Open access

Controversies in Autologous Stem Cell Transplantation for the Treatment of Multiple Myeloma

Written By

Bhavana Bhatnagar and Ashraf Z. Badros

Submitted: 22 May 2012 Published: 13 February 2013

DOI: 10.5772/54115

From the Edited Volume

Innovations in Stem Cell Transplantation

Edited by Taner Demirer

Chapter metrics overview

2,286 Chapter Downloads

View Full Metrics

1. Introduction

The treatment paradigm for multiple myeloma has evolved considerably over the past three decades with the incorporation of autologous stem cell transplantation (ASCT) in upfront therapy for eligible patients, and the use of novel agents. As a result, although multiple myeloma remains an incurable disease, clinical outcomes have significantly improved. In this chapter we will review the seminal studies that established the role of ASCT in multiple myeloma and as well as the current controversies with regard to the role of ASCT in the management of myeloma in the era of novel agents. We will review conditioning regimens, post-transplant maintenance strategies with novel agents and immune modulation. We will summarize the current data on early versus late ASCT, single versus tandem transplant and the role of ASCT in patients with relapsed or progressive disease.

Advertisement

2. The role of autologous stem cell transplantation in multiple myeloma

The advent of autologous stem cell transplantation has changed the therapeutic landscape for the management of multiple myeloma and has been the standard frontline therapy for younger patients with normal renal function since the 1990’s. The standard of care for multiple myeloma patients prior to the incorporation of ASCT was conventional chemotherapy using melphalan and prednisone with the primary goals of treatment being achievement of partial response or disease stabilization. Treatment complications and later resistance were associated with poor outcomes with median overall survival ranged between two and three years.

High-dose chemotherapy was initially explored as a therapeutic approach in the 1980’s after a landmark study demonstrated its effectiveness in inducing 100-percent complete remission rates in nine high-risk multiple myeloma and plasma cell leukemia patients after preconditioning with high-dose melphalan. The observation that high-dose melphalan had significant anti-tumor activity and could overcome primary drug resistance was confirmed in a later study.

Since its initial description, there have been seven randomized clinical trials comparing high-dose ASCT to conventional chemotherapy (Table 1). The first of these trials was conducted by the Intergroupe Français du Myélome (IFM) in which 200 untreated multiple myeloma patients under 65 years of age were randomized to receive either conventional chemotherapy or high dose chemotherapy in combination with ASCT. Response rates were significantly higher in patients receiving high-dose chemotherapy and ASCT compared to those who received conventional chemotherapy alone (81% vs 57%, p<0.001). Furthermore, patients who received high-dose therapy had a higher probability of 5-year event-free survival (28% vs 10%, p = 0.01) and estimated 5-year rate of overall survival (52% vs 12%, p =0.03). Seven years later, the findings from the IFM study were corroborated by the British Medical Research Council Myeloma VII Trial (MRCM-VII) in a larger 407 patient multicenter study.

These findings prompted modifications to the disease response criteria as proposed by the International Myeloma Working Group as the achievement of complete responses (CRs), which were rare using conventional chemotherapy, became more achievable and, most importantly, were found to correlate with survival endpoints.

An additional five prospective randomized trials comparing ASCT to conventional chemotherapy followed. Most, but not all, demonstrated superiority of ASCT to conventional chemotherapy with respect to higher rates of CR and very good partial responses (VGPR) which ultimately translated into longer progression-free survival (PFS). An overall survival (OS) benefit was reported in three of the seven studies [5,6,12]. Differences in methodology and trial design between studies may account for some of the discordance in results. A systematic review and meta-analysis of these randomized trials reported improved overall median PFS with no significant improvement in OS following ASCT when compared to conventional chemotherapy .

