Skip to main content
Top

02-10-2016 | Treatment | Book chapter | Article

Transplantation for Multiple Myeloma

Authors: Yogesh S. Jethava, Frits van Rhee

Publisher: Springer International Publishing

Abstract

Multiple myeloma is a disorder characterized by accumulation of malignant plasma cells in the bone marrow, hypercalcemia, monoclonal protein, and end organ damage. Recently newer generation proteosome inhibitors, monoclonal antibodies and novel agents have been approved by FDA, which is undoubtedly increasing life expectancy of the patients. However, hematopoietic stem cell transplantation still remains the cornerstone of the treatment. In this chapter, we are discussing the autologous stem cell transplant, allogeneic stem cell transplant and total therapy trials with outcomes.

1 Introduction

A Surveillance Epidemiology and End Results (SEER) data indicate that the number of new cases of multiple myeloma (MM) was 6.3 per 100,000 men and women per year. Multiple myeloma is the most common indication for autologous stem cell transplantation (AT). AT is still considered the standard of care for multiple myeloma (MM) patients. In the USA, eligibility for hemopoetic stem cell transplant (HSCT) is principally determined by biological fitness rather than age while in Europe, patients are often considered who are 65 years or younger. New chemotherapeutic agents (e.g., bortezomib, thalidomide, lenalidomide) are being incorporated into treatment paradigm for multiple myeloma as a result, outcomes with AT have been improving. Recent studies with long-term follow-up suggest that cure is now feasible for subsets of patients.

2 Advances in Disease Biology and Risk-Adapted Treatment

Several algorithms for the management of myeloma have been suggested. Most of these incorporate cytogenetics and standard prognostic factors such as serum albumin and beta 2 microglobulin [1].
The cytogenetic-based risk stratification model is demonstrated in Table 1.
Table 1
Risk stratification of myeloma based on cytogenetics
High risk
Intermediate risk
Low risk
17p13 deletion
t (14;16)
t (14;20)
LDH ≥ 2 times institutional upper limit of normal
Features of primary plasma cell leukemia
High-risk gene expression profiling signature
t (4;14)
Deletion 13 or hypodiploidy by conventional karyotyping
Trisomies (hyperdiploidy)
t (11;14)
t (6;14)
Apart from the cytogenetic-based risk model, gene expression profiling offers more robust way of risk stratification of MM [2]. So far, GEP is available in large institutions and the disadvantages of GEP include sample attrition (especially in multicenter trials), expense, requirement for stringent quality control, and sample turnaround time, which limits application. The introduction of a “myeloma PCR kit” has negated these issues and is enabling genetic stratification based on biologic disease risk. Based on RT-PCR data of 70 genes, MM patients are divided into standard risk and high-risk groups. High-risk patients do poorly with all current approaches and should be entered into clinical trials exploring novel therapies and combination of novel drugs. It is conceivable that standard or low-risk patients in the elderly population would be amenable to therapeutic approaches aimed at providing long-term disease control.
In subsequent sections we will discuss
  • Early versus late transplant in MM
  • Single autologous transplant
  • Tandem autologous transplants
  • Evidence related to allogeneic transplant

3 Early Versus Late Transplant in the Era of Novel Agents

Novel agents have resulted in increased response rates especially with the triplet therapy. This has raised the question whether AT should be considered early or reserved for relapse. There are not many studies, comparing up-front AT versus salvage AT. There has been one phase 3 study, reported more than 10 years ago, which examined up-front autologous HSCT compared with rescue autologous HSCT at disease progression/relapse and found no difference in the two approaches [3]. Early reports from two phase 3 trials examining chemotherapy versus up-front tandem autologous HSCT have shown an improved PFS without a difference in OS [4, 5]. Table 2 summarizes studies of early autologous transplantation versus standard chemotherapy. Recent genomic studies have shown that MM patients have significant clonal heterogeneity. Hence one could argue that, in younger patients, AT should be applied to maximize cytoreduction followed by consolidation and maintenance therapy with an objective to eliminate myeloma completely or minimize the myeloma burden to prevent the emergence of treatment-refractory clones. Further, the novel therapies carry the risk of generating drug-refractory disease, which may prove difficult to control even with a melphalan-based AT. At present it is difficult to identify patients with genomically unstable disease and therefore with with aggressive subclones and those who do not harbor more aggressive subclones, allowing for more expectant management. It is also important to realize that there is an increased tendency in the field to treat patients, who are not transplanted up-front, until progression prolonging the exposure to drugs and their potential toxicities. A large randomized study US/French study enrolling 1000 patients is currently in progress comparing the outcomes of patients receiving up-front AT and AT at relapse This study will elucidate more information on the timing of AT transplantation strategy. At present, we recommend up-front AT for the transplant eligible patients.
Table 2
Studies of early autologous transplantation
Trial
Induction
Randomization
Results
Gay et al.
N = 402 [6]
Lenalidomide + Dexamethasone
Arm A-MPR
Arm B-tandem AT with melphalan 200 mg/m2
Follow up 45 months from the diagnosis.
PFS for AT superior. 38 versus 26 months (p < 0.0001)
Italian group
N = 389 [7]
RD
Arm A = CRD
Arm B = AT with mel 200
Second randomization = lenalidomide + prednisone versus lenalidomide maintenance
PFS at 2 years = mel arm 72 % and 61 % for CRD arm. (p < 0.001)

4 Single Autologous Transplant Versus Standard Chemotherapy

Melphalan (MEL) was first introduced in 1962 for the therapy of myeloma [4, 5]. McElwain and Powles first reported the efficacy of high-dose MEL in inducing complete biochemical and bone marrow responses in three of nine patients [8, 9]. Barlogie et al. reported that the prolonged myelosuppression due high-dose MEL therapy could be considerably shortened by infusing autologous bone marrow cells [10]. Several prospective, randomized studies in newly diagnosed patients have addressed whether high-dose Mel is superior to standard chemotherapy (Table 3). These include studies conducted by the Intergroupe Francais de Myélome (IFM), the Medical Research Council of the United Kingdom (MRC), the Group Myelome-Autogreffe (MAG), the Programa para el Estudio de la Terapéutica en Hematopatía Maligna (PETHEMA), and the US Intergroup Trial S9321 [1113]. Collectively, the evidence of both retrospective and prospective studies indicates that high dose treatment (HDT) confers a survival benefit in younger patients (<60 years of age).
Table 3
Randomized trials of standard chemotherapy compared to high-dose Mel treatment
Author
Group
N
Age (years)
Median Follow-up (month)
TRM (%)
CR%(STD vs. HDT)
Median EFS (month)
Median OS (month)
Conclusion
Attal et al. [14]
IFM 90
200
≤65
37 STD, 41 AT
5 versus 7
5 versus 22
18 versus 22
44 versus 57
Benefit in EFS and OS
Child et al. [15]
MRC7
401
≤64
32 STD 40AT
 
