Abstract
Survival of myeloma patients has greatly improved with the use of autologous stem cell transplantation and novel agents, such as proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies. Compared to bortezomib- and lenalidomide-based regimens alone, the addition of high-dose melphalan followed by autologous transplantation significantly improves progression-free survival, although an overall survival benefit was not observed in all trials. Moreover, follow up of recent trials is still too short to show any difference in survival. In the light of these findings, novel agent-based induction followed by autologous transplantation is considered the standard upfront treatment for eligible patients (level of evidence: 1A). Post-transplant consolidation and maintenance treatment can further improve patient outcome (1A). The availability of several novel agents has led to the development of multiple combination regimens such as salvage treatment options. In this context, the role of salvage autologous transplantation and allotransplant has not been extensively evaluated. In the case of prolonged remission after upfront autologous transplantation, another autologous transplantation at relapse can be considered (2B). Patients who experience early relapse and/or have high-risk features have a poor prognosis and may be considered as candidates for clinical trials that, in young and fit patients, may also include an allograft in combination with novel agents (2B). Ongoing studies are evaluating the role of novel cellular therapies, such as inclusion of antibody-based triplets and quadruplets, and chimeric antigen receptor-T cells. Despite encouraging preliminary results, longer follow up and larger patient numbers are needed before the clinical use of these novel therapies can be widely recommended.Introduction
The treatment landscape and clinical outcome of multiple myeloma (MM) patients have changed in the last decades,1 with an improved median survival of 8–10 years. Multiple combinations of proteasome inhibitors (PIs) and immunomodulatory drugs (IMIDs) have been under evaluation in the transplant and non-transplant settings, and studies are still ongoing. Several pre-transplant inductions and post-transplant novel agent-based consolidation and maintenance regimens have been investigated, although direct comparisons between such strategies have rarely been performed. Autologous stem cell transplant (ASCT) is currently considered the standard of care for fit newly diagnosed MM (NDMM) patients, although remarkable results obtained in the non-transplant setting32 with novel agent-based treatment have raised questions as to the role of upfront versus delayed ASCT.
The availability of 2-generation PIs and IMIDs, monoclonal antibodies, histone deacetylase inhibitors, and, more recently, check-point inhibitors and small molecules, has led to the development of multiple salvage options that include different combinations of these drugs. In this context, the role of salvage ASCT and allotransplant have not been extensively evaluated. These exciting advances require a critical review to delineate the merit of different induction, consolidation and maintenance approaches, as well as to define the role of upfront ASCT, salvage ASCT and allotransplant in the novel agent era. These important considerations prompted the European Myeloma Network (EMN) to provide guidelines to harmonize treatment selection. A brief overview of novel cellular therapies, which can be considered the new frontier for transplant, is also provided.
Methodology
Clinical EMN experts on MM developed these recommendations based on published data through August 2017. Expert consensus was used to suggest recommendations in case of inconclusive data. Grades of recommendations were assigned using the GRADE criteria for grade of recommendation (Online Supplementary Table S1). The manuscript underwent revision in 3 rounds until the EMN experts reached mutual consent.
Upfront autologous transplant
The current treatment paradigm for NDMM patients eligible for ASCT consists of 4 phases: pre-transplant induction, transplant, post-transplant consolidation and maintenance.
Pre-transplant induction
Induction treatment generally consists of 3–6 cycles with the goal of achieving rapid disease control, improve symptoms, and allow for subsequent successful stem cell collection. The current standard is a 3-drug bortezomib-based combination. Doxorubicin-bortezomib-dexamethasone (PAD) proved to be superior to standard chemotherapy in a randomized trial,4 and more recently, bortezomib-cyclophosphamide-dexamethasone (VCD) was found to be non-inferior to PAD.5 Improved responses were observed with combinations including both PIs and IMIDs. Indeed, complete response (CR) rates were significantly higher with bortezomib-thalidomide-dexamethasone (VTD) compared with thalidomide-dexamethasone (TD) in 2 randomized trials (35% vs. 14%, P=0.0001; 31% vs. 11%, P<0.001).76 VTD versus VCD improved CR rates (13% vs. 9%, respectively).8 Higher CR rates were reported with bortezomib-dexamethasone plus the 2-generation IMID lenalidomide (VRD) (23–48%) (Table 1).109 No direct, randomized comparisons of PAD versus VTD have been made.
Expected efficacy of a given regimen is one of the main factors to be considered in the treatment choice, the second factor being the expected toxicity. Infections are common events in NDMM, often to the underlying disease itself and to the treatment. The main issue with the use of bortezomib (in particular when combined with thalidomide) is the occurrence of peripheral neuropathy (PNP), which can be decreased substantially with subcutaneous and once-weekly administrations. The main concern with combinations including thalidomide or doxorubicin is the thromboembolic risk. Both PNP and thromboembolism rates seem to be lower when bortezomib is associated with cyclophosphamide (Table 2).
Given that the benefit of bortezomib could be hampered by its neurological side effects, 2-generation PIs with minimal neurotoxicity demonstrated that induction treatment with ixazomib-lenalidomide-dexamethasone (IRD) was very well tolerated (no grade 3–4 PNP, cardiac, liver or renal toxicities) and led to a 12% CR rate.11 Carfilzomib-thalidomide-dexamethasone (KTd)12 or carfilzomib-lenalidomide-dexamethasone (KRd)13 lead to a 18–24% CR rate, although cardiovascular toxicities (mainly hypertension) have been reported.
The impact of depth of response on outcome14 provides the rationale for choosing the most effective induction regimen, provided the toxicity profile is acceptable. Nevertheless, only one randomized trial (Myeloma XI) investigated a response-adapted approach based on the sequential use of chemotherapeutic agents, with different modes of action in patients with a suboptimal response (minimal response/partial response) to thalidomide-based induction. Some 40% of patients upgraded their response with VCD before ASCT and significant improvement in PFS was observed (median 48 vs. 38 months; P<0.0001).15 However, the trial included suboptimal induction regimens (CTD and cyclophosphamide-lenalidomide-dexamethasone) not widely used outside the UK. The current standard of care is bortezomib plus IMIDs or chemotherapy, supported also by the results of two meta-analyses1716 that showed the superiority of bortezomib- over non-bortezomib-induction treatments. Thus, the impact of switching treatment, with currently much more effective induction regimens, still needs to be confirmed.
Autologous transplantation
Several trials compared different chemotherapy regimens to standard high-dose melphalan (200 mg/m, MEL200), showing a favorable risk-benefit profile with MEL200 over busulfan/melphalan, idarubicin/melphalan/cyclophosphamide, BCNU/etoposide/melphalan, melphalan 100/140 mg/m. Conditioning regimens including novel agents have so far only been evaluated in single arm studies.18 Given the efficacy and favorable toxicity profile of MEL200, this regimen remains the standard.
Efficacy of novel-agent treatments in the non-transplant setting, together with a manageable safety profile and the advantage of the administration in the outpatient setting, questioned the role of MEL200-ASCT. Four randomized trials compared MEL200-ASCT versus novel agent-based triplets. In two trials, patients received Rd induction and were randomized to tandem MEL200-ASCT or oral lenalidomide-based chemotherapy [melphalan-prednisone-lenalidomide (MPR)/cyclophosphamide-lenalidomide-dexamethasone (CRD)]. Median PFS was significantly longer for patients randomized to tandem MEL200-ASCT than for those randomized to MPR (43 vs. 22 months; P<0.001) or CRD (43 vs. 28 months; P<0.001). Tandem ASCT also improved the 4-year OS rate versus MPR (82% vs. 65%; P=0.02) or CRD (86% vs. 73%; P=0.004).2019
Two large studies compared MEL200-ASCT versus bortezomib-based regimens. MEL200-ASCT significantly prolonged PFS versus bortezomib-lenalidomide-dexamethasone (VRD)10 (median 50 vs. 36 months; P<0.001), and versus bortezomib-melphalan-prednisone (VMP)21 (3-year PFS 65% vs. 57%; P=0.001). Follow up of these two trials is still too short to show any differences in OS.
