Autologous stem cell transplantation (ASCT) is considered the gold standard in the frontline therapy of younger patients with multiple myeloma because it results in higher complete remission (CR) rates and longer event-free survival than conventional chemotherapy. The greatest benefit from ASCT is obtained in patients achieving CR after transplantation, the likelihood of CR being associated with the M-protein size at the time of transplantation. The incorporation of novel agents results in higher pre- and posttransplantation CR rates. Induction with bortezomib-containing regimens is encouraging in patients with poor-risk cytogenetics. However, longer follow-up is required to assess the impact of this increased CR on long-term survival. The results of posttransplantation consolidation/maintenance with new drugs are encouraging. All this indicates that, in the era of novel agents, high-dose therapy should be optimized rather than replaced. Because of its high transplantation-related mortality, myeloablative allografting has been generally replaced by reduced-intensity conditioning (reduced intensity conditioning allogeneic transplantation). The best results are achieved after a debulky ASCT, with a progression-free survival plateau of 25% to 30% beyond 6 years from reduced intensity conditioning allogeneic transplantation. The development of novel reduced-intensity preparative regimens and peri- and posttransplantation strategies aimed at minimizing graft-versus-host disease, and enhancing the graft-versus-myeloma effect are key issues.

The outcome of patients with multiple myeloma (MM) treated with conventional chemotherapy is unsatisfactory.1-3  A significant survival improvement has been observed for patients diagnosed in the more recent years.4-6  Because the strongest survival increase was noted in patients younger than 60 years, the improvement was attributed, at least in part, to the benefit of high-dose therapy/stem cell transplantation (HDT/SCT). In addition, the long-term results of autologous and allogeneic transplantation show that a number of patients enjoy prolonged progression-free survival (PFS), and a small proportion of them can be cured.7-11  Indeed, MM is the most frequent indication for HDT/SCT in Europe and the United States.7,12  In recent years, the availability of new effective drugs, such as thalidomide, lenalidomide, and bortezomib, as well as the increased experience with the so-called dose-reduced intensity conditioning allogeneic transplantation (Allo-RIC), has resulted in a new scenario in which the role of HDT/SCT needs to be revisited. This review is focused on: (1) the impact of single and tandem autologous transplantation (ASCT) in the outcome of MM patients, (2) the results achieved with allogeneic transplantation (ie, myeloablative and Allo-RIC), and (3) the prospects for improvement with the incorporation of the new drugs in transplantation programs.

Refractory and relapsed disease

The first studies on HDT/SCT in MM were performed in patients with advanced refractory disease. Although the response rate was encouraging, the median event-free survival (EFS) and overall survival (OS) were short.13,14  Although it is clear that refractory and relapsed myeloma patients are not the ideal candidates for autotransplantation, the aforementioned seminal studies showed: (1) the feasibility of the procedure, (2) the high antimyeloma activity, although generally transient, of HDT, and (3) that, even in a poor-risk population, up to one-fourth of the patients achieving complete remission (CR) remained in CR 10 years beyond ASCT.15 

ASCT should be considered whenever possible in MM patients with sensitive relapse. Indeed, in a randomized trial designed to assess the optimal timing of ASCT, survival of patients who underwent a rescue transplantation was identical to that of those receiving ASCT up-front.16  However because of a longer time without symptoms, toxicity, and treatment, the authors recommended performing transplantation up-front.16 

Patients with primarily refractory disease seem to benefit from early ASCT (Table 1).17-21  However, for a meaningful interpretation of the data, the 2 categories of patients considered as primary refractory (ie, primary unresponsive with progressive disease vs minimal response or with no change but without clinical progression, nonresponsive/nonprogressive) should had been analyzed separately. Thus, in the Spanish Programa de Estudio y Tratamiento de las Hemopatías Malignas (PETHEMA) experience, the median survival of 31 patients with primary unresponsive progressive disease who underwent an ASCT was only 21 months.22 

Table 1

ASCT in primary refractory multiple myeloma

ReferenceNo. of patientsAge, yβ2M, mg/LCR, %Median EFS, yMedian OS, y
Alexanian et al,17  (1994) 27 45 2.8 3.5 
Vesole et al18  (1994) 72 50 — 15 1.7 
Singhal et al19  (2002) 43 54 3.3 40 — 
Kumar et al20  (2004) 50 56 2.7 20 2.5 
Alexanian et al21  (2004) 89 52 3.7 16 7* 7* 
ReferenceNo. of patientsAge, yβ2M, mg/LCR, %Median EFS, yMedian OS, y
Alexanian et al,17  (1994) 27 45 2.8 3.5 
Vesole et al18  (1994) 72 50 — 15 1.7 
Singhal et al19  (2002) 43 54 3.3 40 — 
Kumar et al20  (2004) 50 56 2.7 20 2.5 
Alexanian et al21  (2004) 89 52 3.7 16 7* 7* 

ASCT indicates autologous stem cell transplantation; β2M, β2-microglobulin; CR, complete remission; EFS, event-free survival; OS, overall survival; and —, not applicable.

*

In patients achieving CR after ASCT.

Up-front therapy

Single ASCT versus conventional chemotherapy.

Autologous transplantation is considered the gold standard as part of the initial therapy for patients with MM younger than 65 years. However, the results of trials comparing a single autologous transplantation versus conventional chemotherapy have not been uniform across the studies. Five trials comparing ASCT versus conventional chemotherapy have been published (Table 2).23-27  Two of them showed that ASCT significantly increased the CR rate, EFS, and OS.23,24  In contrast, the remaining 3 studies showed no benefit of ASCT in EFS and OS.25-27  Several reasons may have accounted for the discrepancies among these trials. First, in the Spanish PETHEMA trial,25  only patients with chemosensitive disease were randomized and the French MAG study26  included only patients 55 to 65 years of age. Whether randomization at diagnosis in the PETHEMA study or the inclusion of patients younger than 55 years in the MAG trial could have ended up with different results is uncertain. Second, dose intensity in the conventional arms of the PETHEMA (vincristine, bischloroethylnitrosourea [BCNU], melphalan, cytoxan, prednisone [VBMCP]/vincristine, BCNU, adriamycin, dexamethasone [VBAD])25  and in the US Intergroup (VBMCP)27  studies was higher than in the Intergroup Franchophone du Myelome (IFM) and Medical Research Council trials.23,24  A systematic review and meta-analysis of randomized trials, including a total of 2411 patients, of ASCT versus conventional chemotherapy showed a significantly longer PFS in favor of ASCT, with no significant impact on OS.12 

Table 2

Randomized trials: single ASCT versus conventional chemotherapy

ReferenceCR, %Median PFS, moMedian OS, mo
Attal et al23  (1996) 22 vs 5 28 vs 18 57 vs 42 
Child et al24  (2003) 44 vs 9 32 vs 20 55 vs 42 
Bladé et al25  (2005) 30 vs 11 42 vs 34 67 vs 65 
Fermand et al26  (2005) 8.5 vs 7 25 vs 19 47.8 vs 47.6 
Barlogie et al27  (2006) 17 vs 15 25 vs 21 58 vs 53 
ReferenceCR, %Median PFS, moMedian OS, mo
Attal et al23  (1996) 22 vs 5 28 vs 18 57 vs 42 
Child et al24  (2003) 44 vs 9 32 vs 20 55 vs 42 
Bladé et al25  (2005) 30 vs 11 42 vs 34 67 vs 65 
Fermand et al26  (2005) 8.5 vs 7 25 vs 19 47.8 vs 47.6 
Barlogie et al27  (2006) 17 vs 15 25 vs 21 58 vs 53 

ASCT indicates autologous stem cell transplantation; CR, complete remission; EFS, event-free survival; and OS, overall survival.

Single versus double (tandem) ASCT.