8 versus 44
20 versus 32
42 versus 54
Benefit in EFS and OS
Blade et al. [12]
PETHEMA
164
≤65
56
3.6 versus 3.7
11 versus 30
33 versus 42
66 versus 61
No benefit
Palumbo [16]
M97G
194
50–70′
39 STD 41 AT
0 versus 2.1
6 versus 25
16 versus 28
43 versus NR
Benefit in EFS and OS
Fermand et al. [11]
MAG
190
55–65
120
2.1 versus 5.3
4 versus 6
19 versus 25
48 versus 48
Benefit in EFS, but not OS
Segeren et al. [8]
HOVON
261
<65
33
1.3 versus 5.2
13 versus 29
21 versus 22
50 versus 47
No benefit in EFS or OS
Barlogieet al. [13]
USIG
516
<70
76
0.4 versus 3.4
15 versus 17
22 versus 25
54 versus 62
No benefit in EFS or OS
In IFM90 study involving patients younger than 65 years of age, HDT arm had shown higher CR and VGPR rates (38 vs 14 % respectively) [14]. The 5-year OS and EFS rates in the HDT arm were superior (52 vs. 12 %; p = 0.03 and 28 vs. 10 %; p = 0.01) and the best results were seen in patients <60 years with 5-year OS of 70 %. In MRCVII study of 407 patients <65 years, CR rates of 44 versus 8 % (p < 0.001) were reported. HDT with MEL 200 mg/m2 not only increased median survival by almost 1 year (54.1 vs. 42.3 months) but also significantly improved time to progression in this trial.
Few trials such as MAG, PETHEMA, and the US Intergroup studies did not show a definite benefit in terms of OS for MEL-based HDT. This can be explained by several factors. In the US Intergroup Trial, CR rates were similar (17 vs. 15 %) between high-dose MEL arm and standard chemotherapy arm however the conditioning used was reduced dose of Mel 140 mg/m2 and 8 Gy of total body irradiation (TBI). The dose if MEL is certainly inferior to MEL 200 mg/m2 which was used in historical and randomized trials. Also in this trial, the large percentage of patients in the standard chemotherapy arm crossed over to MEL arm, making it difficult to interpret OS [1719].
The MAG study compared VMCP chemotherapy and high-dose treatment, comprising MEL 200 or MEL140 mg/m2 with busulfan 16 mg/kg in patients between 55 and 65 years old. There was a trend to better EFS in the high-dose arm at a median follow-up at 10 years but the OS survival was identical at 48 months in both the arms. However, it is important to note that 22 % of patients in the STD arm received salvage HDT, which may have contributed to equalizing OS in both the arms in this trial.
In the PETHEMA study, randomization to high-dose chemotherapy or standard chemotherapy was not performed at diagnosis. Also, the therapy delivered in the standard arm, comprised of 12 cycles of VBMP/VBAD, which is considerably heavy and it mitigated any survival benefit conferred by HDT [20].
In general, most investigators would agree that single MEL-based high-dose treatment is superior to standard chemotherapy in patients less than 65 years [19].

5 Tandem Autologous Transplant

The role of tandem autologous transplants is explored in various total therapy trials and European trials.

5.1 Total Therapies

Total Therapy I, trial intended to increase the frequency and duration of complete response, thus extending OS [9]. The concept was inspired by the St Jude’s Children’s Hospital Total Therapy (TT) programs for acute leukemia which have made unprecedented progress in curing children with both acute lymphoblastic and myeloid leukemias. It was recognized in adult acute leukemia that cures were only obtained when a CR rate of ≥40 % was accomplished. A single MEL 200 AT resulted in a CR rate of usually no more than 20 % [10, 21, 22]. Thus, using CR as a substitute for survival, the underlying hypothesis was that a more marked increase in CR rate, from less than 5 % with standard MEL-prednisone (MP) to 40 % would produce significant prolongation of EFS and OS, and perhaps attain cure [5]. These observations led to initiation of Total Therapy Trials in MM.
The schema for TT1, TT2, and TT3 trials is already published (Figs. 1 and 2). The results of TT4, TT5, and TT6 trials are still awaited. The long-term results of TT1, which enrolled 231 patients, were published and the median follow-up was an unprecedented 12 years [23]. At 10 and 15 years, respectively, 33 and 17 % of patients are alive; 15 and 7 % are event free; and 18 and 12 % of those achieving CR remain in uninterrupted remission.
The successor Phase III trial, TT2 with 668 enrollees, delivered more intense treatment by intensifying remission induction, by adding consolidation chemotherapy post-tandem AT, and by providing high-dose pulsed dexametasone during maintenance with INF-α [24]. In addition, patients were randomized to receive thalidomide from the outset until disease progression or adverse events.
The thalidomide group had significantly higher CR rates and 5-year EFS compared to the control group (62 vs. 43 %, and 56 vs. 44 %, respectively). However, the 5-year OS was approximately 65 % in both arms of the study, which could be explained by worse post-relapse survival in the thalidomide arm, suggesting that continuous exposure to thalidomide may promote drug resistance.
EFS and OS were adversely influenced by calcium (CA), elevated LDH and albumin <3.5 g/dl.
The non-thalidomide arms in TT2 and TT1 were recently compared in order to examine the potential benefit conferred by dose-intensified induction chemotherapy and post-tandem AT consolidation chemotherapy applied in TT2 without having thalidomide as a confounding variable [25]. The CR rates in both trials were similar at 41 and 43 %. However, the 5-year estimates of continuous CR (45 vs. 32 %), and 5-year EFS (43 vs. 28 %) were significantly superior in TT2, with a trend to improved OS (62 vs. 57 %). Patients who achieved CR in the first year and had tandem AT within 1 year also had superior OS.
The treatment-related mortality was similar in both trials at approximately 7 %. These data indicate that, overall, TT2 without thalidomide was superior to TT1. Since CA is an important prognostic factor, the outcome was examined in both trials in patients with good-risk (normal CA) and high-risk features (abnormal CA). TT2 especially benefited the two-thirds of good-risk patient who entered CR more frequently and had a longer duration of CR with superior EFS and OS. Thaldomide conferred benefit to patients with a standard risk GEP and abnormal metaphase cytogenetics; a surprising finding, which has not been satisfactorily explained. The TT3 trial combined bortezomib with DTPACE portion as induction and consolidation. The initial results of TT3 trial are, respectively, with an unprecedented 80 % of patients remaining in CR at 2 years with a treatment-related mortality of only 5 %.
These TT trials demonstrated that it is feasible in myeloma to achieve a long-term survival of >30 % at 10 years with tandem transplants with high-dose MEL. This also sets a new yardstick against which recently developed newer drugs should be tested.