Indeed, data confirmed that the toxicity profile was better and more manageable in the non-transplant arm, but no increase in toxic deaths was reported with MEL200-ASCT.21201910
Before the introduction of novel agents, several studies showed a prolonged event-free survival (EFS) with double versus single ASCT.22 A subgroup analysis of one of those trials reported an improved OS only in patients achieving less than very good partial response (VGPR) after the first ASCT.23 A more recent integrated analysis of patient-level data from 4 European trials demonstrated that, in patients receiving bortezomib-based induction, the greatest benefit with double versus single ASCT in terms of extended PFS [Hazard Ratio (HR)=0.41] and OS (HR=0.22) was seen in patients with t(4;14) and/or del(17p) who failed CR to induction therapy.24 Preliminary results of the EMN02 trial confirmed that patients receiving double ASCT have a superior PFS in comparison with patients randomized to a single ASCT (3-year PFS 74% vs. 62%; P=0.05). The benefit was particularly evident in patients with high-risk cytogenetics (3-year PFS 65% vs. 41%; HR 0.49, P=0.046).25
On the contrary, the STAMINA trial showed no improvement in PFS in patients receiving double ASCT followed by maintenance versus single ASCT followed by VRD consolidation and lenalidomide maintenance. However, different induction regimen, more effective and prolonged therapy with better disease control before ASCT, as well as non-adherence to the double ASCT policy in 30% of patients can prove to be a limitation of this comparative trial.26
Consolidation regimens
Consolidation is a commonly adopted approach after transplant to improve depth of response. In “naïve” patients, bortezomib consolidation prolonged PFS versus no consolidation (median 27 vs. 20 months, respectively; P=0.05), but no difference in OS was seen.27 In another trial, VTD consolidation increased the CR rate from 15% to 49% and the molecular remission rate from 3% to 18%.14 More recently, post-ASCT consolidation with the same induction regimens was assessed. VTD increased the CR/nCR rate from 63% to 73%.28 Similarly, CR plus stringent CR rate increased from 47% to 50% after VRD.9 Preliminary results of the EMN-02 trial suggest that post-transplant VRD consolidation also prolongs PFS versus no consolidation (3-year PFS 65% vs. 60%, respectively; P=0.045).29 The STAMINA trial did not find any improvement in PFS with single ASCT followed by VRD consolidation and lenalidomide maintenance versus single ASCT followed by lenalidomide maintenance. However, the rate of non-compliance to VRD was sizeable at 12%.26
Similarly to induction phase, combining 2-generation PIs and IMIDs is advantageous also in the consolidation phase, enhancing CR rates from 20% to 32% with IRD, from 31% to 64% with KTD, and from 45% to approximately 70% with KRD.1311
Maintenance regimens
The optimal maintenance regimen should aim at prolonging the remission duration without affecting patients’ quality of life. Although meta-analyses showed a reduced risk of progression (HR=0.65) and death (HR=0.84) with thalidomide maintenance, in the IFM and MRC IX studies, patients with unfavorable cytogenetics did not benefit from this approach.3130 In addition, grade 3–4 PNP (7–19%) and treatment discontinuation due to PNP limit the long-term use of thalidomide.
Bortezomib maintenance seems to be a better option: a landmark analysis of the HOVON-65/GMMG-HD4 trial showed that bortezomib maintenance significantly reduced the risk of progression (P=0.04) and death (P=0.05) as compared with thalidomide, with a similar rate of grade 3–4 PNP (5% vs. 8%).4 Results of this trial also suggest that pre-transplant bortezomib induction followed by bortezomib maintenance significantly reduces the high-risk impact of del(17p) and renal impairment on survival.32 More recently, longer PFS was reported also with the bortezomib-thalidomide combination versus thalidomide alone.33
Lenalidomide is another valid strategy for long-term treatment, with limited neurotoxicity: 4 trials subsequently evaluated lenalidomide maintenance after ASCT,363419 showing a consistent PFS benefit for lenalidomide versus no maintenance (HR range 0.46–0.50). A meta-analysis of the first three randomized trials reported a significant increase also in OS (7-year OS 62% vs. 50%; HR 0.75, P=0.001) across all subgroups analyzed with the exception of patients with high-risk cytogenetics. In the MRC trial, a significant PFS benefit was maintained also in patients with high-risk cytogenetics, but no data on OS are currently available. Main grade 3–4 toxicities were neutropenia (23–51%), and infections (6–13%).353419 Although second primary malignancies (SPMs) were higher with lenalidomide maintenance versus control (hematologic SPM 6.1% vs. 2.8%; solid tumor SPM 7.3% vs. 4.2%),37 the OS benefit outweighed the SPM risk.
Recommendations in NDMM patients eligible for high-dose therapy and ASCT, sequential treatment including novel agent-based induction, upfront transplant, post-transplant bortezomib plus IMIDs consolidation and maintenance is recommended (1A) (Figure 1). Treatment choice should be based on evidence supporting a specific treatment, and on a thorough evaluation of the patient’s characteristics, toxicity of the expected regimens, and availability of drugs in the specific countries (Table 3).
Special considerations
Currently, selection criteria for high-dose therapy include age and comorbidities. However, a definite age cut off, rather than assessment of patient’s biological age, comorbidities, fitness and frailty/comorbidity scores is suboptimal. Besides age, the performance status, and cardiac, pulmonary, hepatic and renal functions should be considered to better evaluate the risk-benefit ratio of transplant for each patient, and specific risk-assessment models, such as the Myeloma Comorbidity Index (MCI) and/or Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI) should be used to better modulate the dose of chemotherapy.4038 Specific considerations refer to patients with renal failure (RF) and elderly patients.
Renal failure (RF)
Approximately 20% of patients have creatinine more than 2 mg/dL at diagnosis. Bortezomib-based regimens remain the cornerstone of management of renal failure (RF). Indeed, higher response rates were reported with PAD versus VAD induction in patients with RF (81% vs. 63%; P=0.31).41 In dialysis patients, bortezomib-based induction versus conventional chemotherapy significantly increased pre-transplant (83% vs. 36%; P=0.02) and post-transplant (100% vs. 58%; P=0.01) overall response rate. Prolonged EFS and a trend towards less time on hemodialysis (6 vs. 17 months) was also reported.42 Combination of bortezomib with high cut-off hemodialysis led to prompt and remarkable responses.43 RF does not appear to affect the quality of stem cell collection.44 Persistent RF or dialysis are not contraindications to high-dose therapy and ASCT,45 since patients may improve renal function after ASCT. Nevertheless, the rate of treatment-related mortality (TRM) ranges from 0 to 29% in different reports and with different melphalan doses.4442 Thus, due to the potentially higher toxicity of 200 mg/m, dose reductions are mandatory, particularly in dialysis patients. Other suggested reductions in case of impaired organ function are reported in Online Supplementary Table S2. Of note, a recent large retrospective analysis showed no significant differences in the 5-year PFS and OS between transplant patients with normal, moderate [glomerular filtration rate (GFR) 30–60 mL/min/1.73 m)] and severe RF (GFR<30). For patients with moderate RF, 5-year PFS was 18% with melphalan 140 mg/m, and 46% with melphalan 200 mg/m (P=0.009); 5-year OS was 67% and 68%, respectively (P=0.52). In patients with severe RF (GFR<30), no differences in 5-year PFS and OS were reported between groups. Relapse remained the primary cause of death in all patient subgroups.46 In this report, 85% patients achieved dialysis independence post ASCT even though, in previous case series, rate of dialysis independence varied from 6% to 25%.44
Of interest, around 10% of younger patients may achieve long-lasting responses, which makes them potential candidates for renal transplantation. However, many issues, including donor availability, the immunosuppression risks and the possible disease relapse on the xenograft, need to be considered. Thus, patients with low-risk disease and with negative minimal residual disease (MRD) might be considered eligible for transplantation in the future but currently, due to limited data, no recommendations can be made.44
Transplant in the elderly
Aging is associated with reduced organ function and drug metabolism, with potentially increased toxicity and TRM. The potential increase in toxicity has led to the evaluation of reduced doses of melphalan conditioning (100–140 mg/m). Many studies, mostly retrospective, observa tional or registry-based, provided encouraging results with ASCT in patients over 65 years of age, with TRM less than 3–4%.47 No differences in TRM (1%) were reported with tandem melphalan 140 mg/m in patients aged 60–65 years versus 65–70 years in the large DSMM II trial.48 Interestingly, a recent study found that ASCT-TRM was 0% with either melphalan 140 mg/m or 200 mg/m, which may partly be due to improvements in supportive therapy and better patient selection.49 A recent European Society for Blood and Marrow Transplantation (EBMT) study confirms increased utilization and safety of ASCT with improved post-transplant survival, particularly in elderly MM patients.50
Former analysis of non-ASCT treatment versus ASCT in the elderly (65–75 years) compared thalidomide-based chemotherapy (MPT) versus reduced-intensity (melphalan 100 mg/m) ASCT in patients aged 65–75 years in the IFM9906 trial. MPT significantly reduced the risk of progression (HR 0.54, P=0.0002)51 and death (HR 0.69, P=0.027), but the lack of novel agents in the pre-ASCT induction and the low melphalan dosing could be a limitation to the study. The rate of toxic deaths was also higher (5%) during induction in the ASCT arm. Other prospective trials subsequently evaluated a sequential approach including novel agent based-induction, consolidation and maintenance. One study showed that PAD induction, followed by MEL100-ASCT, lenalidomide-prednisone consolidation and lenalidomide maintenance was highly efficacious (VGPR rate 82%, 5-year OS 63%) and feasible, in particular for patients under 70 years of age who reported a significantly lower rate of TRM in comparison with elderly patients (5% vs. 19%).52 A recent report suggests that bortezomib consolidation after ASCT may determine clinical outcomes in older patients, who may have been less heavily pre-treated, as in younger patients treated with standard doses of melphalan.53 The phase III DSMM XIII trial compared continuous Rd versus Rd induction followed by tandem melphalan 140 mg/m-ASCT and lenalidomide maintenance. Results of the planned interim analysis showed a 3-year-survival rate of 75% for all patients. A longer follow up is needed to evaluate the potential advantages and disadvantages of combining lenalidomide with high-dose melphalan-ASCT as compared with continuous RD.54
Recommendations: biological age rather than chronological age, PS, and organ function should be considered to better evaluate the risk-benefit ratio of transplant for each patient (1B) (Figure 2). Objective risk-assessment scores, such as the Revised-Myeloma Comorbidity Index (R-MCI) and/or the Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI) can be used to define the appropriate dose of chemotherapy4038 (1B) (Table 4).