The results of 2 trials comparing the efficacy of single versus double ASCT have been published.28,29  The IFM group reported that the median survival was prolonged by 10 months with tandem transplantation and that the OS at 7 years of follow-up for tandem and single ASCT was 42% and 21%, respectively.28  A non–preplanned subset analysis showed that the patients who benefited from a second ASCT were only those failing to achieve at least a very good partial response (VGPR) with the first transplantation. In this subgroup, survival at 7 years was 43% and 11% with tandem and single transplantation, respectively. The Italian Bologna 96 study showed a significant prolongation in EFS with no impact on OS with tandem versus single ASCT.29  A non–preplanned subset analysis of patients not achieving CR or near-CR also showed a significantly longer EFS with double ASCT that did not translate into a significant OS prolongation.29  In addition, neither of these 2 studies was adequately powered to evaluate the equivalence of 1 versus 2 transplantations in patients achieving at least a VGPR after the first transplantation. Finally, whether or not patients who do not achieve at least a VGPR with a first ASCT benefit from a second high-dose procedure should be answered in a clinical trial.

Impact of CR after ASCT

Whether ASCT is beneficial for the majority of MM patients or the benefit comes from certain subsets of patients remains an unsolved issue. It seems that CR achievement is the crucial step for a long-lasting response and prolonged survival. Thus, it has been shown that patients who achieve immunofixation (IFE)–negative CR after ASCT had an EFS and OS significantly longer than those who remained in PR.30,31  In a Spanish PETHEMA trial, Lahuerta et al have shown that the improvement in the depth of response, particularly the achievement of posttransplantation CR, was associated with a significantly longer EFS and OS.32  In a literature review and meta-analysis, the achievement of CR highly correlated with PFS and long-term survival.33  The Spanish group has also shown that the achievement of a negative minimal residual disease by multiparameter flow cytometry (MFC) is a strongest predictor of EFS and OS compared with IFE-negative CR.34  Furthermore, the Italian group has reported that 18% of patients in at least VGPR after ASCT achieved molecular remission by qualitative and quantitative polymerase chain reaction with intensification therapy using bortezomib/thalidomide/dexamethasone (VTD).35  After a median follow-up of 27 months, no patient in molecular remission had relapsed.35  These 2 studies indicate the importance of achieving the lowest possible tumor mass and support the need for more refined/sensitive CR criteria for MM, including not only negative IFE but also MFC and molecular complete remissions. It is probable that most of the long survivors in continued CR in both the Arkansas study with tandem ASCT (Total Therapy I)10  and in our single ASCT series11  enjoy the aforementioned really “stringent” CR and are not only “operational” but also true cures.

The sensitivity to the initial therapy measured by the M-protein size at the time of transplantation is the most important predictor of CR after ASCT.30,36,37  Thus, in patients with an M-protein less than 10 g/L, the likelihood of CR is between 52% and 67%; whereas in those with a serum M-protein higher than 10 g/L or 20 g/L, the probability of CR is 15% and 7%, respectively.31,36  The Mayo Clinic group reported that the M-spike at the time of transplantation was the only predictor for CR and developed a single function to predict the probability of achieving CR with ASCT.37  In a meta-analysis, a strong association between maximal response to induction therapy and long-term survival was found.33 

Rationale

Conventional induction regimens followed by a single or double ASCT have resulted in 30% to 40% IFE-negative CR and a median survival of 6 years in the best circumstances but, unfortunately, with no survival plateau. The introduction of novel drugs (thalidomide, lenalidomide, and bortezomib) has provided the frame for improving the results of the pretransplantation induction therapy.38,39  The higher antimyeloma potency of the new induction regimens should theoretically end up with a higher pretransplantation tumor reduction, resulting in a higher posttransplantation CR rate and, ultimately, an improvement in the long-term survival and potential cures.

Novel induction regimens

During the last years, the combination of thalidomide/dexamethasone (TD) has increasingly replaced VAD40  and has been approved by the US Food and Drug Administration for its use as pretransplantation induction regimen. Although the overall response rate to TD is between 58% and 76%, the CR rate is low.41,42  In addition, TD could not be an optimal regimen for patients with extramedullary disease because of the lack of activity of thalidomide on soft-tissue plasmacytomas.43  Furthermore, of 3 reported studies comparing pretransplantation induction with TD versus VAD,44-46  TD resulted in an improved posttransplantation outcome in only one.46  The incorporation of thalidomide in the Total Therapy II protocol resulted in significantly higher CR rate and EFS with no advantage in OS because of a shorter survival after relapse.47  Of interest, a more recent update shows that the thalidomide arm of the Total Therapy II produced an improvement in both the OS and the response duration in patients with metaphase cytogenetic abnormalities.48 

Two phase 2 trials on the combination of bortezomib/dexamethasone (VD) as induction regimen have shown a pre- and posttransplantation CR rates of 12% and 33%, respectively.49,50  In a trial by the French IFM group, including 482 patients, posttransplantation IFE-negative CR rate and PFS were significantly higher with VD than with VAD.51 

Thalidomide and bortezomib are being used in combination with dexamethasone or anthracyclines, resulting in the so-called triple regimens. The PAD regimen (bortezomib PS 341, adriamycin, dexamethasone) resulted in a pretransplantation overall response of 95% with 24% CR.52  The posttransplantation CR rate after induction with PAD was 43%. The accrual of a large trial from the Dutch-Belgian Hemato-Oncology Cooperative Group comparing PAD versus VAD has recently been completed, but the results are not yet mature.

The M. D. Anderson group first reported the results achieved with VTD in 36 patients.53  The overall response rate after 2 induction cycles was 92% with 19% CR. The posttransplantation response rate was 89% with 31% CR. Cavo et al54  have recently reported that VTD was significantly superior to TD in terms of CR rate both before (21% vs 6%) and after (43% vs 23%) ASCT. The PFS was also significantly longer with VTD. The Spanish PETHEMA group is currently comparing TD versus VTD versus combination chemotherapy with VBMCP/vincristine, BCNU, adriamycin, dexamethasone (4 cycles) plus 2 cycles of bortezomib as pretransplantation induction therapy.55  The preliminary results of this study show that the best regimen is VTD, with a pre- and posttransplantation CR rate of 30% and 49%, respectively. Encouraging results have been reported with RVD (lenalidomide, bortezomib, dexamethasone) in a phase 1/2 trial, including 36% CR/near-CR response rate, even in high-risk groups,56  and a large international transplantation trial using VRD as induction regimen has recently been activated. Finally, the Total Therapy III protocol used at the University of Arkansas with VTD-PACE induction plus tandem ASCT, consolidation with VTD or VRD and maintenance with TD resulted in a CR rate of 56% at 2 years.57  The pre- and posttransplantation CR rates achieved with novel induction regimens are summarized in Table 3.

Table 3

Pre- and post-ASCT CR rate with novel induction regimens

RegimenPre-ASCT, %Post-ASCT, %
Thalidomide/dexamethasone 23-34 
Bortezomib/dexamethasone 12 33 
PAD-1 24 43 
VTD 21-30 43-49 
Total Therapy III — 56 (2 y) 
RegimenPre-ASCT, %Post-ASCT, %
Thalidomide/dexamethasone 23-34 
Bortezomib/dexamethasone 12 33 
PAD-1 24 43 
VTD 21-30 43-49 
Total Therapy III — 56 (2 y) 

ASCT indicates autologous stem cell transplantation; CR, complete remission; PAD, bortezomib PS 341, adriamycin, dexamethasone; VTD, bortezomib/thalidomide/dexamethasone; and —, not applicable.

Will induction with new regimens improve the posttransplantation outcome?

Post-induction IFE-negative CR is higher when new agents are incorporated than with VAD-like regimens, cyclophosphamide/dexamethasone or combination chemotherapy (up to 30% vs ∼ 10%).32,40,42,58  With this higher pretransplantation tumor reduction, a higher posttransplantation CR rate should be expected. Actually, ASCT increases the CR rate in approximately 20% of the patients, irrespective of the regimen used for induction (Table 3).49-55  The real impact of these increased CR rates on the long-term post-ASCT survival requires longer follow-up. Hopefully, the remarkable results of Total Therapy I,10  with 10-year OS of 33% and 7% of patients alive in continued CR after a median follow-up of 12 years (“operational” cures), will be improved. Of interest, induction with bortezomib-containing regimens results in a high CR rate and in an encouraging, at least in the short-term, outcome in patients with high-risk myeloma by overcoming the negative impact of poor cytogenetics.54,55,57  From the currently available data, it seems that a triple regimen, such as VTD or PAD, will result in superior results than a double combination, such as VD or TD. However, we have to wait until the results of the aforementioned large phase 3 studies are mature enough to be certain of whether or not a multidrug induction will produce significantly longer EFS, OS, and superior long-term outcome with a higher rate of “operational” or true cure rate than a gentler new drug approach. Finally, it must be considered that the experience with new agents is still limited, as shown in a recent report demonstrating that bortezomib, in contrast to the general belief, induces canonical nuclear factor-κ B activation.59  This can modify the way we will use proteasome inhibitors in the future.