5.2 European Randomized Studies

In the IFM 94 study, French intergroup compared single transplant with MEL 140 with tandem transplants. In tandem transplant arm comprised of conditioning with MEL 140 and MEL 140/TBI with the first and second transplant, respectively [26]. A superior 7-year probability of EFS and OS in the tandem AT arm (20 vs. 10 % and 42 vs. 21 %, respectively) was observed.
In Bologna, 96 studies showed that tandem AT significantly increased the probability to achieve CR from 33 to 47 % and extended the 5-year EFS from 29 to 17 % [27]. Tandem AT prolonged EFS by approximately 1 year and was related to a higher CR rate. Twenty percent of patients who failed to achieve (n)CR post after the first transplant did so after the second transplant. Patients who were sensitive to conventional chemotherapy with VAD achieved a CR rate of 73 % with double AT, versus 52 % in the single AT group. Conversely, the CR rates in the single and tandem AT arms were similar in patients’ refractory to VAD, suggesting that resistance to conventional chemotherapy was difficult to overcome even by double AT. There was no benefit in terms of OS, which was similar after single and double AT (46 vs. 43 %). The 2-year OS from relapse in the single transplant arm was longer than in the second transplant arm (62 vs. 51 %), which can be explained by the sequential application of salvage therapies. Approximately one-third of the patients received a second, unplanned AT, obscuring the survival benefit conferred by a second planned procedure. In addition, half were treated with novel agents such as thalidomide and bortezomib, which may have the potential to reverse resistance to HDT.
The French MAG 95 used a two × two design in 230 patients younger than 56 years to study the effects of both tandem AT and purging of the autograft using CD34+ selection. Tandem AT using unselected CD34+ cells resulted in improved OS compared to single AT. CD34+ cell selection did not confer survival benefit and was associated with an increased risk of infectious complications [28].
Table 4 summarizes the studies of tandem transplants in MM patients.
Table 4
Randomized trials of single versus double AT
Group
N
Age
Median Follow-up (Mo)
Regimen
CR% (single vs. double)
Median EFS (mo) (single vs. double)
Median OS (single vs. double)
Conclusion
IFM 94 [25]
339
60
75
MEL 140 + TBI and MEL 140
42 versus 50
25 versus 30
48 versus 58
OS/EFS better in double AT
MAG 95 [27]
227
55
53
MEL 140 + CCNU/VP 16/Cy/TBI versus MEL 140 → MEL 140 + TBI
39 versus 37
31 versus 33
49 versus 73
No diff in EFS. OS is better with double AT
Bologna 96 [29]
228
60
55
MEL 200 → MEL 120 + BU 12
35 versus 48
25 versus 35
59 versus 73
OS/EFS better in double AT
GMMG HD2 [30]
210
65
NR
MEL 200 → MEL 200
Not available
23 versus 29
No difference
EFS better with double AT
We recommend that tandem transplant should not be done outside the context of clinical trials. The studies show that tandem AT is well tolerated by younger patients with good performance status. It has acceptable treatment-related morbidity and mortality. Patients who appear to benefit most from a second AT procedure are those who are not in (n)CR after the first AT. Patients with aggressive disease, e.g., abnormal CA, do not seem to do better with tandem AT.

6 Autologous Stem Cell Transplant

6.1 Collection and Processing of Stem Cells

Alkylating agents such as melphalan and the immunomodulatory lenalidomide can impair the collection of hemopoeitic stem cells (HSC). Melphalan is best completely avoided, whilst lenalidomide exposure should be limited to four cycles prior to collection. HSC are typically collected by apheresis from the peripheral blood after stimulation with granulocyte colony-stimulating factor (G-CSF). Peripheral blood progenitor cells (PBPCs) are preferable to bone marrow cells for transplantation due to quicker engraftment and a potential for less contamination of the infused cells with tumor cells. G-CSF alone and G-CSF plus cyclophosphamide are the most common regimens used for stem cell mobilization [3133]. The usual dose of G-CSF is 10 mcg/kg per day subcutaneous. Cyclophosphamide is usually administered at a dose of 1.5–3 g/m2 intravenous for 1 or 2 days. The choice between the two methods is mainly dependent on institutional preference and experience. There are no randomized trials comparing the two approaches. Some institutions use G-CSF plus cyclophosphamide as their standard. G-CSF plus cyclophosphamide has the advantage of providing much higher PBPCs than G-CSF alone, but also carries the risk of longer time to start collection and the risk of neutropenia. Plerixafor is a chemokine receptor type 4 inhibitor (CXCR4) and it impairs the binding of hematopoietic stem cells within the bone marrow microenvironment [34]. Plerixafor is by some, primarily reserved for patients who fail stem cell collection with either G-CSF or G-CSF plus cyclophosphamide, while some use pleraxifor up-front usually together with G-CSF [35, 36]. Once the mobilization regimen is initiated, patients are monitored by peripheral blood CD34 counts. Apheresis is started when the peripheral CD34+ counts reach 20 CD34 cells/microL. Apheresis is performed with a goal of collecting between at least minimum 3 × 106 CD34+ to 6 × 106 CD34+ cells/kg. A minimum of 2 × 106 CD34+ cells/kg is considered essential for one transplant. We routinely collect >20 × 106 CD34+ cells/kg. Infusion of a larger number of stem cells will hasten recovery, minimize blood product use, and reduce area of CRP under the curve, the latter being a surrogate marker for infectious complication. This leaves ample cells for use as salvage transplant or for reestablishing hematopoiesis if multiple lines of salvage therapy have exhausted hematopoiesis. In general, enough PBPCs are harvested for two transplantations. PBPCs are cryopreserved in 5 % dimethylsulfoxide to be thawed at the bedside at the time of infusion [16].

6.2 Preparatory Regimen

The standard conditioning regimen used for AT in multiple myeloma is melphalan at a dose of 200 mg/m2, with dose reductions based on age and renal function.
The use of this dose is primarily based upon two randomized trials that have compared melphalan 200 mg/m2 with a lower dose of melphalan in conditioning for HCT. The French cooperative study compared melphalan 140 mg/m2 plus 8 Gy total body irradiation versus Melphalan 200 mg/m2 in 282 newly diagnosed patients. Patients randomly assigned to melphalan 200 mg/m2 had faster hematologic recovery, less mucositis, less transfusion requirements and, shorter hospitalizations. In this study, survival at 45 months was significantly better in patients receiving melphalan 200 mg/m2 (66 versus 46 %).The Italian study comparing melphalan 200 mg/m2 with melphalan 100 mg/m2 as preparative regimens for HCT in 298 newly diagnosed symptomatic patients <65 years of age [37], patients who received melphalan 200 m/m2 had significantly longer median progression-free survival (31.4 versus 26.2 months) and a trend towards improved projected overall survival at 5 years (62 versus 48 %). Higher percentage of gastrointestinal toxicity (11 versus 1 %), mucositis (17 versus 3 %), need for intravenous broad-spectrum antibiotics (41 versus 29 %), and need for platelet transfusions (56 versus 38 %) was noted in melphalan 200 mg/m2 group. In two other randomized studies, the use of more intensive preparative regimens, such as thiotepa, busulfan, and cyclophosphamide [38] or high-dose idarubicin, cyclophosphamide, and melphalan [39] did not result in better outcomes than melphalan at a dose of 200 mg/m2. Dose adjustments for melphalan for obese patients can lead to a lower dose of melphalan when calculated per kg and could potentially lead to under treatment.