Transplant at relapse
Upfront versus rescue transplant
In the past, several randomized trials confirmed the PFS benefit with early ASCT as compared with chemotherapy. In 3 randomized studies, OS was similar whether ASCT was performed early or at first relapse. Despite similar OS, early ASCT improved the average time without symptoms and reduced treatment-related toxicities in 1 trial.55 However, at the time of these trials, most novel agents were not available. Based on the impressive results of novel agent-based treatments in the non-transplant setting, the option of delaying ASCT until first relapse was reconsidered.5632 In all the recent randomized phase III trials comparing ASCT versus novel agent-based therapies, patients who did not receive ASCT upfront were recommended to receive it at first relapse. A pooled analysis including the GIMEMA and the EMN441 trials showed that only 53% of patients eligible for Mel200-ASCT at diagnosis actually received ASCT at first relapse. Upfront MEL200-ASCT significantly improved not only PFS1, but also PFS2 (4-year PFS2 71% vs. 54%; HR 0.53, P<0.001) and OS (4-year OS 84% vs. 70%; HR 0.51, P<0.001) as compared with oral chemotherapy plus lenalidomide.57 Of note, in the IFM2009 trial, in which up to 79% of patients treated with lenalidomide plus bortezomib upfront were rescued with ASCT at relapse, no differences in OS were noticed.10
Transplant in patients relapsing after prior autograft
Multiple retrospective analyses showed that chemo-sensitivity and remission duration after first ASCT are the most important prognostic factors for long-term disease control after salvage ASCT.5958 Most reports also highlighted the impact of the number of prior therapies on outcome, suggesting that salvage ASCT should be part of the initial salvage strategies, rather than be offered to patients who have failed multiple therapy lines. A retrospective analysis on 1061 patients showed a significantly longer median survival for patients who received salvage ASCT (4 years) versus those who received salvage IMIDs/PIs and no ASCT (3.3 years), and those who received conventional chemotherapy (2.5 years).60 A limitation of this analysis is a possible selection bias as patients who were treated with ASCT may have been in better clinical condition compared with those who were not. Nevertheless, the phase III multicenter randomized Myeloma X trial showed a significant advantage in time to progression (19 vs. 11 months; P<0.001) and OS (67 vs. 52 months; P=0.022) in patients relapsing after a previous ASCT, and then randomized to receive either a second ASCT or oral cyclophosphamide.61 The limitation of these trials, however, is that, even though all patients were re-induced with PAD prior to randomization, the control arm with cyclophosphamide alone can now be considered suboptimal. A recent retrospective EBMT analysis showed that even a third ASCT at relapse may be feasible, with more than 80% of patients achieving at least a PR, although with increased non-relapse mortality. Particularly in severely cytopenic patients in whom hematologic toxicity of conventional treatment may be prohibitive, ASCT may be a rescue option. The option of a third ASCT mostly followed a previous upfront approach with tandem ASCT; some patients received a first ASCT followed by a second ASCT at second relapse and a third ASCT at subsequent relapse. The first scenario resulted in better results with a median OS of more than four years if the relapse occurred after more than three years after the upfront tandem ASCT.62
Recommendations: upfront ASCT remains the standard option for patients eligible for HDT (1A) (Figure 1). A second transplant at relapse should be considered after a minimal duration of remission of 18 months after a first ASCT (1B); this cut off could be extended to 24 months in the context of novel induction/maintenance.63 A second ASCT should be offered as a first salvage therapy rather than after failing multiple lines (2B). Novel-agent based induction and consolidation-maintenance should be adopted also in the elderly (1A).
Allotransplant
When and in which patients
A review of approximately 3000 ASCTs and allo-SCTs, performed in the USA between 2007 and 2009 showed that, overall, 47.1% of ASCTs and only 3.6% of allo-SCTs were performed in MM patients.64 However, the number of allo-SCTs for MM in Europe steadily increased from 1990 to 2012.65 Before new drugs became readily available almost 20 years ago, in a series of “biologically” randomized prospective studies, the concept of splitting myeloablation and graft-versus-myeloma (GvM) by a tandem approach with a standard ASCT followed by a non-myeloablative allo-SCT from a matched sibling or an unrelated donor was explored in NDMM (Table 5).7666 Results were discordant, and this was likely due to differences in study design, target population and post-transplant immunosuppression (Table 3). Moreover, only at long-term follow up were differences in clinical outcomes between arms observed.7473 Of note, at that time, most studies could not include new drugs either at induction or as post allo-SCT maintenance/consolidation.
Partly due to the conflicting results and to the introduction of new drugs, in recent years used allo-SCT has tended to be used as a salvage strategy at relapse, often not in the context of clinical trials. Most reports were single institution or registry analyses. Only a few comparative studies have been conducted, and these are limited by their retrospective nature and/or small patient cohorts (Table 6). In a recent EBMT report65 on 7333 MM patients who underwent allo-SCT between 1990 and 2012, 3405 had received allo-SCT as a second line or beyond regimen; this report showed that 25% of the patient cohort who received allo-SCT more than eight months from the first ASCT survived at ten years, suggesting that cure may have been reached through a GvM mechanism in some patients. Another retrospective EBMT analysis identified patient and donor cytomegalovirus (CMV) seronegativity as the key prognostic factor for better outcome after allo-SCT in relapsed patients.77 One prospective study78 concluded that, with well-matched donors, the non-relapse mortality was 10%, and approximately 20% of patients achieved long-term disease-free survival. The high response rates seen after donor lymphocyte infusions (DLI) administration provide additional evidence for the GvM effect.
Taken together, these studies have showed the feasibility of allo-SCT in relapsed MM; however, given the heterogeneous patient cohorts and differences in conditioning regimens and supportive care, its real role and curative potential has not been clearly established. Both reduced-intensity and myeloablative conditionings have been successfully used and, so far, the choice should be based on center policy and patients’ comorbidities.
Considering the lack of effective therapy for high-risk patients carrying del(17p), gain(1q), t(4;14) and t(14;16) abnormalities, new treatment modalities should be sought in this patient subset. The negative prognostic impact of high-risk cytogenetics appeared to be partly neutralized by GvM in two recent studies. Kröger et al. did not observe significant differences in PFS between patients harboring del17p13 and/or t(4;14) and those without these genetic abnormalities after a median follow up of six years (24% vs. 30%; P=0.70). Depth of remission had a remarkable impact on 5-year PFS: 17% for PR, 41% for CR, 57% for molecular CR, and 85% for sustained molecular CR.79 A French trial also showed no differences in clinical outcomes between t(4;14) and non-t(4;14) patients. Moreover, the 3-year progression rate did not exceed 45% in patients with del(17p).80 Taken together, these findings raise the question as to whether high-risk patients who usually experience poor outcomes and easily develop resistance to novel agents would benefit from allo-SCT earlier in the course of the disease.