How can the efficacy of the high-dose regimens be improved?

The gold standard for HDT in MM remains melphalan 200 mg/m2. Attempts with other drug combinations, such as cyclophosphamide, vepeside, and BCNU-carmustine60 ; the trialkylator regimen thiotepa, busulfan, and cyclophosphamide61 ; an increased melphalan dose to 220 mg/m262 ; or the association of melphalan 140 mg/m2 with total body irradiation (TBI)23,25,63  or busulfan64  have not resulted in an improved outcome. It has recently been shown that the administration of ascorbic acid and arsenic trioxide preceding MEL-200 is safe and could enhance the antimyeloma activity of melphalan.65  Interestingly, bortezomib synergizes with chemotherapy resulting from its effects on DNA repair enzymes. Thus, the combination of melphalan/prednisone with bortezomib in elderly patients has resulted in an impressive 30% IFE-negative CR in the nontransplantation setting.66  In a phase 1 trial investigating the bortezomib dose (1-1.6 mg/m2) and sequence (24 hours before or 24 hours after melphalan administration), the combination of MEL-200 along with the aforementioned bortezomib dose and schedule resulted in a PR rate or better of 93% (14 of 15 patients), with a toxicity profile and engraftment kinetics similar to that observed in an historical control receiving MEL-200 alone.67  The IFM group used bortezomib on days −6, −3, 1, and 4 along with MEL-200 on day −2 in 35 patients with high-risk MM, and the results on engraftment and response were encouraging.68  Thus, the door for enhancing the efficacy of MEL-200 with imaginative combinations with new agents is open.

Is there a role for posttransplantation consolidation/maintenance therapy?

Despite many attempts, the role of maintenance in MM remains controversial and none of the investigated treatments has been established. Thalidomide maintenance prolonged OS in 2 transplantation series.69,70  In the study by the IFM, the thalidomide arm was superior in response rate, EFS, and OS.69  Interestingly, the survival benefit was only observed among patients who failed to achieve at least VGPR after ASCT, suggesting that the benefit was the result of a “consolidation” effect leading to a further reduction in tumor mass. The addition of thalidomide in the Total Therapy II program significantly prolonged the EFS47  as well as the CR duration and OS in the subset of patients with metaphase cytogenetic abnormalities48  and compared favorably with Total Therapy I, with single-agent interferon maintenance, in terms of CR duration, EFS, and OS.71  In the Total Therapy III program, including consolidation with VTD or VRD and maintenance with TD, an unprecedented CR rate of 56% at 2 years of initiation of therapy was achieved.57  In a preliminary report, Palumbo et al72  reported a CR rate of 72% with lenalidomide/prednisone consolidation plus lenalidomide maintenance after induction with PAD and tandem ASCT with MEL-100 in patients 65 to 75 years of age. Interestingly, it has been shown, for the first time outside the allogeneic setting, that consolidation with VTD after ASCT can induce durable molecular remissions.35  The encouraging results of the latter studies have led to the design of a number of phase 3 transplantation trials, including consolidation/maintenance based on the novel drugs thalidomide, lenalidomide, and/or bortezomib. Hopefully, these trials will help us to establish the role of consolidation/maintenance in patients with MM.

Do patients who achieve CR with primary therapy benefit from ASCT intensification?

It could be speculated that patients achieving CR with either conventional chemotherapy or a single transplantation are the most likely to obtain long-term benefit, and perhaps cure, with further intensification with ASCT or with a repeated ASCT, respectively. However, the M. D. Anderson group has consistently reported that patients who achieve CR with conventional chemotherapy and who do not receive a transplantation have the same prolonged PFS and OS as those attaining CR after ASCT.31,73  In the same direction, patients who achieve CR or VGPR with a single transplantation do not benefit from a second ASCT.29  On the other hand, a Mayo Clinic study showed that patients who were in CR at the time of ASCT had a similar survival as those achieving CR after ASCT only.74  Thus, whether patients in CR with primary therapy would benefit from ASCT intensification is unknown. With the availability of novel agents yielding high CR rates as primary treatment, the question has become clinically relevant, and only a randomized trial would conclusively answer it. Such a study should ideally include sequential minimal residual disease studies with MFC and molecular analysis to establish from what CR level further treatment is beneficial or not. In this sense, the Arkansas group has recently reported on the importance of not only achieving CR but also sustaining CR by applying a time-dependent statistical methodology to the patients included in Total Therapy I, II, and III.75  These results support the investigation of CR consolidation, especially in high-risk patients, to determine what treatment and for how long beyond CR it is still necessary.

Is there still a role for ASCT in the era of novel agents?

ASCT is an important tool to further decrease the tumor mass after induction therapy. Theoretically, the higher degree of response achieved with the new induction regimens should be the first step toward a higher CR rate after transplantation, the “sine qua non” condition for an improved survival. On the other hand, melphalan has an unquestionable efficacy in MM, and its high-dose administration in the transplantation procedure is an excellent way to optimize its antimyeloma activity. Furthermore, the mechanisms of action of the novel agents are different from that of high-dose melphalan; thus, the 2 treatment steps should be considered complementary. This is in line with the recent concept of the so-called “cancer stem cells.” This is a small population of cells able to self-renew and responsible for the tumor to sustain.76  These cancer stem cells are biologically distinct from the bulk of differentiated cancer cells that characterize the disease. Novel antimyeloma agents, such as bortezomib, efficiently inhibit myeloma cells but appear to have little activity against myeloma stem cells “in vitro.” These novel drugs could indeed produce dramatic responses on the bulk of differentiated plasma cells but can have limited activity against myeloma stem cells responsible for disease persistence and regrowth.76  In this scenario, intensification with high-dose melphalan appears most appropriate and, rather than replaced, ASCT must be further explored in the era of novel agents. To answer this question, a large international trial of induction with VRD followed by randomization to ASCT versus VRD consolidation with ASCT at relapse has just been activated.

Myeloablative conditioning

The allogeneic transplantation has the advantage over the autologous transplantation that the graft does not contain tumor cells and the potential for a graft versus myeloma (GVM) effect.77,78  However, the allogeneic transplantation in MM has 2 major shortcomings: a transplantation-related mortality ranging from 30% to 50% and a high posttransplantation relapse rate.7,8,79,80  Nevertheless, 10% to 20% of patients undergoing an allogeneic transplantation are long-term disease-free, many of them in molecular remission.7,8,79,80  The European Group for Blood and Marrow Transplantation (EBMT) reported a significant decrease in transplantation-related mortality (TRM) over time (ie, 30% between 1994 and 1998 vs 46% in the previous period).8  In a more recent analysis, the TRM with myeloablative allogeneic transplantation during the period 1998 to 2002 was still 37% (Table 4).7  The main attempts to reduce the TRM have been: (1) the use of peripheral blood progenitor cells and (2) T-cell depletion. In the EBMT series, including 770 patients who underwent a myeloablative allograft, the TRM was not different between peripheral blood stem cell (n = 401) and bone marrow (n = 369) recipients.81  Concerning T-cell depletion, the results have been disappointing. In the Dutch-Belgian Hemato-Oncology Cooperative Group, a series of 53 patients in which T cell–depleted allogeneic transplantation was part of the front-line therapy, median survival from transplantation was only 25 months.82  In a series of 66 patients from the Dana-Farber Cancer Institute, who received a T cell–depleted allogeneic graft, the nonrelapse TRM was 35%, with a PFS at 4 years of 23%.78 

Table 4

Myeloablative versus RIC allogeneic transplantation

ProcedureNo. of patientsTRM, %CR rate, %PFS, % at 3 yOS, % at 3 y
Myeloablative 196 37 53.4 18.9 50.8 
RIC 320 24 33.6 34.5 38.1 
    P  .002 < .001 < .001 NS 
ProcedureNo. of patientsTRM, %CR rate, %PFS, % at 3 yOS, % at 3 y
Myeloablative 196 37 53.4 18.9 50.8 
RIC 320 24 33.6 34.5 38.1 
    P  .002 < .001 < .001 NS 

Data are from the European Group for Blood and Marrow Transplantation experience, 1998 to 2002.7 

RIC indicates reduced intensity conditioning; TRM, transplantation-related mortality; CR, complete remission; OS, overall survival; PFS, progression-free survival; and NS, not significant.