6.3 Special Circumstances

(a) Patients with renal insufficiency—Randomized trials that have shown benefit with HCT compared with chemotherapy have mainly studied patients with serum creatinine <2.0 mg/dL. The data for melphalan use in patients with serum creatinine >2.0 mg/dL comes from a retrospective review of 81 patients with MM and renal failure (plasma creatinine >2 mg/dL) who underwent autologous HCT [40]. Sixty patients who received melphalan 200 mg/m2, excessive toxicity was noted in patients than the subsequent 21 patients who received melphalan 140 mg/m2. The patients who received melphalan 200 mg/m2 had significantly higher rates of severe pulmonary toxicity (57 versus 17 %) and mucositis (93 versus 67 %). In the patients with creatinine >2.0 mg/dl, the treatment-related mortality, event-free, and overall survival were not significantly different between melphalan 200 mg/m2 and melphalan 140 mg/m2.
(b) Older adults—In order to reduce toxicity in patients over 65 years of age, various strategies are studied. For example, once two sequential courses of an intermediate dose of melphalan (100 mg/m2) followed by hematopoietic stem cell rescue (MEL100) has been studied as an alternative to high-dose melphalan 200 mg/m2. In a multicenter trial of 194, in the subgroup of 80 patients aged 65–70, higher rates of near complete plus partial remissions (47 versus 16 %), longer median event-free survival (28 versus 16 months), and overall survival (58 versus 37 months) were observed in patients receiving melphalan 100 mg/m2. However, higher short-term toxicity (e.g., mucositis, fever, need for red cell, or platelet transfusions) was noted. The French Intergroupe Francophone du Myelome (IFM) group reported on 447 previously untreated patients with myeloma patients aged 65–75 years who received melphalan, prednisone, and thalidomide (MPT) or melphalan and prednisone alone (MP) or tandem autologous HCT using MEL100 [41]. There was no difference in median overall survival between the MP and transplant arms. This study suggests that an intermediate dose of melphalan may not be the optimal conditioning regimen. At present, we use melphalan 200 mg/m2 as standard conditioning regimen for HCT myeloma patients and reduce the dose to 140 mg/m2 in patients with renal impairment or those over the age of 65.

6.4 Care During Transplantation

Autologous hematopoietic cell transplantation (HCT) has been performed in both the inpatient and outpatient settings. Approximately 24 h after completion of the preparative chemotherapy, peripheral blood progenitor cells (PBPCs) are reinfused. A period of pancytopenia follows. Red blood cell and platelet transfusions are administered as necessary while hematopoietic colony-stimulating factors (i.e., G-CSF) are used to speed neutrophil engraftment. Neutrophil engraftment usually occurs by day 10–11 and platelet counts are expected to recover to greater than 20,000 by day 16 [42]. Red blood cell transfusion requirements during autologous HCT are usually minimal. HCT without transfusion support, although not ideal has recently been reported in a series of 50 Jehova’s witnesses with acceptable toxicity [43]. At our institution, approximately 80–90 % of patients undergo autologous transplantation entirely as an outpatient, with daily monitoring until full engraftment has occurred [44]. Patients who undergo HCT are at risk for bacterial, viral, and fungal infections, the time course of which varies in the post-transplant period, according to the degree of immune deficiency and cytopenia induced by the transplantation procedure. Approximately 40 % of patients with multiple myeloma undergoing autologous HCT will experience febrile neutropenia [45]. As a result, prophylactic therapies to prevent infection including antiviral and antifungal drugs are recommended during the period of increased risk. In addition, all markers of potential infection must be investigated thoroughly.

6.5 Maintenance After AT

The absence of a plateau on survival curves post HDT and AT with ongoing relapses justifies the exploration of maintenance strategies with the prolongation of response duration as the goal. In addition, AT may result in a very low tumor burden, which may be susceptible to long-term suppression by drugs or immunomodulatory agents. A number of agents have been or are being explored for maintenance, including interferon alpha (IFN-α), corticosteroids, biphosphonates, thalidomide, lenalidomide, bortezomib, and combinations of these drugs. Several studies show that consolidation and maintenance post AT can further reduce tumor burden and improve outcome.
IFN was one of the first drugs to be studied as maintenance post AT. It yielded a small benefit in a meta-analysis from the European Group for Blood and Marrow Transplantation (EBMT) [46]. The study by Australian myeloma group (with a median follow-up 5.4 years), thalidomide administered in combination with prednisolone for 1 year was found to significantly prolong PFS and OS compared with prednisolone alone: 5-year PFS was 27 % for Thal/Pred versus 15 % for Pred, P = 0.005; 5-year OS was 66 versus 47 %, respectively, P = 0.007) [47]. Minimum thalidomide exposure required was at least 8 months to gain a PFS and OS advantage and there was no difference between the two arms regarding overall response rate to salvage therapy or post relapse. This suggested that acquired resistance in this study was not an important issue for thalidomide-treated patients.
In the meta-analysis of five trials involving patients, Morgan et al. found a significant late OS benefit for thalidomide (P < 0.001, 7-year difference hazard ratio (HR) = 12.3; 95 % confidence interval, 5.5–19.0). Similarly, Lenalidomide has been investigated in the post-transplant setting in three large studies. In the study conducted by the IFM group, patients were randomized to lenalidomide maintenance until progression or no maintenance following a single or tandem ASCT step and two cycles of lenalidomide consolidation in both arms [48]. The lenalidomide was stopped at a median of 2 years (range 1–3 years) due to concerns regarding second primary malignancies. With a median follow-up of 67 months from randomization, the PFS for patients who had received lenalidomide maintenance was significantly longer than for those who had not received any maintenance therapy (46 versus 24 months, P < 0.001) [49]. Although OS did not achieve statistical significance in two arms (82 versus 81 months, respectively, P = 0.8), the second PFS and survival after the first progression were shorter in the lenalidomide maintenance arm. In addition, the cumulative incidence of second primary malignancy was significantly higher with lenalidomide. When examining cytogenetic risk, progression was superior for the lenalidomide arm for patients with or without 13q deletion and without t(4;14) or 17p deletion, but did not reach significance for patients with either t(4;14) or 17p deletion. The CALGB conducted a large placebo-controlled randomized study of lenalidomide maintenance following ASCT [50]. In this study, lenalidomide was administered until disease progression. 86 of 128 eligible patients received lenalidomide maintenance. At a median follow-up of 34 months, 37 % who received lenalidomide maintenance and 58 % who received placebo had disease progression or had died. The median time to progression was 46 months in the lenalidomide group and 27 months in the placebo group (P < 0.001). The median OS in both arms had not been reached, with 85 % of the lenalidomide-arm patients and 77 % of the placebo-arm patients who were alive at the time of the analysis having died (P < 0.03). The rate of second primary malignancy appeared to plateau at 2 years post SCT. This study was updated in 2013. At median follow-up of 48 months, the intent-to-treat analysis demonstrated that the OS was 80 % for the lenalidomide arm and 70 % for the placebo group (P = 0.008) with a continued PFS advantage in the lenalidomide arm. In the study by GIMEMA group, at a median follow-up of 51.2 months, the median PFS was superior with lenalidomide maintenance (PFS: 41.9 months for lenalidomide versus 21.6 months for placebo, P < 0.0001), but 3-year OS was not significantly prolonged (3-year OS: 88.0 % versus 79.2 %, respectively, P = 0.14).
Bortezomib maintenance therapy has been investigated in two phase 3 studies, the HOVON and GMMG studies. A landmark analysis from the start of maintenance showed that OS was superior for patients on the bortezomib arm, while PFS was comparable. The limitation of these studies was that, while the results of these trials confirmed the important role of bortezomib-containing regimen in the treatment of newly diagnosed MM, the design of the trials made it difficult to interpret the role of bortezomib maintenance difficult as it was not possible to delineate the individual contributions of induction versus the maintenance treatment as part of this design. Furthermore, the OS benefit was seen only for patients with del17p cytogenetic abnormalities and those who presented in renal failure. In a large randomized phase 3 study conducted by the Spanish myeloma group, the combination of bortezomib and thalidomide was compared to thalidomide and to Interferon as maintenance therapy and a significant benefit in PFS was found for the VT combination with a median follow-up of 34.9 months [15].