Evidence of graft-versus-myeloma effect
Response to DLIs is often seen as proof of GvM effect. However, the prolonged post-relapse survival reported after tandem auto-allo-SCT upfront suggests an important synergy between novel agents and GvM.7473 In several reports, DLIs have been used as salvage treatment. Beitinjaneh et al.81 reported on 23 of 162 patients with MM receiving DLI post allo-SCT for residual or relapsed disease: 22% achieved VGPR or better with a median duration of 21.8 months. Similarly, an analysis of EBMT registry data reported a response rate of 63% in 70 patients when DLI was given pre-emptively and 52% when given at relapse.82 Ladetto et al. reported a gradual reduction of residual disease with longer follow up. Minimal residual disease negativity, detected by molecular methods, remained low up to three months post alloSCT, then increased up to 44% at six and 47% at 12 months.83 Importantly, these patients did not receive any maintenance/consolidation treatment. These findings also compared favorably with the molecular analysis conducted by the same group in patients undergoing autografting and VTD consolidation.14 Finally, although not univocal, many trials reported a favorable association between development of chronic graft-versus-host disease (GvHD) and prolonged PFS and OS,8584 again supporting a GvM effect.
Novel agents and graft-versus-myeloma effect
Although the introduction of ‘new drugs’ has made allografting a less attractive treatment option because of its toxicity, the mechanisms of action of new drugs and immune-mediated GvM effects are by no means mutually exclusive.7473 Given that one of the most important predictors of survival is the response at the time of transplant, and the major limitation remains disease recurrence as for all other treatments, the new anti-MM drugs may strongly improve outcomes of allo-SCT. Moreover, the concept of maintenance treatment was also recently introduced in the setting of allografting. Bortezomib has been used before allo-SCT and early after allo-SCT to eliminate residual disease and to decrease GvHD incidence and severity based on its presumed immunomodulatory potency in at least two prospective studies8786 on 16 and 12 high-risk MM patients, respectively. Both trials proved feasible and safe and, based on these results, the expert panel agree that larger confirmatory studies should be designed.
Lenalidomide is also of interest in the allo-SCT setting, although this should be considered with caution because of the risk of GvHD flares if given too soon after transplant. Three trials898886 demonstrated that post allo-SCT lenalidomide maintenance was feasible and contributed to further reduce MM tumor burden with PFS rates of 52% at three years;88 63% at three years,89 and 60% at two years.90 GvHD flares were observed in 28–47% of cases.
Update on current studies
At the 2016 American Society of Hematology meeting (December 2016), reports on myeloma and allo-SCT mainly focused on interactions of new drugs and GvM effect, and three groups unanimously reported remarkable responses to new drugs used as post-allo-SCT salvage, clearly showing a synergism with GvM effect. A retro spective study comparing OS after relapse from upfront auto-allo (n=178) versus double auto-SCT (n=404) was conducted through the registry of the Center for International Blood and Marrow Transplant Research (CIBMTR).91 Despite a higher risk population (46% of early relapse from 2 SCT vs. 26%) in the allo-SCT group, long-term reduction in post-relapse mortality (HR for death in auto-auto-SCT=1.55; P=0.0052) was observed. This was clearly attributable to improved response to salvage therapy due to the donor-derived immunological milieu that potentiated the immune effects of new agents. Similarly, Giaccone et al. showed prolonged OS from 1 relapse post tandem auto-allo-SCT compared to double auto-SCT (89.8 months vs. 23.5 months; P=0.009).92 López-Corral et al. reported similar pre-transplant and post-transplant response rates and durability of response achieved with new drugs before and after allo-SCT; responses post allo-SCT were at least similar in proportion and durability to those observed in the pre-transplant setting, which is in contrast to the usual course of the disease outside the allo-SCT setting.93 Another study reported on 18 high-risk MM patients who received upfront auto-SCT followed by RIC allo-SCT and bortezomib as maintenance, which was overall well tolerated, although 4 of 18 had asymptomatic Epstein Barr virus (EBV) reactivation. Depth of response improved after bortezomib, with 67% of patients in CR or stringent CR.94
Daratumumab has also demonstrated encouraging efficacy in 10 heavily pre-treated relapsed/refractory patients after allo-SCT. The safety profile was good with, in the majority of cases, non-severe adverse effects (AEs) mostly after the first infusion; 5 of 9 evaluable patients responded and all responding patients maintained their responses 7, 14, 35, 54 and 84 days after the first administration.95 Cook et al. monitored immune biomarkers with the use of lenalidomide after T-cell-depleted reduced intensity conditioning (RIC)-alloSCT, showing that the agent allowed sustained quantitative and functional reconstitution of donor immune homeostasis.96 McKiernan et al.97 reported a long-term comparison in patients receiving allo-SCT as upfront consolidation (n=75) or as salvage therapy (n=43). The 10-year OS for patients who received allo-SCT as salvage was 36% versus 68% for the consolidation group (P=0.0007). Of note, having undergone 2 or more prior auto-SCTs predicted for a higher risk of mortality (P=0.05). Chronic GvHD was favorable, associated with a 36% improvement in OS (P=0.0008).
Recommendations: previous studies that did not include novel agents reported long-term molecular remissions, and possibly cure, in patient subsets. Well-designed prospective trials combining GvM and new drugs may become urgent in young high-risk/ultra high-risk patients whose treatment remains an unmet clinical need. However, there are no current data supporting an upfront allograft. A clinical indication or recommendation may also become “early relapse” after first-line treatment (including the new PI and IMIDs) which identifies patients at very poor prognosis independent of other prognostic factors (Table 7). Re-induction to obtain tumor shrinking using novel drugs as a bridge to transplant is highly recommended/mandatory in this setting.98 Novel agent-based combinations should be considered also in association with DLI in case of relapse after allogeneic transplant.
Future developments
Treatment for MM has undergone a dramatic improvement in the past decade given the considerable advances in the understanding of the disease pathogenesis and the approval of numerous novel drugs and combinations for the disease. However, despite the development of novel agents which target not only MM cells but also the microenvironment,99 the prognosis of patients with early relapsed/refractory MM remains poor. Thus, new therapeutic modalities are urgently needed to overcome resistance to current therapies. Several immunotherapies have recently been proposed which, among others, include monoclonal antibodies, antibody-drug conjugates, chimeric antigen receptor T-cell therapy (CAR-T cells), tumor vaccines and immune checkpoint inhibitors.100 Preliminary results observed in patients with B-cell hematologic malignancies with infusion of T cells genetically modified to express synthetic CARs against the lineage-specific surface antigen CD19 were impressive. T cells engineered with an anti-CD19 CAR induced CR also in a patient with MM.101 Recently, a number of other CAR-T cells have been designed to target surface antigens expressed by MM cells and include CD38,102 CD138,103 CD269, the B-cell maturation antigen (BCMA),104 κ light chains,105 CS1 (CD319)106 and CD44v6.107 However, despite their efficacy, CAR-T cells have raised many concerns on their short- and long-term toxicities, in particular, the development of life-threatening cytokine release syn drome (CRS) and prolonged aplasia of the healthy counterparts.108 Genetic modifications of cells belonging to the innate immune system, such as natural killer (NK) cells, are also being explored, and modification of the human NK-cell lines NKL and NK-92 with a lentiviral vector encoding for CS1 and CD138 CARs has proven to be feasible.109 However, several steps to optimize and validate CAR-modified NK cells have to be undertaken before their wider clinical use can be considered.
Conclusions
Over the last two decades, changes in the treatment paradigm for MM patients have dramatically improved survival. Clearly, results of the most recently published trials confirm the role of ASCT in the era of novel agents, with new drugs administered both in the pre-transplant and post-transplant phases. The expert panel emphasizes that current clinical research should maintain a balance between treatment efficacy and quality of life, identify the optimal sequencing of treatment, the appropriate tools for patient selection, evaluate costs of prolonged novel-agent application versus transplant remission efficacy, and treatment-free intervals, and it should identify how to best induce long-term remission.110 In the future, objective, prospective and proficiently performed fitness tools may prove to be of benefit before intensive treatment is start ed, especially since fitness assessments made by patients and physicians are not as objective as fitness evaluations derived from well-defined tests and scores. Future randomized studies will also need to evaluate the role of ASCT as salvage treatment in the context of the novel combinations currently available as salvage options.