The GVM effect of donor lymphocyte infusion (DLI) has led to the use of DLI in the treatment of both persistent disease or relapse after allogeneic transplantation. The GVM effect of DLI is associated with the development of graft-versus-host disease (GVHD)83  as well as with an antibody response to highly expressed myeloma-associated antigens.84  The response rate to DLI ranges from 45% to 61%, with the CR rate of 20% to 30%.83,85  Unfortunately, the proportion of patients in whom the response lasts for more than 1 year is only approximately 20%.83,85 

Myeloablative allogeneic versus autologous transplantation

In a retrospective case-control study by the EBMT, comparing the results of allogeneic and autologous transplantation, the survival was significantly longer in patients who underwent the autologous procedure.86  However, the relapse rate was higher with ASCT and the prospects for a long-term outcome were better with the allogeneic transplantation. Of interest, Corradini et al9  reported that molecular remission was achieved in only 7% of patients undergoing autologous transplantation versus 50% in the allogeneic setting. The same authors highlighted the impact of molecular remission after allogeneic transplantation on the risk of relapse.9  Thus, none of 16 patients with a negative molecular status after allogeneic transplantation had relapsed at 5 years after transplantation, whereas all 13 patients who remained molecular positive relapsed within the 5 years after allogeneic transplantation.

Reduced-intensity conditioning allogeneic transplantation

The Allo-RIC was introduced in an attempt to decrease the transplantation-related toxicity while retaining the beneficial GVM effect.87-93  The conditioning regimens consisted of: (1) fludarabine/melphalan with or without in vivo T-cell depletion with antithymocyte globulin (ATG) or alemtuzumab or (2) low-dose TBI with or without fludarabine. The results of early studies on Allo-RIC can be summarized as follows: (1) a TRM of approximately 20%, (2) an incidence of acute and chronic GVHD approximately 30% and 50%, respectively, (3) a CR rate up to 50%, (4) a negative effect of T-cell depletion with ATG or alemtuzumab, and (5) a low tumor burden at the time of transplantation as the main factor associated with long-term survival.

Tandem autologous/Allo-RIC transplantation

Considering the importance of a low tumor mass at the time of transplantation for the success of Allo-RIC, the use of ASCT to reduce the tumor burden followed by Allo-RIC has been investigated. Kröger et al94  used a conditioning regimen consisting of fludarabine/melphalan/ATG. In this study, the incidence of acute and chronic GVHD was 38% and 40%, respectively, with a TRM of 11% at 100 days. The CR rate was of 73%. In a subsequent study from the same group, including 22 patients who received an unrelated allograft, the incidence of GVHD was almost identical and the CR rate after the allogeneic procedure was 40%.95  Thus, Allo-RIC from unrelated donors is feasible and its results seem comparable with those achieved with HLA-identical siblings. The long-term results of 2 studies of ASCT with MEL-200 followed by Allo-RIC from identical siblings conditioned with the “Seattle approach” of 2 Gy TBI are summarized in Table 5.96,97  Of interest, the development of chronic GVHD was not associated with the achievement of CR or with disease relapse. An encouraging PFS plateau between 25% and 30% beyond 6 years from Allo-RIC was observed in both studies.96,97 

Table 5

Tandem ASCT followed by Allo-RIC

ReferenceNo. of patientsMedian follow-up, yaGVHD (II-IV)/cGVHD, %CR rate, %EFS, moOS at 5 y
Rotta et al96  (2009) 102 6.6 42/74 57 36 NR 
Bruno et al97  (2009) 100 38/74 53 37 NR 
ReferenceNo. of patientsMedian follow-up, yaGVHD (II-IV)/cGVHD, %CR rate, %EFS, moOS at 5 y
Rotta et al96  (2009) 102 6.6 42/74 57 36 NR 
Bruno et al97  (2009) 100 38/74 53 37 NR 

ASCT indicates autologous stem cell transplantation; allo-RIC, reduced-intensity conditioning allogeneic transplantation; aGVHD, acute graft-versus-host disease; cGVHD, chronic graft-versus-host disease; CR, complete remission; EFS, event-free survival; OS, overall survival; and NR, not reached.

Double autologous versus tandem auto/Allo-RIC transplantation

Three studies have been published comparing the efficacy of a tandem double ASCT versus single autograft followed by Allo-RIC in patients with newly diagnosed MM with an available sibling donor.98-100  The TRM ranged from 10% to 16% in the 3 studies. The French IFM study98  reported no benefit in terms of CR, EFS, and OS from Allo-RIC versus a second autologous transplantation. In contrast, the Italian group99  found an increased CR rate and a significant survival advantage in favor of Allo-RIC, along with a survival plateau beyond 4 years from allografting. In the Spanish PETHEMA study,100  there were no significant differences in EFS and OS between the 2 groups. However, the curves of Allo-RIC patients showed an encouraging plateau beyond 3 years of follow-up (Table 6). The different results achieved in these 3 studies can be explained through the differences in the inclusion criteria and conditioning regimens (Table 7).

Table 6

Double ASCT versus ASCT/Allo-RIC

ReferenceNo. of patientsCR rate, %EFS, moOS, mo
Garban et al98  (2006) 166 vs 51 32.5 vs 32.6 31.7 vs 35 42.7 vs 35 
    P  NS NS .07 
Bruno et al99  (2007) 82 vs 80 26 vs 55 29 vs 35 54 vs 80 
    P  .004 .02 .01 
Rosiñol et al100  (2008) 85 vs 25 11 vs 40%* 26 vs 19.6 58 vs NR 
    P  .01 NS NS 
ReferenceNo. of patientsCR rate, %EFS, moOS, mo
Garban et al98  (2006) 166 vs 51 32.5 vs 32.6 31.7 vs 35 42.7 vs 35 
    P  NS NS .07 
Bruno et al99  (2007) 82 vs 80 26 vs 55 29 vs 35 54 vs 80 
    P  .004 .02 .01 
Rosiñol et al100  (2008) 85 vs 25 11 vs 40%* 26 vs 19.6 58 vs NR 
    P  .01 NS NS 

ASCT indicates autologous stem cell transplantation; allo-RIC, reduced-intensity conditioning allogeneic transplantation; CR, complete remission; EFS, event-free survival; OS, overall survival; NS, not significant; and NR, not reached.

*

Response improvement with second transplantation.

Table 7

Double ASCT versus ASCT/Allo-RIC: differences in study design

ReferenceInclusion criteriaConditioning
Garban et al98  (2006) High-risk (high β2M, 13q−) Fludarabine/busulfan/ATG 
Bruno et al99  (2007) All patients TBI (2 Gy) 
Rosiñol et al100  (2008) No CR/nCR with first ASCT Fludarabine/ MEL-140 
ReferenceInclusion criteriaConditioning
Garban et al98  (2006) High-risk (high β2M, 13q−) Fludarabine/busulfan/ATG 
Bruno et al99  (2007) All patients TBI (2 Gy) 
Rosiñol et al100  (2008) No CR/nCR with first ASCT Fludarabine/ MEL-140 

ASCT indicates autologous stem cell transplantation; allo-RIC, reduced-intensity conditioning allogeneic transplantation; TBI, total body irradiation; ATG, antithymocyte globulin; CR, complete remission; and nCR, near complete remission.