7 Importance of MRD

7.1 Is Complete Remission (CR) Important?

In the context of AT trials for myeloma, disappearance of the M-protein is generally thought to play a pivotal role in predicting superior survival [9, 14, 18, 47, 49, 51]. However, several observations have challenged the notion that a CR is an absolute requirement for prolongation of survival in myeloma. The Mayo Clinic has reported that patients who underwent AT have similar PFS and OS irrespective of whether they achieved CR [52]. Patients with CA in TT2 had similar CR rates of 40 % compared to those without CA, yet their median survival was significantly shorter post high-dose MEL therapy. A similar observation applied to the thalidomide arm in TT2 where a higher CR rate and EFS did not translate into superior survival. The median time to CR in TT2 was 12 months despite the application of an intensive therapeutic program [24] In addition, the disappearance of MRI-defined focal lesions (FL), which are potential sites for surviving resistant myeloma cells, lags 2 years behind the disappearance of M-protein from blood and urine [53]. Paradoxically more aggressive disease features such as CA, elevated LDH, and IgA isotype are predictors of CR [25]. We have recently reported that high serum free light chain levels at diagnosis and a rapid reduction after induction therapy are predictive for achieving CR [54]. These parameters reflect more proliferative myeloma which, on the one hand, is inherently more sensitive to combination chemotherapy, and on the other is linked to shorter EFS and OS due to rapid myeloma regrowth when tumor reduction is insufficient.
Conversely, patients with myeloma evolving from monoclonal gammopathy of unknown significance (MGUS) or with smoldering myeloma have significantly lower CR rates, yet equal EFS and OS compared to patients with de novo myeloma. The lower CR rate in these patients is likely to be due to a lower proliferative rate, reflecting more indolent disease, which is less susceptible to eradication by both standard- and high-dose chemotherapy. Interestingly, these patients also have fewer MRI-defined FL. Patients with MGUS-like myeloma have favorable clinical characteristics, with lower CR rates yet superior survival compared to patients with non-MGUS-like myeloma. Attainment of CR in myeloma is often a gradual and cumulative process. It appears, therefore, that achieving a CR is only critical for prolonging survival in a truly high-risk group of myeloma that thus far can only be captured by gene expression profiling [55]. These data also suggest that the use of CR as a surrogate marker for OS and EFS in clinical trials of novel agents cannot be applied without characterizing myeloma at the molecular level and should be used with extreme caution.

7.2 MRD Detection by Flow Cytometry

Results of multicolor flow cytometry and deep-sequencing studies suggest that among patients achieving a complete response, MRD-negative status is associated with significant improvements in PFS and OS. The UK Medical Research Council IX trial assessed the importance of achieving MRD. There was a significant improvement in OS for each log depletion in MRD level. The median OS was 1 year for ≥10 %, 4 years for 1 to <10 %, 5.9 years for 0.1 to <1 %, 6.8 years for 0.01 to <0.1 %, and more than 7.5 years for <0.01 % MRD level. The detection of minimal residual disease MRD in myeloma using a 0.01 % threshold (10(−4)) after treatment is an independent predictor of PFS [56].

8 Allogeneic Transplantation for MM

8.1 Introduction

Allogeneic hematopoietic cell transplantation (HCT) is potentially the curative treatment for multiple myeloma. However its use is limited because of high rate of treatment-related mortality and its efficacy compared with autologous HCT is not fully established. With the advent of new proteasome inhibitors and IMiDs, role of allogeneic transplant needs to be scrutinized. In subsequent sections, we will review the role of allogeneic transplant in MM.

8.2 Myeloablative HCT

Myeloablative transplants require that patients receive high-dose chemotherapy with or without total body radiation, followed by donor stem cell infusion. Myeloablative transplants are not the preferred modality and are very infrequently performed. Hence this is not discussed further in this chapter.

8.3 Syngeneic HCT

Syngeneic allogeneic transplants are from the identical twin donor and only a limited number of syngeneic transplants have been performed in multiple myeloma. The EBMT analyzed 25 syngeneic transplants and compared the results with 250 patients who underwent either autologous or allogeneic HCT (n = 125, each) HCT [57]. The TRM was substantially lower with only two patients dying due to transplant-related toxicity as compared to myeloablative allogeneic transplant. Overall survival for the patients undergoing syngenic transplants was 73 months, significantly better than that of the case-matched autologous transplants (44 months); both groups outperformed the allogeneic transplants. The Seattle Marrow Transplant Team performed syngeneic transplants on five patients with multiple myeloma [58]. Four patients responded to therapy, while one patient died one month after transplant from cytomegalovirus-associated interstitial pneumonitis. Response durations for these four patients were 6, 17, 18, and more than 72 months. These reports are useful in exploring the various treatment options but do not support the use of syngeneic HCT in place of an autologous HCT if a twin donor is available.

8.4 Non-myeloablative Allogeneic HCT

The results of non-myeloablative T-cell depleted transplants have been summarized in Table 5. Longer-term follow-up of these studies is awaited. It is unclear what impact non-myeloablative HCT will have given the improved outcomes with autologous HCT after the introduction of novel agents utilized in induction, consolidation, and maintenance, in the era of novel agents. In current scenario, non-myeloablative-allogeneic transplantation remains investigational and its role needs to be validated in the era of novel agents.
Table 5
Summary of non-myeloablative T-cell depleted transplants
Study
Previous AT
Results
EBMT [59]
N = 413
44.6 % had undergone two or more prior autologous HCTs
Median PFS—9.6 months
Median OS—24.7 months
5-year survival rate—30 %
Bacigalupo et al. [60]
N = 33 had one or more than one previous autologous transplant
CR in 62 %
TRM = 13 %
Median follow-up 22 months
PFS = 45 % and OS = 37 %
Shimoni et al. [61]
N = 50
Non relapse mortality = 26 %
PFS = 26 % and OS = 34 %
Median follow-up time of 6.4 years

