containing bortezomib, cyclophosphamide and dexamethasone, followed by bortezomib, thalidomide and dexamethasone as frontline therapy for multiple myeloma.
Br J Haematol
2010
, vol. 
148
 
4
(pg. 
562
-
568
)
66
Ludwig
 
H
Greil
 
R
Masszi
 
T
, et al. 
Bortezomib, thalidomide, and dexamethasone (VTD) versus VTD plus cyclophosphamide as induction therapy in previously untreated multiple myeloma patients eligible for HDT-ASCT: a randomized phase 2 trial [abstract].
Blood
2009
, vol. 
114
  
22
 
Abstract 2312
67
Pineda-Roman
 
M
Zangari
 
M
Haessler
 
J
, et al. 
Sustained complete remissions in multiple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2.
Br J Haematol
2008
, vol. 
140
 
6
(pg. 
625
-
634
)
68
Wang
 
M
Delasalle
 
K
Giralt
 
S
Alexanian
 
R
Rapid control of previously untreated multiple myeloma with bortezomib-lenalidomide-dexamethasone (BLD).
Hematology
2010
, vol. 
15
 
2
(pg. 
70
-
73
)
69
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
)
70
Roussel
 
M
Avet-Loiseau
 
H
Moreau
 
P
, et al. 
Frontline therapy with bortezomib, lenalidomide and dexamethasone (VRD) followed by autologous stem cell transplantation, VRD consolidation and lenalidomide maintenance in newly diagnosed multiple myeloma patients: primary results of the IFM 2008 phase II study [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 624
71
Kumar
 
S
Flinn
 
I
Noga
 
SJ
, et al. 
Bortezomib, dexamethasone, cyclophosphamide and lenalidomide combination for newly diagnosed multiple myeloma: phase I results from the multicenter EVOLUTION study.
Leukemia
2010
, vol. 
24
 
7
(pg. 
1350
-
1356
)
72
Kumar
 
S
Flinn
 
IW
Richardson
 
PG
, et al. 
Novel three- and four-drug combination regimens of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide, for previously untreated multiple myeloma: results from the multicenter, randomized, phase 2 EVOLUTION study [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 621
73
Jakuboviak
 
AJ
Reece
 
DE
Craig
 
C
, et al. 
Lenalidomide, bortezomib, pegylated liposomal doxorubicin and dexamethasone in newly diagnosed multiple myeloma: updated results of a phase I/II MMRC trial [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 132
74
Fonseca
 
R
Bergsagel
 
PL
Drach
 
J
, et al. 
International Myeloma Working Group molecular classification of multiple myeloma: spotlight review.
Leukemia
2009
, vol. 
23
 
12
(pg. 
2210
-
2221
)
75
Stewart
 
AK
Bergsagel
 
PL
Greipp
 
PR
, et al. 
A practical guide to defining high-risk myeloma for clinical trials, patient counseling and choice of therapy.
Leukemia
2007
, vol. 
21
 
3
(pg. 
529
-
534
)
76
Gertz
 
MA
Lacy
 
MQ
Dispenzieri
 
A
, et al. 
Clinical implications of t(11;14) (q13;q32), t(4;14) (p16.3;q32) and −17p13 in myeloma patients treated with high-dose therapy.
Blood
2005
, vol. 
106
 
8
(pg. 
2837
-
2840
)
77
Cavo
 
M
Terragna
 
C
Renzulli
 
M
, et al. 
Poor outcome with front-line autologous transplantation in t(4;14) multiple myeloma: low complete remission rate and short duration of remission.
J Clin Oncol
2006
, vol. 
24
 
3
(pg. 
e4
-
e5
)
78
Avet Loiseau
 
H
Leleu
 
X
Roussel
 
M
, et al. 
Bortezomib plus dexamethasone induction improves outcome of patients with t(4;14) but not outcome of patients with del(17p).
J Clin Oncol
2010
, vol. 
28
 
30
(pg. 
4630
-
4634
)
79
Zamagni
 
E
Testoni
 
N
Terragna
 
C
, et al. 
Prognostic impact of cytogenetic abnormalities on outcomes of newly diagnosed multiple myeloma patients treated with thalidomide-dexamethasone incorporated into double autologous stem-cell transplantation: an analysis of 593 patients [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 3562
80
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
-
3294
)
81
Morgan
 