Myeloablative versus Allo-RIC transplantation

The high TRM associated with conventional conditioning allogeneic transplantation has resulted in an almost universal switch from conventional to Allo-RIC. The EBMT group has reported a retrospective study comparing the results achieved in 196 patients who received a myeloablative conditioning versus those of 320 patients who underwent an Allo-RIC allografted between 1998 and 2002 (Table 4).7  Although the CR rate and PFS were favorable to the conventional conditioning, the OS was not significantly different: the higher TRM with conventional conditioning was compensated by a lower relapse rate. The main shortcoming of this study is that the 2 populations were not entirely comparable because the patients in the Allo-RIC group were older (median, 51 vs 45 years), had more resistant disease, and had been more heavily pretreated. In addition, there was an increased use of T-cell depletion in the Allo-RIC group, which was associated with a lower CR rate and an increased relapse rate.

Which is the best allogeneic transplantation approach?

The TRM of approximately 20% higher with myeloablative conditioning has resulted in a shift to Allo-RIC. However, the final outcome with the 2 conditioning approaches seems to be similar because the higher TRM with myeloablative conditioning is compensated by a lower relapse rate. Therefore, the only way to answer the question would be a randomized trial. With the current data, Allo-RIC from either related or unrelated donors seems the most promising allogeneic approach, but there may still be a role for myeloablative allografting in selected patients (ie, younger patients with poor cytogenetics).101 

Who are the patients most likely to benefit from Allo-RIC?

Patients with sensitive disease after a debulky autologous transplantation are the most likely to benefit from Allo-RIC. In our opinion, the patients already in CR after either primary therapy or ASCT should not be submitted to the risk of any allogeneic procedure. Patients with sensitive relapse could also benefit from Allo-RIC. However, it is doubtful that patients with advanced disease benefit from Allo-RIC. In any event, taking into account the TRM of 10% to 20%, the chronic GVHD of 50% to 70%, and the controversial results on a meaningful survival plateau, Allo-RIC should only be conducted in controlled clinical trials, including patients with high-risk myeloma for whom other therapies are insufficient.102 

Double ASCT or tandem ASCT/Allo-RIC?

The results of the 3 prospective trials on double autologous versus auto/Allo-RIC are controversial.102  Our results in patients not achieving CR or near-CR with an autologous transplantation are similar to those reported in the Italian study in favor of Allo-RIC. The differences among these studies can be explained through the different study design (Tables 67).98-100  Hopefully, the results of the large prospective studies of the EBMT and the US Bone Marrow Transplant Clinical Trials will help to clarify the role of auto/Allo-RIC.

How can the results of Allo-RIC be improved?

The following points are essential for improving the outcome of patients undergoing Allo-RIC: (1) use a prior debulky autologous transplantation, (2) limit the procedure to patients with sensitive disease, (3) use the best conditioning with fludarabine/melphalan or low-dose TBI with or without fludarabine and with no T-cell depletion, and (4) optimize DLI (ie, with low-dose thalidomide) for suboptimal responses.103  The incorporation of bortezomib in the allogeneic procedure could decrease acute GVHD while retaining the GVM effect,104,105  and trials including bortezomib in the conditioning and after engraftment are ongoing. It has been shown that lenalidomide is highly effective in patients relapsing after allogeneic transplantation.106  Interestingly, lenalidomide increased the CD4+FoxP3+ cells, a specific marker of regulatory T cells.106  The immunostimulatory effect of lenalidomide deserves further investigation in the allogeneic transplantation setting. It is obvious, to actually improve the long-term outcome of patients undergoing Allo-RIC, that there is a need for developing novel reduced-intensity preparative regimens107  as well as peri- and posttransplantation strategies (ie, expansion of T regulatory cells) aimed at minimizing the GVHD and enhancing the GVM effect.108 

This work was supported in part by Instituto Carlos III (Spanish Institute of Health; grants 06/0020/0005 and 08/0147).

Contribution: J.B. wrote the first draft; and L.R., M.T.C., M.R., and E.C. reviewed and provided critical revisions on the article.

Conflict-of-interest disclosure: J.B. received honoraria and grant support from Janssen-Cilag and Celgene; L.R. and M.T.C. received honoraria from Janssen-Cilag and Celgene. The remaining authors declare no competing financial interests.

Correspondence: Joan Bladé, Hematology Department, Villaroel 170, Hospital Clínic, Barcelona, Spain; e-mail: jblade@clinic.ub.es.

1
Kyle
 
RA
Rajkumar
 
SV
Multiple myeloma.
Blood
2008
, vol. 
111
 
1
(pg. 
2962
-
2972
)
2
Bladé
 
J
Rosiñol
 
L
Advances in therapy of multiple myeloma.
Curr Opin Oncol
2008
, vol. 
20
 
6
(pg. 
697
-
704
)
3
Brenner
 
H
Gondos
 
A
Pulte
 
D
Expected long-term survival of patients diagnosed with multiple myeloma in 2006-2010.
Haematologica
2009
, vol. 
94
 
2
(pg. 
270
-
275
)
4
Kristinsson
 
SY
Landgren
 
O
Dickman
 
PW
Derolf
 
AR
Bjorkholm
 
M
Patterns of survival in multiple myeloma: a population-based study of patients diagnosed in Sweden from 1973 to 2003.
J Clin Oncol
2007
, vol. 
25
 
15
(pg. 
1993
-
1999
)
5
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
)
6
Kumar
 
SK
Rajkumar
 
SV
Dispenzieri
 
A
, et al. 
Improved survival in multiple myeloma and impact of novel therapies.
Blood
2008
, vol. 
111
 
5
(pg. 
2516
-
2520
)
7
Crawley
 
C
Iacobelli
 
S
Björkstrand
 
B
, et al. 
Reduced-intensity conditioning for myeloma: lower nonrelapse mortality but higher relapse rates compared with myeloablative conditioning.
Blood
2007
, vol. 
109
 
8
(pg. 
3588
-
3594
)
8
Gahrton
 
G
Svensson
 
H
Cavo
 
M
, et al. 
Progress in allogeneic bone marrow and peripheral blood stem cell transplantation for multiple myeloma: a comparison between transplants performed 1983-93 and 1994-98 at European Group for Blood and Marrow Transplantation centres.
Br J Haematol
2001
, vol. 
113
 
1
(pg. 
209
-
216
)
9
Corradini
 
P
Cavo
 
M
Lokhorst
 
H
, et al. 
Molecular remission after myeloablative allogeneic stem cell transplantation predicts a better relapse-free survival in patients with multiple myeloma.
Blood
2003
, vol. 
102
 
5
(pg. 
1927
-
1929
)
10
Barlogie
 
B
Tricot
 
GJ
Van Rhee
 
F
, et al. 
Long-term outcome results of the first tandem autotransplant trial for multiple myeloma.
Br J Haematol
2006
, vol. 
135
 
2
(pg. 
158
-
164
)
11
Rovira
 
M
Rosiñol
 
L
Fernández-Avilés
 
F
, et al. 
Is there a curative potential of autologous stem cell transplantation in multiple myeloma? Long-term results from a single institution series.
Bone Marrow Transplant
2009
, vol. 
43
 
suppl 1
pg. 
S147
 
12
Koreth
 
J
Cutler
 
SC
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
)
13
Barlogie
 
B
Hall
 
R
Zander
 
A
Dicke
 
KA
Alexanian
 
R
High-dose melphalan with autologous bone marrow transplantation for multiple myeloma.
Blood
1986
, vol. 
67
 
5
(pg. 
1298
-
1301
)
14
Barlogie
 
B
Alexanian
 
R
Dicke
 
KA
, et al. 
High-dose chemoradiotherapy and autologous bone marrow transplantation for resistant multiple myeloma.
Blood
1987
, vol. 
70
 
3
(pg. 
869
-
872
)
15
Barlogie
 
B
Jagannath
 
S
Naucke
 
S
, et al. 
Long-term follow-up after high-dose therapy for high-risk multiple myeloma.
Bone Marrow Transplant
1998
, vol. 
21
 
11
(pg. 
1101
-
1107
)
16
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
)
17
Alexanian
 