8.5 Role of Auto/Allo Transplants

The role of auto–allo versus tandem autologous transplant is debatable. A meta-analysis of 1822 patients with previously untreated myeloma comparing double autologous HCT versus a single autologous HCT followed by non-myeloablative allogeneic HCT [62], allogeneic HCT was associated with greater treatment-related mortality (relative risk [RR] 3.3; 95 % CI 2.2-4.8 (RR 1.4; 95 % CI 1.1–1.8), but similar overall survival at and beyond 36 months.
The following is a brief summary of the largest trials conducted in this setting
BMT CTN Trial [63]:
  • 625 patients with standard risk myeloma in this trial, 189 with an HLA-identical sibling donor were assigned to receive a myeloablative autologous HCT followed by a non-myeloablative allogeneic HCT.
Hovon 50 [64]:
  • 260 patients with autologous transplant-randomly assigned to RIC sib allo (n = 122 patients) or maintenance with thalidomide or interferon alpha.
  • Median follow-up 77 months.
  • When compared with maintenance therapy, allogeneic HCT was associated with similar rates of progression-free (28 versus 22 % respectively) and overall (55 %) survival at 6 years.
French IFM [65]:
  • Patients with high-risk myeloma (deletion 13 by FISH or an elevated beta-2 microglobulin level) were included in this trial.
  • No benefit with autologous HCT followed by reduced-intensity allogeneic HCT compared with tandem autologous HCT [16].
Spanish PETHEMA [15]:
  • Prospectively compared the use of autologous (85 patients) or reduced-intensity allogeneic (25 patients) HCT in 110 patients with myeloma who failed to achieve a complete or near complete remission after an initial autologous HCT.
  • At a median follow-up after second transplantation of 5.2 years of patients who received an allogeneic HCT, there was no difference in event-free or overall survival rates.
However, the Italian study of 162 patients showed better event-free survival in patients who underwent sib matched allogeneic transplant [66].
The authors of this chapter noted that hardly any genetic data was included in these trials. Hence in the era of genomically driven personalized medicine, the role of auto/allo remains investigational and the data needs to be carefully analyzed.