The trend in survival improvement is likely to continue in the future with new classes of drugs [such as monoclonal antibodies (MoAbs)] and 2-generation PIs and IMIDs moving in the upfront setting. If most patients can now expect long-term disease control, the optimal definition of high-risk disease and the specific treatment for these patients remains a major challenge. Based on the available data, the opinion of the expert committee is that allotransplant in combination with novel agents might be considered in the context of clinical trials for high-risk patients who are willing to accept the TRM for a chance of a better long-term survival. Moreover, cellular therapies, that for the moment are still highly experimental, should be optimized and made more widely available and cost approved so they can be included in our treatment armamentarium.
Acknowledgments
This work is supported by the Deutsche Krebshilfe (grants 1095969 and 111424 to ME and RW). The expert panel thanks all the investigators of the EMN group in the different countries for their support.
Footnotes
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/103/2/197
- Received June 28, 2017.
- Accepted December 5, 2017.
References
- Kumar SK, Dispenzieri A, Lacy MQ. Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients. Leukemia. 2014; 28(5):1122-1128. PubMedhttps://doi.org/10.1038/leu.2013.313Google Scholar
- San Miguel JF, Schlag R, Khuageva NK. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008; 359(9):906-917. PubMedhttps://doi.org/10.1056/NEJMoa0801479Google Scholar
- Benboubker L, Dimopoulos MA, Dispenzieri A. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014; 371(10):906-917. PubMedhttps://doi.org/10.1056/NEJMoa1402551Google Scholar
- Sonneveld P, Schmidt-Wolf IGH, van der Holt B. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/GMMG-HD4 trial. J Clin Oncol. 2012; 30(24):2946-2955. PubMedhttps://doi.org/10.1200/JCO.2011.39.6820Google Scholar
- Mai EK, Bertsch U, Dürig J. Phase III trial of bortezomib, cyclophosphamide and dexamethasone (VCD) versus bortezomib, doxorubicin and dexamethasone (PAd) in newly diagnosed myeloma. Leukemia. 2015; 29(8):1721-1729. PubMedhttps://doi.org/10.1038/leu.2015.80Google Scholar
- Rosiñol L, Oriol A, Teruel AI. Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pre-transplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood. 2012; 120(8):1589-1596. PubMedhttps://doi.org/10.1182/blood-2012-02-408922Google Scholar
- Cavo M, Tacchetti P, Patriarca F. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, dou ble autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3. Lancet. 2010; 376(9758):2075-2085. PubMedhttps://doi.org/10.1016/S0140-6736(10)61424-9Google Scholar
- Moreau P, Hulin C, Macro M. VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood. 2016; 127(21):2569-2574. PubMedhttps://doi.org/10.1182/blood-2016-01-693580Google Scholar
- Roussel M, Lauwers-Cances V, Robillard N. Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: a phase II study by the Intergroupe Francophone du Myélo. J Clin Oncol. 2014; 32(25):2712-2717. PubMedhttps://doi.org/10.1200/JCO.2013.54.8164Google Scholar
- Attal M, Lauwers-Cances V, Hulin C. Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N Engl J Med. 2017; 376(14):1311-1320. Google Scholar
- Moreau P, Hulin C, Caillot D. Ixazomib-Lenalidomide-Dexamethasone (IRd) Combination before and after Autologous Stem Cell Transplantation (ASCT) Followed By Ixazomib Maintenance in Patients with Newly Diagnosed Multiple Myeloma (NDMM): A Phase 2 Study from the Intergroupe Francophone. Blood. 2016; 128(22):674. Google Scholar
- Wester R, van der Holt B, Asselbergs E. Phase 2 Study of Carfilzomib, Thalidomide, and Low-Dose Dexamethasone As Induction/Consolidation in Newly Diagnosed, Transplant Eligible Patients with Multiple Myeloma, the Carthadex Trial. Blood. 2016; 128(22):1141. Google Scholar
- Roussel M, Lauwers-Cances V, Robillard N. Frontline Therapy with Carfilzomib, Lenalidomide, and Dexamethasone (KRd) Induction Followed By Autologous Stem Cell Transplantation, Krd Consolidation and Lenalidomide Maintenance in Newly Diagnosed Multiple Myeloma (NDMM) Patients: Primary Results of the Intergroupe Francophone Du MyéLome (IFM) Krd Phase II Study. Blood. 2016; 128(22):1142. Google Scholar
- Ferrero S, Ladetto M, Drandi D. Long-term results of the GIMEMA VEL-03-096 trial in MM patients receiving VTD consolidation after ASCT: MRD kinetics’ impact on survival. Leukemia. 2015; 29(3):689-695. PubMedhttps://doi.org/10.1038/leu.2014.219Google Scholar
- Jackson GH, Davies FE, Pawlyn C. Response Adapted Induction Treatment Improves Outcomes for Myeloma Patients; Results of the Phase III Myeloma XI Study. Blood. 2016; 128(22):244. Google Scholar
- Sonneveld P, Goldschmidt H, Rosiñol L. Bortezomib-based versus non-bortezomib-based induction treatment before autologous stem-cell transplantation in patients with previously untreated multiple myeloma: a meta-analysis of phase III randomized, controlled trials. J Clin Oncol. 2013; 31(26):3279-3287. PubMedhttps://doi.org/10.1200/JCO.2012.48.4626Google Scholar
- Nooka AK, Kaufman JL, Behera M. Bortezomib-containing induction regimens in transplant-eligible myeloma patients. Cancer. 2013; 119(23):4119-4128. PubMedhttps://doi.org/10.1002/cncr.28325Google Scholar
- Shah N, Callander N, Ganguly S. Hematopoietic Stem Cell Transplantation for Multiple Myeloma: Guidelines from the American Society for Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 2015; 21(7):1155-1166. PubMedGoogle Scholar
- Palumbo A, Cavallo F, Gay F. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med. 2014; 371(10):895-905. PubMedhttps://doi.org/10.1056/NEJMoa1402888Google Scholar
- Gay F, Oliva S, Petrucci MT. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol. 2015; 16(16):1617-1629. PubMedhttps://doi.org/10.1016/S1470-2045(15)00389-7Google Scholar
- Cavo M, Beksac M, Dimopoulos MA. Intensification Therapy with Bortezomib-Melphalan-Prednisone Versus Autologous Stem Cell Transplantation for Newly Diagnosed Multiple Myeloma: An Intergroup, Multicenter, Phase III Study of the European Myeloma Network (EMN02/HO95 MM Trial). Blood. 2016; 128(22):673. Google Scholar
- Fermand JP, Alberti C, Marolleau JP. Single versus tandem high dose therapy (HDT) supported with autologous blood stem cell (ABSC) transplantation using unselected or CD34-enriched ABSC: Results of a two by two designed randomized trial in 230 young patients with multiple myeloma (MM). Hematol J. 2003; 4(Suppl 1):S59. Google Scholar
- Attal M, Harousseau J-L, Facon T. Single versus Double Autologous Stem-Cell Transplantation for Multiple Myeloma. N Engl J Med. 2003; 349(26):2495-2502. PubMedhttps://doi.org/10.1056/NEJMoa032290Google Scholar
- Cavo M, Salwender H, Rosiñol L. Double Vs Single Autologous Stem Cell Transplantation After Bortezomib-Based Induction Regimens For Multiple Myeloma: An Integrated Analysis Of Patient-Level Data From Phase European III Studies. Blood. 2013; 122(21):767. Google Scholar
- Cavo M, Petrucci MT, Di Raimondo F. Upfront Single Versus Double Autologous Stem Cell Transplantation for Newly Diagnosed Multiple Myeloma: An Intergroup, Multicenter, Phase III Study of the European Myeloma Network (EMN02/HO95 MM Trial). Blood. 2016; 128(22):991. Google Scholar