R
Dimopoulos
 
MA
Hester
 
J
Delasalle
 
K
Champlin
 
R
Early myeloablative therapy for multiple myeloma.
Blood
1994
, vol. 
84
 
12
(pg. 
4278
-
4282
)
18
Vesole
 
DH
Barlogie
 
B
Jagannath
 
S
, et al. 
High-dose therapy for refractory multiple myeloma: improved prognosis with better supportive care and double transplants.
Blood
1994
, vol. 
84
 
3
(pg. 
950
-
956
)
19
Singhal
 
S
Powles
 
R
Sirohi
 
B
, et al. 
Response to induction chemotherapy is not essential to obtain survival benefit from high-dose melphalan and autotransplantation in myeloma.
Bone Marrow Transplant
2002
, vol. 
30
 
10
(pg. 
673
-
679
)
20
Kumar
 
S
Lacy
 
MQ
Dispenzieri
 
A
, et al. 
High-dose therapy and autologous stem cell transplantation for multiple myeloma poorly responsive to initial therapy.
Bone Marrow Transplant
2004
, vol. 
34
 
2
(pg. 
161
-
167
)
21
Alexanian
 
R
Weber
 
D
Delasalle
 
K
Handy
 
B
Champlin
 
R
Giralt
 
S
Clinical outcome with intensive therapy for patients with primary resistant multiple myeloma.
Bone Marrow Transplant
2004
, vol. 
34
 
3
(pg. 
229
-
234
)
22
Bladé
 
J
Rosiñol
 
L
García-Sanz
 
R
, et al. 
A PETHEMA study of high-dose therapy/stem cell support, including tandem transplant, in primary refractory multiple myeloma (MM): identification of two populations with different outcome.
J Clin Oncol
2007
, vol. 
25
 
suppl 1
pg. 
446a
 
23
Attal
 
M
Harousseau
 
JL
Stoppa
 
AM
, et al. 
A prospective randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma.
N Engl J Med
1996
, vol. 
335
 
2
(pg. 
91
-
97
)
24
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
)
25
Bladé
 
J
Rosiñol
 
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
)
26
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
)
27
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
)
28
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
)
29
Cavo
 
M
Tosi
 
P
Zamagni
 
E
, et al. 
Prospective, randomized study of single compared with double autologous transplantation for multiple myeloma: Bologna 96 clinical study.
J Clin Oncol
2007
, vol. 
25
 
17
(pg. 
2434
-
2441
)
30
Bladé
 
J
Esteve
 
J
Rives
 
S
, et al. 
High-dose therapy autotransplantation/intensification vs continued standard chemotherapy in multiple myeloma in first remission: results of a non-randomized study from a single institution.
Bone Marrow Transplant
2000
, vol. 
26
 
8
(pg. 
845
-
849
)
31
Alexanian
 
R
Weber
 
D
Giralt
 
S
, et al. 
Impact of complete remission with intensive therapy in patients with responsive multiple myeloma.
Bone Marrow Transplant
2001
, vol. 
27
 
10
(pg. 
1037
-
1043
)
32
Lahuerta
 
JJ
Mateos
 
MV
Martínez-López
 
J
, et al. 
Influence of pre- and post-transplantation responses on outcome of patients with multiple myeloma: sequential improvement of response and achievement of complete response are associated with longer survival.
J Clin Oncol
2008
, vol. 
26
 
35
(pg. 
5775
-
5782
)
33
van de Velde
 
HJK
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
)
34
Paiva
 
B
Vidriales
 
MB
Cerveró
 
J
, et al. 
Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation.
Blood
2008
, vol. 
112
 
10
(pg. 
4017
-
4023
)
35
Ladetto
 
M
Pagliano
 
G
Ferrero
 
S
, et al. 
Major tumour shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma.
J Clin Oncol
2010
3
22
 
[Epub ahead of print]
36
Nadal
 
E
Giné
 
E
Bladé
 
J
, et al. 
High-dose therapy/autologous stem cell transplantation in patients with chemosensitive myeloma: predictors of complete remission.
Bone Marrow Transplant
2004
, vol. 
33
 
1
(pg. 
61
-
64
)
37
Dingli
 
D
Pacheco
 
JM
Dispenzieri
 
A
, et al. 
Serum M-spike and transplant outcome in patients with multiple myeloma.
Cancer Sci
2007
, vol. 
98
 
7
(pg. 
1035
-
1040
)
38
Bensinger
 
W
Stem-cell transplantation for multiple myeloma in the era of novel drugs.
J Clin Oncol
2008
, vol. 
26
 
3
(pg. 
480
-
492
)
39
San Miguel
 
J
Harousseau
 
JL
Joshua
 
D
Anderson
 
KC
Individualizing treatment of patients with myeloma in the era of novel agents.
J Clin Oncol
2008
, vol. 
26
 
3
(pg. 
480
-
492
)
40
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. 
108
 
1
(pg. 
35
-
39
)
41
Rajkumar
 
SV
Blood
 
E
Vesole
 
D
, et al. 
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 Group.
J Clin Oncol
2006
, vol. 
24
 
3
(pg. 
431
-
436
)
42
Rajkumar
 
SV
Rosiñol
 
L
Hussein
 
M
, et al. 
A multicenter, randomized, double-blind, placebo-controlled study of thalidomide plus dexamethasone versus dexamethasone as initial therapy for newly diagnosed multiple myeloma.
J Clin Oncol
2008
, vol. 
26
 
13
(pg. 
2171
-
2177
)
43
Rosiñol
 
L
Cibeira
 
MT
Bladé
 
J
, et al. 
Escape of extramedullary disease to the thalidomide effect in multiple myeloma.
Haematologica
2004
, vol. 
89
 
7
(pg. 
832
-
836
)
44
Macro
 
M
Divine
 
M
Uzunhan
 
Y
, et al. 
Dexamethasone plus thalidomide compared with VAD as a pretransplant treatment in newly diagnosed multiple myeloma.
Blood
2006
, vol. 
108
 
11
pg. 
22a
  
Abstract 57
45
Thomas
 
SK
Giralt
 
SA
Wang
 
M
, et al. 
Survival outcomes of patients receiving thalidomide/dexamethasone for previously untreated multiple myeloma.
Blood
2006
, vol. 
108
 
11
pg. 
1019a
  
Abstract 3569
46
Lokhorst
 
HM
Bertsch
 
U
Sonneveld
 
P
, et al. 
Thalidomide in indection treatment increases the very good partial response (VGPR) rate before and after high-dose therapy in patients with previously untreated multiple myeloma.
Haematologica
2008
, vol. 
93
 
1
(pg. 
124
-
127
)
47
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
)
48
Barlogie
 
B
Pineda-Roman
 
M
van Rhee
 
F
, et al. 
Thalidomide arm of Total Therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities.
Blood
2008
, vol. 
112
 
8
(pg. 
3115
-
3121
)
49
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.
Haematologica
2006
, vol. 
91
 
11
(pg. 
1498
-
1505
)
50
Rosiñol
 
L
Oriol
 
A
Mateos
 
MV
, et al. 
A phase II trial of alternating bortezomib and dexamethasone as induction regimen prior to autologous stem cell transplantation in younger patients with multiple myeloma.
J Clin Oncol
2007
, vol. 
25
 
28
(pg. 
4452
-
4458
)
51
Harouseau
 
JL
Avet-Loiseau
 
H
Attal
 
M
, et al. 
High complete and very good partial response rate with bortezomib-dexamethasone as induction prior to ASCT in newly diagnosed patients with high-risk myeloma: results of the IFM-2005-01 phase 3 trial.
Blood
2009
, vol. 
114
 
22
pg. 
149a
  
Abstract 353
52
Popat
 
R
Oakervee
 
HE
Hallam
 
S
, et al. 
Bortezomib, doxorubicin and dexamethasone (PAD) frontline treatment of multiple myeloma: updated results after long-term follow-up.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
512
-
516
)
53
Wang
 
M
Giralt
 
S
Delasalle
 
K
, et al. 
Bortezomib in combination with thalidomide-dexamethasone for previously untreated multiple myeloma.
Hematology
2007
, vol. 
12
 
3
(pg. 
235
-
236
)
54
Cavo
 
M
Tachetti
 
P
Patriarca
 
, et al. 
Superior complete response rate and progression-free survival after autologous transplantation with up-front Velcade-thalidomide-dexamethasone compared with thalidomide-dexamethasone in newly diagnosed multiple myeloma.
Blood
2008
, vol. 
112
 