8.6 Future Strategies

It is important to recognize that MM is complex disease with significant clonal heterogeneity. Transplantation helps in early reduction of clonal diversity and increases the likelihood of cure. Long-term follow-up studies show that AT can achieve profound cytoreduction and likely cure a portion of patients even before the introduction of novel drugs. Total Therapy 1 (TT1), the first tandem AT trial for myeloma, enrolled 231 patients; with a median follow-up of 17 years, 23 remain alive and progression-free with a plateau on the overall survival (OS) curve appearing around 14 years [23]. Martinez-Lopez et al. reported on 344 patients who received AT between 1989 and 1998 who had a median follow-up of 153 months. A plateau in OS appeared after 11 years and, with a further follow-up of 5 years, no relapses were observed [67]. Targeted therapy has already demonstrated some efficacy, but may merely select for resistant subclones, unless a given mutation drives the disease as recently has been described for KRAS/NRAS activating mutations. High-risk myeloma even with modern genetic analysis will likely remain a challenging disease to treat in years to come. AT, tandem autotransplants, autotransplant followed by reduced-intensity allogeneic transplant and allogeneic transplant will remain an important option in the therapeutic armamentarium of myeloma. Novel immune therapeutic agent anti-CD38 antibodies, i.e., daratumomab, checkpoint blockade inhibitors, cellular therapies, and vaccines will likely become available very soon and this can enhance the anti-myeloma response without transplant-associated risks. It is conceivable that such strategies will be complimentary and not replacement for transplant.
Literature
1.
Stewart AK, Bergsagel PL, Greipp PR, Dispenzieri A, Gertz MA, Hayman SR et al (2007) A practical guide to defining high-risk myeloma for clinical trials, patient counseling and choice of therapy. Leukemia 21(3):529–534CrossRefPubMed
2.
Zhan F, Hardin J, Kordsmeier B, Bumm K, Zheng M, Tian E et al (2002) Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood 99(5):1745–1757CrossRefPubMed
3.
Fermand JP, Ravaud P, Chevret S, Divine M, Leblond V, Belanger C et al (1998) 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 92(9):3131–3136PubMed
4.
Bergsagel DE, Ross SW, Baker DT (1962) Evaluation of new chemotherapeutic agents in the treatment of multiple myeloma. V. 1-Aminocyclopentanecarboxylic acid (NSC-1026). Cancer Chemother Rep Part 1 21:101–106
5.
Alexanian R, Haut A, Khan AU, Lane M, McKelvey EM, Migliore PJ et al (1969) Treatment for multiple myeloma. Combination chemotherapy with different melphalan dose regimens. JAMA 208(9):1680–1685CrossRefPubMed
6.
Gay F, Hajek R, Diramondo F et al (2013) Cyclophosphamidelenalidomide-dexamethasone vs autologous transplant in newly diagnosed myeloma: a phase 3 trial. Clin Lymphoma Myeloma Leuk 13(suppl 1):S40
7.
Boccadoro M, Cavallo F, Gay F, Di Raimondo F, Nagler A, Montefusco V (2013) Melphalan/prednisone/lenalidomide (MPR) versus high-dose melphalan and autologous transplantation (MEL200) plus lenalidomide maintenance or no maintenance in newly diagnosed multiple myeloma (MM) patients. J Clin Oncol 31(suppl):8509
8.
Segeren CM, Sonneveld P, van der Holt B, Vellenga E, Croockewit AJ, Verhoef GE et al (2003) Overall and event-free survival are not improved by the use of myeloablative therapy following intensified chemotherapy in previously untreated patients with multiple myeloma: a prospective randomized phase 3 study. Blood 101(6):2144–2151CrossRefPubMed
9.
Barlogie B, Jagannath S, Desikan KR, Mattox S, Vesole D, Siegel D et al (1999) Total therapy with tandem transplants for newly diagnosed multiple myeloma. Blood 93(1):55–65PubMed
10.
Barlogie B, Alexanian R, Dicke KA, Zagars G, Spitzer G, Jagannath S et al (1987) High-dose chemoradiotherapy and autologous bone marrow transplantation for resistant multiple myeloma. Blood 70(3):869–872PubMed
11.
Fermand JP, Katsahian S, Divine M, Leblond V, Dreyfus F, Macro M et al (2005) 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: Official J Am Soc Clin Oncol 23(36):9227–9233CrossRef
12.
Blade J, Rosinol L, Sureda A, Ribera JM, Diaz-Mediavilla J, Garcia-Larana J et al (2005) 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 106(12):3755–3759CrossRefPubMed
13.
Barlogie B, Kyle RA, Anderson KC, Greipp PR, Lazarus HM, Hurd DD et al (2006) Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321. J Clin Oncol: Official J Am Soc Clin Oncol 24(6):929–936CrossRef
14.
Attal M, Harousseau JL, Stoppa AM, Sotto JJ, Fuzibet JG, Rossi JF et al (1996) A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med 335(2):91–97CrossRefPubMed
15.
Rosinol L, Oriol A, Teruel AI, Hernandez D, Lopez-Jimenez J, de la Rubia J et al (2012) Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood 120(8):1589–1596CrossRefPubMed
16.
Berz D, McCormack EM, Winer ES, Colvin GA, Quesenberry PJ (2007) Cryopreservation of hematopoietic stem cells. Am J Hematol 82(6):463–472CrossRefPubMedPubMedCentral
17.
Moreau P, Facon T, Attal M, Hulin C, Michallet M, Maloisel F et al (2002) Comparison of 200 mg/m(2) melphalan and 8 Gy total body irradiation plus 140 mg/m(2) melphalan as conditioning regimens for peripheral blood stem cell transplantation in patients with newly diagnosed multiple myeloma: final analysis of the Intergroupe Francophone du Myelome 9502 randomized trial. Blood 99(3):731–735CrossRefPubMed
18.
Desikan KR, Tricot G, Dhodapkar M, Fassas A, Siegel D, Vesole DH et al (2000) Melphalan plus total body irradiation (MEL-TBI) or cyclophosphamide (MEL-CY) as a conditioning regimen with second autotransplant in responding patients with myeloma is inferior compared to historical controls receiving tandem transplants with melphalan alone. Bone Marrow Transplant 25(5):483–487CrossRefPubMed
19.
Blade J, Vesole DH, Gertz M (2003) High-dose therapy in multiple myeloma. Blood 102(10):3469–3470CrossRefPubMed
20.
Barlogie B, Smith L, Alexanian R (1984) Effective treatment of advanced multiple myeloma refractory to alkylating agents. N Engl J Med 310(21):1353–1356CrossRefPubMed
21.
Harousseau JL, Milpied N, Laporte JP, Collombat P, Facon T, Tigaud JD et al (1992) Double-intensive therapy in high-risk multiple myeloma. Blood 79(11):2827–2833PubMed
22.
Fermand JP, Levy Y, Gerota J, Benbunan M, Cosset JM, Castaigne S et al (1989) Treatment of aggressive multiple myeloma by high-dose chemotherapy and total body irradiation followed by blood stem cells autologous graft. Blood 73(1):20–23PubMed
23.
Barlogie B, Tricot GJ, van Rhee F, Angtuaco E, Walker R, Epstein J et al (2006) Long-term outcome results of the first tandem autotransplant trial for multiple myeloma. Br J Haematol 135(2):158–164CrossRefPubMed
24.
Barlogie B, Tricot G, Anaissie E, Shaughnessy J, Rasmussen E, van Rhee F et al (2006) Thalidomide and hematopoietic-cell transplantation for multiple myeloma. N Engl J Med 354(10):1021–1030CrossRefPubMed
25.
Barlogie B, Tricot G, Rasmussen E, Anaissie E, van Rhee F, Zangari M et al (2006) Total therapy 2 without thalidomide in comparison with total therapy 1: role of intensified induction and posttransplantation consolidation therapies. Blood 107(7):2633–2638CrossRefPubMed
26.
Attal M, Harousseau JL, Facon T, Guilhot F, Doyen C, Fuzibet JG et al (2003) Single versus double autologous stem-cell transplantation for multiple myeloma. N Engl J Med 349(26):2495–2502CrossRefPubMed
27.
Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F et al (2007) Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. J Clin Oncol: Official J Am Soc Clin Oncol 25(17):2434–2441CrossRef
28.
Fermand JP, Alberti C, Morolleau JP (2003) Single versus tandem high dose therapy (HDT) supported with autlogous blood stem cell (ABSC) transplantation using unselected or CD34- enriched ABSC: results of a two by two designed radomized trial in 230 young aptients with multiple myeloma (MM). Hematol J 4:S59
29.
Goldschmidt H (2005) Single vs double high dose therapy in multiple myeloma: second analysis of the GMMG-HD2 trial. Haematologica 90:38
30.
Cavo M, Cellini C, Zamagni E (2005) Update on high dose therapy—Italian studies. Haematologica 90:39
31.
Vesole DH, Tricot G, Jagannath S, Desikan KR, Siegel D, Bracy D et al (1996) Autotransplants in multiple myeloma: what have we learned? Blood 88(3):838–847PubMed
32.
Awan F, Kochuparambil ST, Falconer DE, Cumpston A, Leadmon S, Watkins K et al (2013) Comparable efficacy and lower cost of PBSC mobilization with intermediate-dose cyclophosphamide and G-CSF compared with plerixafor and G-CSF in patients with multiple myeloma treated with novel therapies. Bone Marrow Transplant 48(10):1279–1284CrossRefPubMed
33.