GJ
Davies
 
FE
Gregory
 
WM
, et al. 
Thalidomide maintenance significantly improves progression-free survival (PFS) and overall survival (OS) of myeloma patients when effective relapse treatments are used: MRC Myeloma IX results [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 623
82
Avet Loiseau
 
H
Soulier
 
J
Fermand
 
JP
, et al. 
Impact of high-risk cytogenetics and prior therapy on outcomes in patients with advanced relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone.
Leukemia
2010
, vol. 
24
 
3
(pg. 
623
-
628
)
83
Reece
 
D
Song
 
KW
Fu
 
T
, et al. 
Influence of cytogenetics in patients with relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone: adverse effect of deletion 17p13.
Blood
2009
, vol. 
114
 
3
(pg. 
522
-
525
)
84
Kapoor
 
P
Kumar
 
S
Fonseca
 
R
, et al. 
Impact of risk stratification on outcome among patients with multiple myeloma receiving initial therapy with lenalidomide and dexamethasone.
Blood
2009
, vol. 
114
 
3
(pg. 
518
-
521
)
85
Dimopoulos
 
MA
Terpos
 
E
Chanan-Khan
 
A
, et al. 
Renal impairment in patients with multiple myeloma: a consensus statement on behalf of the International Myeloma Working Group.
J Clin Oncol
2010
, vol. 
28
 
33
(pg. 
4976
-
4984
)
86
Tosi
 
P
Zamagni
 
E
Tacchetti
 
P
, et al. 
Thalidomide-dexamethasone as induction therapy prior to autologous stem-cell transplantation in patients with newly diagnosed multiple myeloma and renal insufficiency.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
8
(pg. 
1115
-
1121
)
87
San-Miguel
 
JF
Richardson
 
PG
Sonneveld
 
P
, et al. 
Efficacy and safety of bortezomib in patients with renal impairment: results from the APEX phase 3 study.
Leukemia
2008
, vol. 
22
 
4
(pg. 
842
-
849
)
88
Ghobrial
 
IM
Rajkumar
 
SV
Management of thalidomide toxicity.
J Support Oncol
2003
, vol. 
1
 
3
(pg. 
194
-
205
)
89
Plasmati
 
R
Pastorelli
 
F
Cavo
 
M
, et al. 
Neuropathy in multiple myeloma treated with thalidomide: a prospective study.
Neurology
2007
, vol. 
69
 
6
(pg. 
573
-
581
)
90
Tosi
 
P
Zamagni
 
E
Cellini
 
C
, et al. 
Neurological toxicity of long-term (>1 yr) thalidomide therapy in patients with multiple myeloma.
Eur J Haematol
2005
, vol. 
74
 
3
(pg. 
212
-
216
)
91
Mateos
 
M-V
Management of treatment-related adverse events in patients with multiple myeloma.
Cancer Treat Rev
2010
, vol. 
36
 
suppl 2
(pg. 
524
-
532
)
92
Palumbo
 
A
Rajkumar
 
SV
Dimopoulos
 
MA
, et al. 
Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma.
Leukemia
2008
, vol. 
22
 
2
(pg. 
414
-
423
)
93
Zamagni
 
E
Brioli
 
A
Tacchetti
 
P
Zannetti
 
B
Pantani
 
L
Cavo
 
M
Multiple myeloma, venous thromboembolism and treatment related risk of thrombosis.
Semin Thromb Hemost
2011
, vol. 
37
 
3
(pg. 
209
-
219
)
94
Kumar
 
S
Giralt
 
S
Stadtmauer
 
EA
, et al. 
Mobilization in myeloma revisited: IMWG consensus perspectives on stem cell collection following initial therapy with thalidomide-, lenalidomide-, or bortezomib-containing regimens.
Blood
2009
, vol. 
114
 
9
(pg. 
1729
-
1735
)
95
Argyriou
 
AA
Iconomou
 
G
Kalofonos
 
HP
Bortezomib-induced peripheral neuropathy in multiple myeloma: a comprehensive review of the literature.
Blood
2008
, vol. 
112
 
5
(pg. 
1593
-
1599
)
96
Reeder
 
CB
Reece
 
DE
Kukreti
 
V
, et al. 
Once- versus twice-weekly bortezomib induction therapy with CyBorD in newly diagnosed multiple myeloma.
Blood
2010
, vol. 
115
 