- Stadtmauer EA, Pasquini MC, Blackwell B. Comparison of Autologous Hematopoietic Cell Transplant (autoHCT), Bortezomib, Lenalidomide (Len) and Dexamethasone (RVD) Consolidation with Len Maintenance (ACM), Tandem Autohct with Len Maintenance (TAM) and Autohct with Len Maintenance (AM) for upFront. Blood. 2016; 128(22):LBA-1. Google Scholar
- Mellqvist U-H, Gimsing P, Hjertner O. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood. 2013; 121(23):4647-4654. PubMedhttps://doi.org/10.1182/blood-2012-11-464503Google Scholar
- Cavo M, Pantani L, Petrucci MT. Bortezomib-thalidomide-dexamethasone is superior to thalidomide-dexamethasone as consolidation therapy after autologous hematopoietic stem cell transplantation in patients with newly diagnosed multiple myeloma. Blood. 2012; 120(1):9-19. PubMedhttps://doi.org/10.1182/blood-2012-02-408898Google Scholar
- Sonneveld P, Beksac M, van der Holt B. Consolidation Followed By Maintenance Therapy Versus Maintenance Alone in Newly Diagnosed, Transplant Eligible Patients with Multiple Myeloma (MM): A Randomized Phase 3 Study of the European Myeloma Network (EMN02/HO95 MM Trial). ASH. 2016; 128(22):242. Google Scholar
- Attal M, Harousseau J-L, Leyvraz S. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood. 2006; 108(10):3289-3294. PubMedhttps://doi.org/10.1182/blood-2006-05-022962Google Scholar
- Morgan GJ, Gregory WM, Davies FE. The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis. Blood. 2012; 119(1):7-15. PubMedhttps://doi.org/10.1182/blood-2011-06-357038Google Scholar
- Goldschmidt H, Lokhorst HM, Mai EK. Bortezomib before and after high-dose therapy in myeloma: long-term results from the phase III HOVON-65/GMMG-HD4 trial. Leukemia. 2017. Google Scholar
- Rosiñol L, Oriol A, Teruel AI. Bortezomib and thalidomide maintenance after stem cell transplantation for multiple myeloma: a PETHEMA/GEM trial. Leukemia. 2017; 31(9):1922-1927. Google Scholar
- Attal M, Lauwers-Cances V, Marit G. Lenalidomide Maintenance after Stem-Cell Transplantation for Multiple Myeloma. N Engl J Med. 2012; 366(19):1782-1791. PubMedhttps://doi.org/10.1056/NEJMoa1114138Google Scholar
- McCarthy PL, Owzar K, Hofmeister CC. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012; 366(19):1770-1781. PubMedhttps://doi.org/10.1056/NEJMoa1114083Google Scholar
- Jackson GH, Davies FE, Pawlyn C. Lenalidomide Is a Highly Effective Maintenance Therapy in Myeloma Patients of All Ages; Results of the Phase III Myeloma XI Study. Blood. 2016; 128(22):1143. Google Scholar
- McCarthy PL, Holstein SA, Petrucci MT. Lenalidomide Maintenance After Autologous Stem-Cell Transplantation in Newly Diagnosed Multiple Myeloma: A Meta-Analysis. J Clin Oncol. 2017; 35(29):3279-3289. Google Scholar
- Saad A, Mahindra A, Zhang M-J. Hematopoietic cell transplant comorbidity index is predictive of survival after autologous hematopoietic cell transplantation in multiple myeloma. Biol Blood Marrow Transplant. 2014; 20(3):402-408.e1. Google Scholar
- Labonté L, Iqbal T, Zaidi MA. Utility of comorbidity assessment in predicting transplantation-related toxicity following autologous hematopoietic stem cell transplantation for multiple myeloma. Biol Blood Marrow Transplant. 2008; 14(9):1039-1044. PubMedhttps://doi.org/10.1016/j.bbmt.2008.06.019Google Scholar
- Engelhardt M, Domm A-S, Dold SM. A concise revised myeloma comorbidity Index as a valid prognostic instrument in a large cohort of 801 multiple myeloma patients. Haematologica. 2017; 102(5):910-921. PubMedhttps://doi.org/10.3324/haematol.2016.162693Google Scholar
- Scheid C, Sonneveld P, Schmidt-Wolf IGH. Bortezomib before and after autologous stem cell transplantation overcomes the negative prognostic impact of renal impairment in newly diagnosed multiple myeloma: a subgroup analysis from the HOVON-65/GMMG-HD4 trial. Haematologica. 2014; 99(1):148-154. PubMedhttps://doi.org/10.3324/haematol.2013.087585Google Scholar
- Breitkreutz I, Heiss C, Perne A. Bortezomib improves outcome after SCT in multiple myeloma patients with end-stage renal failure. Bone Marrow Transplant. 2014; 49(11):1371-1375. PubMedhttps://doi.org/10.1038/bmt.2014.165Google Scholar
- Zannetti BA, Zamagni E, Santostefano M. Bortezomib-based therapy combined with high cut-off hemodialysis is highly effective in newly diagnosed multiple myeloma patients with severe renal impairment. Am J Hematol. 2015; 90(7):647-652. PubMedhttps://doi.org/10.1002/ajh.24035Google Scholar
- Gavriatopoulou M, Terpos E, Kastritis E, Dimopoulos MA. Current treatments for renal failure due to multiple myeloma. Expert Opin Pharmacother. 2016; 17(16):2165-2177. Google Scholar
- Dimopoulos MA, Sonneveld P, Leung N. International Myeloma Working Group Recommendations for the Diagnosis and Management of Myeloma-Related Renal Impairment. J Clin Oncol. 2016; 34(13):1544-1557. PubMedhttps://doi.org/10.1200/JCO.2015.65.0044Google Scholar
- Mahindra A, Hari P, Fraser R. Autologous hematopoietic cell transplantation for multiple myeloma patients with renal insufficiency: a center for international blood and marrow transplant research analysis. Bone Marrow Transplant. 2017. Google Scholar
- Ozaki S, Shimizu K. Autologous stem cell transplantation in elderly patients with multiple myeloma: past, present, and future. Biomed Res Int. 2014; 2014:394792. Google Scholar
- Straka C, Liebisch P, Salwender H. Autotransplant with and without induction chemotherapy in older multiple myeloma patients: long-term outcome of a randomized trial. Haematologica. 2016; 101(11):1398-1406. PubMedhttps://doi.org/10.3324/haematol.2016.151860Google Scholar
- Garderet L, Beohou E, Caillot D. Upfront autologous stem cell transplantation for newly diagnosed elderly multiple myeloma patients: a prospective multicenter study. Haematologica. 2016; 101(11):1390-1397. PubMedhttps://doi.org/10.3324/haematol.2016.150334Google Scholar
- Auner HW, Szydlo R, Hoek J. Trends in autologous hematopoietic cell transplantation for multiple myeloma in Europe: increased use and improved outcomes in elderly patients in recent years. Bone Marrow Transplant. 2015; 50(2):209-215. PubMedhttps://doi.org/10.1038/bmt.2014.255Google Scholar
- Facon T, Mary JY, Hulin C. 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; 370(9594):1209-1218. PubMedhttps://doi.org/10.1016/S0140-6736(07)61537-2Google Scholar
- Gay F, Magarotto V, Crippa C. Bortezomib induction, reduced-intensity transplantation, and lenalidomide consolidation-maintenance for myeloma: updated results. Blood. 2013; 122(8):1376-1383. PubMedhttps://doi.org/10.1182/blood-2013-02-483073Google Scholar
- Straka C, Knop S, Vogel M. Bortezomib Consolidation Following Autologous Transplant Equalizes the Outcome for Older Patients with Less Intensive Pretreatment Compared to Younger Patients with Newly Diagnosed Multiple Myeloma. Blood. 2016; 128(22):516. Google Scholar
- Straka C, Schaefer-Eckart K, Bassermann F. Lenalidomide with Low-Dose Dexamethasone (Rd) Continuously Versus Rd Induction, Tandem MEL140 with Autologous Transplantation and Lenalidomide Maintenance: Planned Interim Analysis of a Prospective Randomized Trial in Patients 60–75 Years of Age with Mult. Blood. 2014; 124(21):3969. Google Scholar
- Fermand JP, Ravaud P, Chevret S. High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: upfront or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood. 1998; 92(9):3131-3136. PubMedGoogle Scholar
- Koreth J, Cutler CS, Djulbegovic B. 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; 13(2):183-196. PubMedhttps://doi.org/10.1016/j.bbmt.2006.09.010Google Scholar