Hosing C, Qazilbash MH, Kebriaei P, Giralt S, Davis MS, Popat U et al (2006) Fixed-dose single agent pegfilgrastim for peripheral blood progenitor cell mobilisation in patients with multiple myeloma. Br J Haematol 133(5):533–537CrossRefPubMed
34.
DiPersio JF, Stadtmauer EA, Nademanee A, Micallef IN, Stiff PJ, Kaufman JL et al (2009) Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood 113(23):5720–5726PubMed
35.
Giralt S, Stadtmauer EA, Harousseau JL, Palumbo A, Bensinger W, Comenzo RL et al (2009) International myeloma working group (IMWG) consensus statement and guidelines regarding the current status of stem cell collection and high-dose therapy for multiple myeloma and the role of plerixafor (AMD 3100). Leukemia 23(10):1904–1912CrossRefPubMed
36.
Kumar S, Giralt S, Stadtmauer EA, Harousseau JL, Palumbo A, Bensinger W et al (2009) Mobilization in myeloma revisited: IMWG consensus perspectives on stem cell collection following initial therapy with thalidomide-, lenalidomide-, or bortezomib-containing regimens. Blood 114(9):1729–1735CrossRefPubMed
37.
Palumbo A, Bringhen S, Bruno B, Falcone AP, Liberati AM, Grasso M et al (2010) Melphalan 200 mg/m(2) versus melphalan 100 mg/m(2) in newly diagnosed myeloma patients: a prospective, multicenter phase 3 study. Blood 115(10):1873–1879CrossRefPubMed
38.
Anagnostopoulos A, Aleman A, Ayers G, Donato M, Champlin R, Weber D et al (2004) Comparison of high-dose melphalan with a more intensive regimen of thiotepa, busulfan, and cyclophosphamide for patients with multiple myeloma. Cancer 100(12):2607–2612CrossRefPubMed
39.
Fenk R, Schneider P, Kropff M, Huenerlituerkoglu AN, Steidl U, Aul C et al (2005) High-dose idarubicin, cyclophosphamide and melphalan as conditioning for autologous stem cell transplantation increases treatment-related mortality in patients with multiple myeloma: results of a randomised study. Br J Haematol 130(4):588–594CrossRefPubMed
40.
Badros A, Barlogie B, Siegel E, Roberts J, Langmaid C, Zangari M et al (2001) Results of autologous stem cell transplant in multiple myeloma patients with renal failure. Br J Haematol 114(4):822–829CrossRefPubMed
41.
Facon T, Mary JY, Hulin C, Benboubker L, Attal M, Pegourie B et al (2007) 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 370(9594):1209–1218CrossRefPubMed
42.
Schmitz N, Linch DC, Dreger P, Goldstone AH, Boogaerts MA, Ferrant A et al (1996) Randomised trial of filgrastim-mobilised peripheral blood progenitor cell transplantation versus autologous bone-marrow transplantation in lymphoma patients. Lancet 347(8998):353–357CrossRefPubMed
43.
Ford PA, Grant SJ, Mick R, Keck G (2015) Autologous stem-cell transplantation without hematopoietic support for the treatment of hematologic malignancies in Jehovah’s witnesses. J Clin Oncol: Official J Am Soc Clin Oncol 33(15):1674–1679CrossRef
44.
Gertz MA, Ansell SM, Dingli D, Dispenzieri A, Buadi FK, Elliott MA et al (2008) Autologous stem cell transplant in 716 patients with multiple myeloma: low treatment-related mortality, feasibility of outpatient transplant, and effect of a multidisciplinary quality initiative. Mayo Clin Proc 83(10):1131–1138CrossRefPubMed
45.
Jones JA, Qazilbash MH, Shih YC, Cantor SB, Cooksley CD, Elting LS (2008) In-hospital complications of autologous hematopoietic stem cell transplantation for lymphoid malignancies: clinical and economic outcomes from the Nationwide Inpatient Sample. Cancer 112(5):1096–1105CrossRefPubMed
46.
Bjorkstrand B, Svensson H, Goldschmidt H, Ljungman P, Apperley J, Mandelli F et al (2001) Alpha-interferon maintenance treatment is associated with improved survival after high-dose treatment and autologous stem cell transplantation in patients with multiple myeloma: a retrospective registry study from the European Group for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant 27(5):511–515CrossRefPubMed
47.
Davies FE, Forsyth PD, Rawstron AC, Owen RG, Pratt G, Evans PA et al (2001) The impact of attaining a minimal disease state after high-dose melphalan and autologous transplantation for multiple myeloma. Br J Haematol 112(3):814–819CrossRefPubMed
48.
Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T et al (2012) Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med 366(19):1782–1791CrossRefPubMed
49.
Shaughnessy J, Jacobson J, Sawyer J, McCoy J, Fassas A, Zhan F et al (2003) Continuous absence of metaphase-defined cytogenetic abnormalities, especially of chromosome 13 and hypodiploidy, ensures long-term survival in multiple myeloma treated with Total Therapy I: interpretation in the context of global gene expression. Blood 101(10):3849–3856CrossRefPubMed
50.
McCarthy PL, Owzar K, Hofmeister CC, Hurd DD, Hassoun H, Richardson PG et al (2012) Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med 366(19):1770–1781CrossRefPubMedPubMedCentral
51.
Child JA, Morgan GJ, Davies FE, Owen RG, Bell SE, Hawkins K et al (2003) High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 348(19):1875–1883CrossRefPubMed
52.
Rajkumar SV, Fonseca R, Dispenzieri A, Lacy MQ, Witzig TE, Lust JA et al (2000) Effect of complete response on outcome following autologous stem cell transplantation for myeloma. Bone Marrow Transplant 26(9):979–983CrossRefPubMed
53.
Walker R, Barlogie B, Haessler J, Tricot G, Anaissie E, Shaughnessy JD Jr et al (2007) Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications. J Clin Oncol: Official J Am Soc Clin Oncol 25(9):1121–1128CrossRef
54.
van Rhee F, Bolejack V, Hollmig K, Pineda-Roman M, Anaissie E, Epstein J et al (2007) High serum-free light chain levels and their rapid reduction in response to therapy define an aggressive multiple myeloma subtype with poor prognosis. Blood 110(3):827–832CrossRefPubMedPubMedCentral
55.
Haessler J, Shaughnessy JD Jr, Zhan F, Crowley J, Epstein J, van Rhee F et al (2007) Benefit of complete response in multiple myeloma limited to high-risk subgroup identified by gene expression profiling. Clin Cancer Res: Official J Am Assoc Cancer Res 13(23):7073–7079CrossRef
56.
Rawstron AC, Gregory WM, de Tute RM, Davies FE, Bell SE, Drayson MT et al (2015) Minimal residual disease in myeloma by flow cytometry: independent prediction of survival benefit per log reduction. Blood 125(12):1932–1935CrossRefPubMedPubMedCentral
57.
Gahrton G, Svensson H, Bjorkstrand B, Apperley J, Carlson K, Cavo M et al (1999) Syngeneic transplantation in multiple myeloma—a case-matched comparison with autologous and allogeneic transplantation. European Group for Blood and Marrow Transplantation. Bone Marrow Transplant 24(7):741–745CrossRefPubMed
58.
Fefer A, Cheever MA, Greenberg PD (1986) Identical-twin (syngeneic) marrow transplantation for hematologic cancers. J Natl Cancer Inst 76(6):1269–1273PubMed
59.
Auner HW, Szydlo R, van Biezen A, Iacobelli S, Gahrton G, Milpied N et al (2013) 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 48(11):1395–1400CrossRefPubMed
60.
Majolino I, Davoli M, Carnevalli E, Locasciulli A, Di Bartolomeo P, Scime R et al (2007) Reduced intensity conditioning with thiotepa, fludarabine, and melphalan is effective in advanced multiple myeloma. Leuk lymphoma 48(4):759–766CrossRefPubMed
61.
Shimoni A, Hardan I, Ayuk F, Schilling G, Atanackovic D, Zeller W et al (2010) Allogenic hematopoietic stem-cell transplantation with reduced-intensity conditioning in patients with refractory and recurrent multiple myeloma: long-term follow-up. Cancer 116(15):3621–3630CrossRefPubMed
62.
Armeson KE, Hill EG, Costa LJ (2013) Tandem autologous vs autologous plus reduced intensity allogeneic transplantation in the upfront management of multiple myeloma: meta-analysis of trials with biological assignment. Bone Marrow Transplant 48(4):562–567CrossRefPubMed
63.
Krishnan A, Pasquini MC, Logan B, Stadtmauer EA, Vesole DH, Alyea E 3rd et al (2011) 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 12(13):1195–1203CrossRefPubMedPubMedCentral
64.
Lokhorst HM, van der Holt B, Cornelissen JJ, Kersten MJ, van Oers M, Raymakers R et al (2012) Donor versus no-donor comparison of newly diagnosed myeloma patients included in the HOVON-50 multiple myeloma study. Blood 119(26):6219–6225; quiz 399
65.
Garban F, Attal M, Michallet M, Hulin C, Bourhis JH, Yakoub-Agha I et al (2006) 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 107(9):3474–3480CrossRefPubMed
66.
Bruno B, Rotta M, Patriarca F, Mordini N, Allione B, Carnevale-Schianca F et al (2007) A comparison of allografting with autografting for newly diagnosed myeloma. New Engl J Med 356(11):1110–1120CrossRefPubMed
67.
Martinez-Lopez J, Blade J, Mateos MV, Grande C, Alegre A, Garcia-Larana J et al (2011) Long-term prognostic significance of response in multiple myeloma after stem cell transplantation. Blood 118(3):529–534CrossRefPubMed