16
(pg. 
3416
-
3417
)
97
Bringhen
 
S
Larocca
 
A
Rossi
 
D
, et al. 
Efficacy and safety of once-weekly bortezomib in multiple myeloma patients.
Blood
2010
, vol. 
116
 
23
(pg. 
4745
-
4753
)
98
Mateos
 
MV
Oriol
 
A
Martinez-Lopez
 
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 randomized trial.
Lancet Oncol
2010
, vol. 
11
 
10
(pg. 
934
-
941
)
99
Delforge
 
M
Bladé
 
J
Dinopoulos
 
MA
, et al. 
Treatment-related peripheral neuropathy in multiple myeloma: the challenge continues.
Lancet Oncol
2010
, vol. 
11
 
11
(pg. 
1086
-
1095
)
100
Chanan-Khan
 
A
Sonneveld
 
P
Schuster
 
MW
, et al. 
Analysis of herpes zoster events among bortezomib-treated patients in the phase III APEX study.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4784
-
4790
)
101
Vickrey
 
E
Allen
 
S
Mehta
 
J
Singhal
 
S
Acyclovir to prevent reactivation of varicella zoster virus (herpes zoster) in multiple myeloma patients receiving bortezomib therapy.
Cancer
2009
, vol. 
115
 
1
(pg. 
229
-
232
)
102
Mellqvist
 
U-H
Westin
 
J
Gimsing
 
P
, et al. 
Improved response rate with bortezomib consolidation after high dose melphalan: first results of a Nordic Myeloma Study Group randomized phase III trial [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 530
103
Attal
 
M
Harousseau
 
JL
Marit
 
G
Caillot
 
D
Stoppa
 
AM
Benboubker
 
L
Lenalidomide after autologous transplantation for myeloma: first analysis of a prospective, randomized study of the Intergroupe Francophone Du Myelome (IFM 2005 02) [abstract].
Blood
2009
, vol. 
114
 
22
 
Abstract 529
104
Cavo
 
M
Perrone
 
G
Buttignol
 
S
, et al. 
Bortezomib-thalidomide-dexamethasone compared with thalidomide-dexamethasone as induction and consolidation therapy before and after double autologous transplantation in newly diagnosed multiple myeloma: results from a randomized phase III study [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 42
105
Terragna
 
C
Zamagni
 
E
Petrucci
 
MT
, et al. 
Molecular remission after bortezomib-thalidomide-dexamethasone compared with thalidomide-dexamethasone as consolidation therapy after double autologous transplantation for multiple myeloma: results of a qualitative and quantitative analysis [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 861
106
Mihelic
 
R
Kaufman
 
JL
Lonial
 
S
Maintenance therapy in multiple myeloma.
Leukemia
2007
, vol. 
21
 
6
(pg. 
1150
-
1157
)
107
Spencer
 
A
Prince
 
HM
Roberts
 
AW
, et al. 
Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure.
J Clin Oncol
2009
, vol. 
27
 (pg. 
1788
-
1793
)
108
Stewart
 
AK
Trudel
 
S
Bahlis
 
NJ
, et al. 
A randomized phase III trial of thalidomide and prednisone as maintenance therapy following autologous stem cell transplantation in patients with multiple myeloma: the NCIC CTG MY.10 trial [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 39
109
Cavo
 
M
Pantani
 
L
Tacchetti
 
P
, et al. 
Thalidomide maintenance in multiple myeloma: certainties and controversies.
J Clin Oncol
2009
, vol. 
27
 
32
(pg. 
e186
-
e187
)
110
McCarthy
 
PL
Owzar
 
K
Anderson
 
KC
, et al. 
Phase III intergroup study of lenalidomide versus placebo maintenance therapy following single autologous hematopoietic stem cell transplantation for multiple myeloma: CALGB 100104 [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 37
111
Attal
 
M
Lauwers
 
WC
Marit
 
G
, et al. 
Maintenance treatment with lenalidomide after transplantation for myeloma: final analysis of the IFM 2005-02 [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 310
112
Moreau
 
P
Pylypenko
 
HV
Grosicki
 
S
, et al. 
A phase 3 prospective randomized international study (MMY-3021) comparing subcutaneous and intravenous administration of bortezomib in patients with relapsed multiple myeloma [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 312
Sign in via your Institution