- Gay F, Oliva S, Petrucci MT. Autologous transplant vs oral chemotherapy and lenalidomide in newly diagnosed young myeloma patients: a pooled analysis. Leukemia. 2017; 31(8):1727-1734. Google Scholar
- Jimenez-Zepeda VH, Mikhael J, Winter A. Second Autologous Stem Cell Transplantation as Salvage Therapy for Multiple Myeloma: Impact on Progression-Free and Overall Survival. Biol Blood Marrow Transplant. 2012; 18(5):773-779. PubMedhttps://doi.org/10.1016/j.bbmt.2011.10.044Google Scholar
- Auner HW, Szydlo R, Rone A. Salvage autologous stem cell transplantation for multiple myeloma relapsing or progressing after upfront autologous transplantation. Leuk Lymphoma. 2013; 54(10):2200-2204. PubMedhttps://doi.org/10.3109/10428194.2013.773998Google Scholar
- Grövdal M, Nahi H, Gahrton G. Autologous stem cell transplantation versus novel drugs or conventional chemotherapy for patients with relapsed multiple myeloma after previous ASCT. Bone Marrow Transplant. 2015; 50(6):808-812. Google Scholar
- Cook G, Ashcroft AJ, Cairns DA. The effect of salvage autologous stem-cell transplantation on overall survival in patients with relapsed multiple myeloma (final results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a randomised, open-label, phase 3 trial. Lancet Haematol. 2016; 3(7):e340-e351. Google Scholar
- Garderet L, Iacobelli S, van Biezen A. Outcome of Third Salvage Autologous Stem Cell Transplantation in Multiple Myeloma. Blood. 2016; 128(22):993. PubMedhttps://doi.org/10.1182/blood-2015-11-680918Google Scholar
- Moreau P, San Miguel J, Sonneveld P. Multiple myeloma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017; 28(suppl_4):iv52-iv61. Google Scholar
- Kumar S, Zhang M-J, Li P. Trends in allogeneic stem cell transplantation for multiple myeloma: a CIBMTR analysis. Blood. 2011; 118(7):1979-1988. PubMedhttps://doi.org/10.1182/blood-2011-02-337329Google Scholar
- Sobh M, Michallet M, Gahrton G. Allogeneic hematopoietic cell transplantation for multiple myeloma in Europe: trends and outcomes over 25 years. A study by the EBMT Chronic Malignancies Working Party. Leukemia. 2016; 30(10):2047-2054. Google Scholar
- Bruno B, Rotta M, Patriarca F. A Comparison of Allografting with Autografting for Newly Diagnosed Myeloma. N Engl J Med. 2007; 356(11):1110-1120. PubMedhttps://doi.org/10.1056/NEJMoa065464Google Scholar
- Garban F, Attal M, Michallet M. 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; 107(9):3474-3480. PubMedhttps://doi.org/10.1182/blood-2005-09-3869Google Scholar
- Moreau P, Garban F, Attal M. Long-term follow-up results of IFM99-03 and IFM99-04 trials comparing nonmyeloablative allotransplantation with autologous transplantation in high-risk de novo multiple myeloma. Blood. 2008; 112(9):3914-3915. PubMedhttps://doi.org/10.1182/blood-2008-07-168823Google Scholar
- Rosinol L, Perez-Simon JA, Sureda A. A prospective PETHEMA study of tandem autologous transplantation versus autograft followed by reduced-intensity conditioning allogeneic transplantation in newly diagnosed multiple myeloma. Blood. 2008; 112(9):3591-3593. PubMedhttps://doi.org/10.1182/blood-2008-02-141598Google Scholar
- Krishnan A, Pasquini MC, Logan B. Autologous haemopoietic stem-cell transplantation followed by allogeneic or autologous haemopoietic stem-cell transplantation in patients with multiple myeloma (BMT CTN 0102): a phase 3 biological assignment trial. Lancet Oncol. 2011; 12(13):1195-1203. PubMedhttps://doi.org/10.1016/S1470-2045(11)70243-1Google Scholar
- Lokhorst HM, van der Holt B, Cornelissen JJ. Donor versus no-donor comparison of newly diagnosed myeloma patients included in the HOVON-50 multiple myeloma study. Blood. 2012; 119(26):6219-6225. PubMedhttps://doi.org/10.1182/blood-2011-11-393801Google Scholar
- Björkstrand B, Iacobelli S, Hegenbart U. Tandem Autologous/Reduced-Intensity Conditioning Allogeneic Stem-Cell Transplantation Versus Autologous Transplantation in Myeloma: Long-Term Follow-Up. J Clin Oncol. 2011; 29(22):3016-3022. PubMedhttps://doi.org/10.1200/JCO.2010.32.7312Google Scholar
- Giaccone L, Storer B, Patriarca F. Long-term follow-up of a comparison of nonmyeloablative allografting with autografting for newly diagnosed myeloma. Blood. 2011; 117(24):6721-6727. PubMedhttps://doi.org/10.1182/blood-2011-03-339945Google Scholar
- Gahrton G, Iacobelli S, Björkstrand B. Autologous/reduced-intensity allogeneic stem cell transplantation vs autologous transplantation in multiple myeloma: long-term results of the EBMT-NMAM2000 study. Blood. 2013; 121(25):5055-5063. PubMedhttps://doi.org/10.1182/blood-2012-11-469452Google Scholar
- Bruno B, Sorasio R, Patriarca F. Unrelated donor haematopoietic cell transplantation after non-myeloablative conditioning for patients with high-risk multiple myeloma. Eur J Haematol. 2007; 78(4):330-337. PubMedGoogle Scholar
- Georges GE, Maris MB, Maloney DG. Nonmyeloablative Unrelated Donor Hematopoietic Cell Transplantation to Treat Patients with Poor-Risk, Relapsed, or Refractory Multiple Myeloma. Biol Blood Marrow Transplant. 2007; 13(4):423-432. PubMedhttps://doi.org/10.1016/j.bbmt.2006.11.011Google Scholar
- Auner HW, Szydlo R, van Biezen A. Reduced intensity-conditioned allogeneic stem cell transplantation for multiple myeloma relapsing or progressing after autologous transplantation: a study by the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2013; 48(11):1395-1400. PubMedhttps://doi.org/10.1038/bmt.2013.73Google Scholar
- Kröger N, Shimoni A, Schilling G. Unrelated stem cell transplantation after reduced intensity conditioning for patients with multiple myeloma relapsing after autologous transplantation. Br J Haematol. 2010; 148(2):323-331. PubMedhttps://doi.org/10.1111/j.1365-2141.2009.07984.xGoogle Scholar
- Kröger N, Badbaran A, Zabelina T. Impact of High-Risk Cytogenetics and Achievement of Molecular Remission on Long-Term Freedom from Disease after Autologous–Allogeneic Tandem Transplantation in Patients with Multiple Myeloma. Biol Blood Marrow Transplant. 2013; 19(3):398-404. PubMedhttps://doi.org/10.1016/j.bbmt.2012.10.008Google Scholar
- Roos-Weil D, Moreau P, Avet-Loiseau H. Impact of genetic abnormalities after allogeneic stem cell transplantation in multiple myeloma: a report of the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire. Haematologica. 2011; 96(10):1504-1511. PubMedhttps://doi.org/10.3324/haematol.2011.042713Google Scholar
- Beitinjaneh AM, Saliba R, Bashir Q. Durable responses after donor lymphocyte infusion for patients with residual multiple myeloma following non-myeloablative allogeneic stem cell transplant. Leuk. Lymphoma. 2012; 53(8):1525-1529. PubMedhttps://doi.org/10.3109/10428194.2012.656635Google Scholar
- Crawley C, Lalancette M, Szydlo R. Outcomes for reduced-intensity allogeneic transplantation for multiple myeloma: an analysis of prognostic factors from the Chronic Leukaemia Working Party of the EBMT. Blood. 2005; 105(11):4532-4539. PubMedhttps://doi.org/10.1182/blood-2004-06-2387Google Scholar
- Ladetto M, Ferrero S, Drandi D. Prospective molecular monitoring of minimal residual disease after non-myeloablative allografting in newly diagnosed multiple myeloma. Leukemia. 2016; 30(5):1211-1214. Google Scholar
- Passera R, Pollichieni S, Brunello L. Allogeneic Hematopoietic Cell Transplantation from Unrelated Donors in Multiple Myeloma: Study from the Italian Bone Marrow Donor Registry. Biol Blood Marrow Transplant. 2013; 19(6):940-948. PubMedhttps://doi.org/10.1016/j.bbmt.2013.03.012Google Scholar