11
pg. 
65a
  
Abstract 158
55
Rosiñol
 
L
Cibeira
 
MT
Martínez
 
J
, et al. 
Thalidomde/dexamethasone (TD) vs bortezomib-Velcade/thalidomide/dexamethasone vs VBMCP/VBAD/Velcade as induction therapy prior to autologous stem cell transplantation: first results of a phase III trial from the Spanish PETHEMA group.
Blood
2008
, vol. 
112
 
11
pg. 
244a
  
Abstract 654
56
Richardson
 
P
Lonial
 
S
Jakubowiak
 
A
, et al. 
Lenalidomide, bortezomib, and dexamethasone in patients with newly diagnosed multiple myeloma: encouraging efficacy in high risk groups with updated results of a phase I/II study.
Blood
2008
, vol. 
112
 
11
pg. 
41a
  
Abstract 92
57
Barlogie
 
B
Anassie
 
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
)
58
Mellqvist
 
UH
Lenhoff
 
S
Jhonsen
 
HE
, et al. 
Cyclophosphamide and dexamethasone is an efficient initial treatment before high-dose melphalan and autologous stem cell transplantation in patients with newly diagnosed multiple myeloma: results of a randomized comparison with vincristine, doxorubicin, and dexamethasone.
Cancer
2008
, vol. 
112
 
1
(pg. 
129
-
135
)
59
Hideshima
 
T
Ikeda
 
H
Chauban
 
D
, et al. 
Bortezomib induces canonical nuclear factor-κB activation in multiple myeloma cells.
Blood
2009
, vol. 
114
 
5
(pg. 
1046
-
1052
)
60
Ventura
 
GJ
Barlogie
 
B
Hester
 
JP
, et al. 
High dose cyclophosphamide, BCNU and VP-16 with autologous blood stem cell support for refractory multiple myeloma.
Bone Marrow Transplant
1990
, vol. 
5
 
4
(pg. 
265
-
268
)
61
Shimoni
 
A
Smith
 
TL
Aleman
 
A
, et al. 
Thiotepa, busulfan, cyclophosphamide (TBC) and autologous hematopoietic transplantation: an intensive regimen for the treatment of multiple myeloma.
Bone Marrow Transplant
2001
, vol. 
27
 
8
(pg. 
821
-
828
)
62
Moreau
 
P
Milpied
 
N
Mahé
 
B
, et al. 
Melphalan 220 mg/m2 followed by peripheral blood stem cell transplantation in 27 patients with advanced multiple myeloma.
Bone Marrow Transplant
1999
, vol. 
23
 
10
(pg. 
1003
-
1006
)
63
Moreau
 
P
Facon
 
T
Attal
 
M
, et al. 
Comparison of 200 mg/m2 melphalan and 8 Gy total body irradiation plus 140 mg/m2 melphalan as conditioning regimens for peripheral blood stem cell transplantation in patients with newly diagnosed multiple myeloma: final analysis of the Intergroupe Francophone du Myélome 9502 randomized trial.
Blood
2002
, vol. 
99
 
3
(pg. 
731
-
735
)
64
Carreras
 
E
Rosiñol
 
L
Terol
 
MJ
, et al. 
Veno-occlusive disease of the liver after high-dose cytoreductive therapy with busulphan and melphalan for autologous blood stem cell transplantation in multiple myeloma patients.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
12
(pg. 
1448
-
1454
)
65
Qazilbash
 
MH
Saliba
 
RM
Nieto
 
Y
, et al. 
Arsenic trioxide with ascorbic acid and high-dose melphalan: results of a phase II randomized trial.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
12
(pg. 
1401
-
1407
)
66
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
)
67
Lonial
 
S
Kaufman
 
J
Langston
 
AA
, et al. 
A randomized phase I trial of melphalan plus bortezomib as conditioning for autologous transplant for myeloma: the effect of sequence of administration.
Blood
2007
, vol. 
110
 
11
pg. 
288a
  
Abstract 949
68
Roussel
 
M
Moreau
 
P
Huynh
 
A
, et al. 
Bortezomib and high-dose melphalan as conditioning regimen before autologous stem cell transplantation in patients with de novo multiple myeloma: a phase II study of the Intergroup Francophone du Myéloma (IFM).
Blood
2010
, vol. 
115
 
1
(pg. 
32
-
37
)
69
Attal
 
M
Harousseau
 
JL
Leyvraz
 
S
, et al. 
Maintenance therapy with thalidomide improves survival in multiple myeloma patients.
Blood
2006
, vol. 
108
 
10
(pg. 
3289
-
3294
)
70
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
 
11
(pg. 
1788
-
1793
)
71
Zangari
 
M
van Rhee
 
F
Anaissie
 
E
, et al. 
Eight-year median survival in multiple myeloma after total therapy 2: roles of thalidomide and consolidation chemotherapy in the context of total therapy 1.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
433
-
444
)
72
Palumbo
 
A
Falco
 
P
Gay
 
F
, et al. 
Bortezomib-doxorubicin-dexamethasone as induction prior to reduced intensity autologous transplantation followed by lenalidomide as consolidation/maintenance in elderly untreated myeloma patients.
Blood
2008
, vol. 
112
 
11
pg. 
65a
  
Abstract 159
73
Wang
 
M
Delasalle
 
K
Thomas
 
S
Giralt
 
S
Alexanian
 
R
Complete remission represents the major surrogate marker of long survival in multiple myeloma.
Blood
2006
, vol. 
108
 
11
pg. 
123a
  
Abstract 403
74
Dingli
 
D
Pacheco
 
JM
Nowakowski
 
GS
, et al. 
Relationship between depth of response and outcome in multiple myeloma.
J Clin Oncol
2007
, vol. 
25
 
31
(pg. 
4933
-
4937
)
75
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
)
76
Huff
 
CA
Matsui
 
W
Smith
 
BD
Jones
 
RJ
The paradox of response and survival in cancer therapeutics.
Blood
2006
, vol. 
107
 
2
(pg. 
431
-
434
)
77
Tricot
 
G
Vesole
 
DH
Jagannath
 
S
Hilton
 
J
Munshi
 
N
Barlogie
 
B
Graft-versus-myeloma effect: proof of principle.
Blood
1996
, vol. 
87
 
3
(pg. 
1196
-
1198
)
78
Alyea
 
E
Weller
 
E
Schlossman
 
R
, et al. 
Outcome after autologous and allogeneic stem cell transplantation for patients with multiple myeloma: impact of graft-versus-myeloma effect.
Bone Marrow Transplant
2003
, vol. 
32
 
12
(pg. 
1145
-
1151
)
79
Gahrton
 
G
Tura
 
S
Ljungman
 
P
, et al. 
Prognostic factors in allogeneic bone marrow transplantation for multiple myeloma.
J Clin Oncol
1995
, vol. 
13
 (pg. 
1312
-
1322
)
80
Bensinger
 
WI
Buckner
 
CD
Anasetti
 
C
, et al. 
Allogeneic marrow transplantation for multiple myeloma: an analysis of risk factors and outcome.
Blood
1996
, vol. 
88
 
7
(pg. 
2787
-
2793
)
81
Gahrton
 
G
Iacobelli
 
S
Bandini
 
G
, et al. 
Peripheral blood or bone marrow cells in reduced-intensity or myeloablative conditioning allogeneic HLA identical sibling donor transplantation for multiple myeloma.
Haematologica
2007
, vol. 
92
 
11
(pg. 
1513
-
1518
)
82
Lokhorst
 
HM
Segeren
 
CM
Verdonck
 
LF
, et al. 
Partially T-cell depleted allogeneic stem-cell transplantation for first-line treatment of multiple myeloma: a prospective evaluation of patients treated in the phase III study Hovon 24 MM.
J Clin Oncol
2003
, vol. 
21
 
9
(pg. 
1728
-
1733
)
83
Lokhorst
 
HM
Wu
 
K
Verdonck
 
LF
, et al. 
The occurrence of graft-versus-host disease is the major predictive factor for response to donor lymphocyte infusions in multiple myeloma.
Blood
2004
, vol. 
103
 