- Donato ML, Siegel DS, Vesole DH. The Graft-Versus-Myeloma Effect: Chronic Graft-Versus-Host Disease but Not Acute Graft-Versus-Host Disease Prolongs Survival in Patients with Multiple Myeloma Receiving Allogeneic Transplantation. Biol Blood Marrow Transplant. 2014; 20(8):1211-1216. Google Scholar
- Michallet M, Sobh M, El-Cheikh J. Evolving strategies with immunomodulating drugs and tandem autologous/allogeneic hematopoietic stem cell transplantation in first line high risk multiple myeloma patients. Exp Hematol. 2013; 41(12):1008-1015. Google Scholar
- Caballero-Velázquez T, López-Corral L, Encinas C. Phase II clinical trial for the evaluation of bortezomib within the reduced intensity conditioning regimen (RIC) and post-allogeneic transplantation for high-risk myeloma patients. Br J Haematol. 2013; 162(4):474-482. PubMedhttps://doi.org/10.1111/bjh.12410Google Scholar
- Kröger N, Zabelina T, Klyuchnikov E. Toxicity-reduced, myeloablative allograft followed by lenalidomide maintenance as salvage therapy for refractory/relapsed myeloma patients. Bone Marrow Transplant. 2013; 48(3):403-407. PubMedhttps://doi.org/10.1038/bmt.2012.142Google Scholar
- Kneppers E, van der Holt B, Kersten M-J. Lenalidomide maintenance after non-myeloablative allogeneic stem cell transplantation in multiple myeloma is not feasible: results of the HOVON 76 Trial. Blood. 2011; 118(9):2413-2419. PubMedhttps://doi.org/10.1182/blood-2011-04-348292Google Scholar
- Alsina M, Becker PS, Zhong X. Lenalidomide maintenance for high-risk multiple myeloma after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2014; 20(8):1183-1189. PubMedhttps://doi.org/10.1016/j.bbmt.2014.04.014Google Scholar
- Htut M, D’Souza A, Bruno B. Survival after Relapse Following Tandem Allogeneic Vs. Tandem Autologous Hematopoietic Cell Transplantation (HCT) for Myeloma (MM). Blood. 2016; 128(22):833. Google Scholar
- Giaccone L, Evangelista A, Patriarca F. Prolonged Follow-up Confirmed a Role for Upfront Tandem Auto-Allo Transplant in Multiple Myeloma Also in the Era of New Drugs. Blood. 2016; 128(22):3469. Google Scholar
- López Corral L, Caballero Velázquez T, López Godino O. Response to Proteosome Inhibitors and Immunomodulatory Drugs before and after Allogeneic Transplantation in Patients with Multiple Myeloma: A Long Term Follow up Study. Blood. 2016; 128(22):3436. Google Scholar
- LeBlanc R, Ahmad I, Terra R. Bortezomib Consolidation after Nonmyeloablative Allogeneic Stem Cell Transplantation Leads to a High Incidence of Immunophenotypic Complete Response in Young and/or High-Risk Multiple Myeloma Patients. Blood. 2016; 128(22):2306. Google Scholar
- Klyuchnikov E, von Pein U-M, Ayuk FA. Daratumumab Is an Effective and Safe Salvage Therapy in Relapsed/Refractory Patients with Multiple Myeloma after Allogeneic Stem Cell Transplantation. Blood. 2016; 128(22):3437. Google Scholar
- Cook G, Carter CR, Brock K. Immune Biomarkers Identify Sustained Quantitative and Functional Immune Reconstitution in the Setting of Adjunctive Lenalidomide Following T-Depleted RIC-Allo SCT for Multiple Myeloma. Blood. 2016; 128(22):4585. Google Scholar
- McKiernan P, Siegel DS, Vesole DH. Long-Term Survival Is Demonstrated in Patients with Multiple Myeloma Treated with Allogeneic Hematopoietic Stem Cell Transplantation in Both the Consolidation and Salvage Settings. Blood. 2016; 128(22):2302. Google Scholar
- Patriarca F, Giaccone L, Onida F. New drugs and allogeneic hematopoietic stem cell transplantation for hematological malignancies: do they have a role in bridging, consolidating or conditioning transplantation treatment?. Expert Opin Biol Ther. 2017; 17(7):821-836. Google Scholar
- Ludwig H, Weisel K, Petrucci MT. Olaptesed pegol, an anti-CXCL12/SDF-1 Spiegelmer, alone and with bortezomib-dexamethasone in relapsed/refractory multiple myeloma: a Phase IIa Study. Leukemia. 2017; 31(4):997-1000. Google Scholar
- Rodríguez-Otero P, Paiva B, Engelhardt M, Prósper F, San Miguel JF. Is immunotherapy here to stay in multiple myeloma?. Haematologica. 2017; 102(3):423-432. PubMedhttps://doi.org/10.3324/haematol.2016.152504Google Scholar
- Garfall AL, Maus MV, Hwang W-T. Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma. N Engl J Med. 2015; 373(11):1040-1047. PubMedhttps://doi.org/10.1056/NEJMoa1504542Google Scholar
- Suck G, Odendahl M, Nowakowska P. NK-92: an “off-the-shelf therapeutic” for adoptive natural killer cell-based cancer immunotherapy. Cancer Immunol Immunother. 2016; 65(4):485-492. Google Scholar
- Jiang H, Zhang W, Shang P. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol Oncol. 2014; 8(2):297-310. PubMedhttps://doi.org/10.1016/j.molonc.2013.12.001Google Scholar
- Tai Y-T, Anderson KC. Targeting B-cell maturation antigen in multiple myeloma. Immunotherapy. 2015; 7(11):1187-1199. PubMedhttps://doi.org/10.2217/imt.15.77Google Scholar
- Yang J, Zhu H, Tan Z. Comparison of two functional kappa light-chain transcripts amplified from a hybridoma. Biotechnol Appl Biochem. 2013; 60(3):289-297. Google Scholar
- Chu J, Deng Y, Benson DM. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014; 28(4):917-927. PubMedhttps://doi.org/10.1038/leu.2013.279Google Scholar
- Casucci M, Nicolis di Robilant B, Falcone L. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013; 122(20):3461-3472. PubMedhttps://doi.org/10.1182/blood-2013-04-493361Google Scholar
- Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016; 127(26):3321-3330. PubMedhttps://doi.org/10.1182/blood-2016-04-703751Google Scholar
- Chu J, Deng Y, Benson DM. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014; 28(4):917-927. PubMedhttps://doi.org/10.1038/leu.2013.279Google Scholar
- Engelhardt M, Ihorst G, Caers J, Gunther A, Wasch R. Autotransplants in older multiple myeloma patients: hype or hope in the era of novel agents?. Haematologica. 2016; 101(11):1276-1278. PubMedhttps://doi.org/10.3324/haematol.2016.154807Google Scholar
- Karlin L, Arnulf B, Chevret S. Tandem autologous non-myeloablative allogeneic transplantation in patients with multiple myeloma relapsing after a first high dose therapy. Bone Marrow Transplant. 2011; 46(2):250-256. PubMedhttps://doi.org/10.1038/bmt.2010.90Google Scholar
- Bashir Q, Khan H, Orlowski RZ. Predictors of prolonged survival after allogeneic hematopoietic stem cell transplantation for multiple myeloma. Am J Hematol. 2012; 87(3):272-276. PubMedGoogle Scholar
- Patriarca F, Einsele H, Spina F. Allogeneic Stem Cell Transplantation in Multiple Myeloma Relapsed after Autograft: A Multicenter Retrospective Study Based on Donor Availability. Biol Blood Marrow Transplant. 2012; 18(4):617-626. PubMedhttps://doi.org/10.1016/j.bbmt.2011.07.026Google Scholar
- Wirk B, Byrne M, Dai Y, Moreb JS. Outcomes of Salvage Autologous Versus Allogeneic Hematopoietic Cell Transplantation for Relapsed Multiple Myeloma After Initial Autologous Hematopoietic Cell Transplantation. J Clin Med Res. 2013; 5(3):174-184. Google Scholar
- Freytes CO, Vesole DH, LeRademacher J. Second transplants for multiple myeloma relapsing after a previous autotransplant-reduced-intensity allogeneic vs autologous transplantation. Bone Marrow Transplant. 2014; 49(3):416-421. PubMedhttps://doi.org/10.1038/bmt.2013.187Google Scholar
- Klyuchnikov E, Wolschke C, Badbaran A. Allogeneic Stem Cell Transplantation As Salvage Therapy for First Relapse after Autografting in Multiple Myeloma Patients. Blood. 2016; 128(22):4619. Google Scholar