11
(pg. 
4362
-
4364
)
84
Bellucci
 
R
Wu
 
CJ
Chiaretti
 
S
, et al. 
Complete response to donor lymphocyte infusions in multiple myeloma is associated with antibody responses to highly expressed antigens.
Blood
2004
, vol. 
103
 
2
(pg. 
656
-
663
)
85
Salama
 
M
Nevill
 
T
Marcellus
 
D
, et al. 
Donor lymphocyte infusions for multiple myeloma.
Bone Marrow Transplant
2000
, vol. 
26
 
11
(pg. 
1179
-
1184
)
86
Björkstrand
 
B
Ljungman
 
P
Svensson
 
H
, et al. 
Allogeneic bone marrow transplantation versus autologous stem cell transplantation in multiple myeloma: a retrospective case-matched study from the European Group for Blood and Marrow Transplantation.
Blood
1996
, vol. 
88
 
12
(pg. 
4711
-
4718
)
87
Giralt
 
S
Aleman
 
A
Anagnostopoulos
 
A
, et al. 
Fludarabine/melphalan conditioning for allogeneic transplantation in patients with multiple myeloma.
Bone Marrow Transplant
2002
, vol. 
30
 
6
(pg. 
367
-
373
)
88
Einsele
 
H
Schafer
 
HJ
Hebart
 
H
, et al. 
Follow-up of patients with progressive multiple myeloma undergoing allograft after reduced-intensity conditioning.
Br J Haematol
2003
, vol. 
121
 
3
(pg. 
411
-
418
)
89
Pérez-Simón
 
JA
Martino
 
R
Alegre
 
A
, et al. 
Chronic but not acute graft-versus-host disease improves outcome in multiple myeloma patients after non-myeloablative allogeneic transplantation.
Br J Haematol
2003
, vol. 
121
 
1
(pg. 
104
-
108
)
90
Lee
 
CK
Badros
 
A
Barlogie
 
B
, et al. 
Prognostic factors in allogeneic transplantation for patients with high-risk multiple myeloma alter reduced intensity conditioning.
Exp Hematol
2003
, vol. 
31
 
1
(pg. 
73
-
80
)
91
Peggs
 
KS
MacKinnon
 
S
Williams
 
CD
, et al. 
Reduced-intensity transplantation with in vivo T-cell depletion and adjuvant dose-escalating donor lymphocyte infusions for chemotherapy-sensitive myeloma: limited efficacy of graft-versus-tumor activity.
Biol Blood Marrow Transplant
2003
, vol. 
9
 
4
(pg. 
257
-
265
)
92
Crawley
 
C
Lalancette
 
M
Szydlo
 
R
, et al. 
Outcomes for reduced-intensity allogeneic transplantation for multiple myeloma: an analysis of prognostic factors from the Chronic Leukemia Myeloma Working Party of the EBMT.
Blood
2005
, vol. 
105
 
11
(pg. 
4532
-
4539
)
93
Kröger
 
N
Pérez-Simón
 
JA
Myint
 
H
, et al. 
Relapse to prior autograft and chronic graft-versus-host disease are the strongest prognostic factors for outcome of melphalan/fludarabine-based dose-reduced allogeneic stem cell transplantation in patients with multiple myeloma.
Biol Blood Marrow Transplant
2004
, vol. 
10
 
10
(pg. 
698
-
708
)
94
Kröger
 
N
Schwerdtfeger
 
R
Kiehl
 
M
, et al. 
Autologous stem cell transplantation followed by a dose reduced allograft induces high complete remission rate in multiple myeloma.
Blood
2002
, vol. 
100
 
3
(pg. 
755
-
760
)
95
Kröger
 
N
Cottfried
 
H
Schweerdtfeger
 
R
, et al. 
Unrelated stem cell transplantation in multiple myeloma after reduced intensity conditioning with pre-transplantation antithymocyte globulin is highly effective with low transplantation-related mortality.
Blood
2002
, vol. 
100
 
12
(pg. 
3919
-
3924
)
96
Rotta
 
M
Storer
 
BE
Sahebi
 
F
, et al. 
Long-term outcome of patients with multiple myeloma after autologous hematopoietic cell transplantation and nonmyeloablative allografting.
Blood
2009
, vol. 
113
 
14
(pg. 
3383
-
3391
)
97
Bruno
 
B
Rotta
 
M
Patriarca
 
F
, et al. 
Nonmyeloablative allografting for newly diagnosed multiple myeloma: the experience of the Gruppo Italiano Trapianti di Midollo.
Blood
2009
, vol. 
113
 
14
(pg. 
3375
-
3382
)
98
Garban
 
F
Attal
 
M
Michallet
 
M
, et al. 
Prospective comparison of autologous stem cell transplantation followed by dose-reduced allograft (IFM99-03 trial) with tandem autologous stem cell transplantation (IFM99-04 trial) in high risk de novo multiple myeloma.
Blood
2006
, vol. 
107
 
9
(pg. 
3474
-
3480
)
99
Bruno
 
B
Rotta
 
M
Patriarca
 
F
, et al. 
A comparison of allografting with autograft for newly diagnosed myeloma.
N Engl J Med
2007
, vol. 
356
 
11
(pg. 
1110
-
1120
)
100
Rosiñol
 
L
Pérez-Simón
 
JA
Sureda
 
A
, et al. 
A prospective PETHEMA study of tandem autologous transplantation versus autograft followed by reduced-intensity conditioning allogeneic transplantation in newly diagnosed multiple myeloma.
Blood
2008
, vol. 
112
 
9
(pg. 
3591
-
3593
)
101
Cook
 
G
Bird
 
JM
Marks
 
DI
In pursuit of the allo-immune response in multiple myeloma: where do we go from here?
Bone Marrow Transplant
2009
, vol. 
43
 (pg. 
91
-
99
)
102
Stewart
 
KA
Reduced-intensity allogeneic transplantation for myeloma: reality bites [editorial].
Blood
2009
, vol. 
113
 
14
(pg. 
3135
-
3136
)
103
Kröger
 
N
Shimoni
 
A
Zagrivnaja
 
M
, et al. 
Low-dose thalidomide and donor lymphocyte infusion as adoptive immunotherapy after allogeneic stem cell transplantation in patients with multiple myeloma.
Blood
2004
, vol. 
104
 
10
(pg. 
3361
-
3363
)
104
Blanco
 
B
Pérez-Simón
 
JA
Sánchez-Abarca
 
LI
, et al. 
Bortezomib induces selective depletion of alloreactive T-lymphocytes and decreases the production of Th1 cytokines.
Blood
2006
, vol. 
107
 
9
(pg. 
3575
-
3583
)
105
Kröger
 
N
Zabelina
 
T
Ayuk
 
F
, et al. 
Bortezomib after dose-reduced allogeneic stem cell transplantation for multiple myeloma to enhance or maintain remission status.
Exp Hematol
2006
, vol. 
34
 
6
(pg. 
770
-
775
)
106
Minnema
 
MC
van der Veer
 
MS
Aarts
 
T
Emmelot
 
M
Mutis
 
T
Lokhorst
 
HM
Lenalidomide alone or in combination with dexamethasone is highly effective in patients with relapsed multiple myeloma after allogeneic stem cell transplantation and increases the frequency of CD4+ Foxp3+ T cells.
Leukemia
2009
, vol. 
23
 
3
(pg. 
605
-
607
)
107
Kohrt
 
IE
Turnbull
 
BB
Heydari
 
K
, et al. 
TLI and ATG conditioning with low risk of graft-versus-host disease retains antitumour reactions after allogeneic hematopoietic cell transplantation from related and unrelated donors.
Blood
2009
, vol. 
114
 
5
(pg. 
1099
-
1109
)
108
Pillai
 
AB
George
 
TI
Dult
 
S
Strober
 
S
Host natural killer T-cells induce an IL-4 dependent expansion of donor CD4+ CD25+ Foxp3+ Tregs that protect against graft-versus-host disease.
Blood
2009
, vol. 
113
 
18
(pg. 
4458
-
4467
)
Sign in via your Institution