In summary, high dose chemotherapy and ASCT has markedly improved the depth of response, overall response rates, and length of progression-free survival in multiple myeloma patients. Most importantly, ASCT has improved overall survival from a median of 36 months to 50-55 months, thereby establishing it as the standard of care for multiple myeloma patients under the age of 65 with normal renal function. However, there remains considerable heterogeneity between myeloma patients with regard to underlying disease characteristics and post-ASCT clinical responses. A number of prognostic markers have been identified that influence disease response to chemotherapy, ASCT and survival, specifically age, elevated β-2-microglobulin levels, LDH and serum free light chain ratio. Additionally, the recognition of recurrent chromosomal abnormalities, which have been reported in as many as 90% of patients has allowed myeloma patients to be categorized into low, intermediate and high risk groups on the basis of these aberrations. Translocation (4;14), t(14;20), deletion 17p and gain of 1q have been well associated with poor disease responses and negatively impact overall survival. A recent update from the IFM group have demonstrated a 75% 8-year survival rate in patients who did not have these chromosomal abnormalities and β-2-microglobulin values less than 5.5mg/L.

Trial/group(Year of publication) No. Patients Age, years Median Follow-up Response Rates (%) (CCT vs ASCT) EFS, mos (CCT vs ASCT) OS, mos (CCT vs ASCT)
IFM, (1996) 200 <65 7 years ORR: 57 vs 81 18 vs 28 44 vs 57
CR: 5 vs 22
VGPR: 9 vs 16
MAG91 (1998) 185 55-65 58 mos ORR: 62 vs 86 19 vs 24 50 vs 55
CR: 5 vs 19
BMRC VII, (2003) 407 <65 42 mos ORR: 67 vs 90 19 vs 31 42 vs 54
CR: 8 vs 44
PR: 40 vs 42
Italian MMSG M97G(2004) 194 50-70 39 mos ORR: 42 vs 73 15.6 vs 28 42 vs 58+
nCR: 6 vs 25
MAG95 (2005) 190 55-65 120 mos ORR: 77 vs 70 19 vs 31 42 vs 54
CR +MRD: 20 vs 36
PR: 38.5 vs 26
MR: 18 vs 7
PETHEMA, (2005) 164 <65 44 mos CR: 11 vs 30 33 vs 42 66 vs 61
US Intergroup 9321 (2006) 516 ≤70 76 mos CR: 15 vs 17 14% vs 17%** 38% vs 38%**

Table 1.

Randomized trials comparing ASCT to conventional chemotherapy (CCT)

Abbreviations: IFM:,Intergroupe Français du Myélome, MAG: Myélome Autogreffe, BMRC: British Medical Research Council, MMSG: Multiple Myeloma Study Group, PETHEMA: Programa para el Estudio de la Terapéutica en Hemaopatia Maligna, ORR: Overall response rate, CR: Complete remission, nCR: Near complete remission, VGPR: Very good partial response, PR: Partial response, MRD: Minimal residual disease, MR: Minimal response


** 7-year estimated EFS and OS rate


While clinical outcomes have improved significantly since the widespread implementation of ASCT, there are several unanswered questions relating to the use of ASCT in multiple myeloma, particularly in the era of novel therapies, which remain as areas of active investigation. However, before these controversies can be fully addressed, it is important to understand the role of novel agents and their impact on myeloma management before discussing their current use in the context of ASCT.

Advertisement

3. Immune modulation and the advent of novel agents

The concept of immune modulation was formulated and developed after a greater understanding of the complex interaction between myeloma cells and their microenvironment as well as the discovery that myeloma cells, through a variety of mechanisms, are inherently able to evade host natural immune defenses, thereby potentiating their own survival. The immune dysregulation that is known to accompany multiple myeloma is believed to be the result of multiple biological pathways and mechanisms including excess production of myeloma–derived cytokines, inadequate antigen presentation, resistance to NK-cell lysis and impaired activity of B, T and NK cells. Additionally, multiple myeloma is also associated with defective humoral and cellular immunity leading to abnormal B-cell differentiation and function. Reduced numbers of CD4+ T cells, abnormal Th1/Th2 CD4+ T-cell ratios, impaired cytotoxic T-cell responses, dysfunction of NK and NK T-cells and abnormal dendritic cell function further compound the immune dysfunction associated with multiple myeloma.

The immunomodulatory drugs (IMiDs), lenalidomide and pomalidomide are thalidomide analogs that were specifically developed in response to the resurgence of interest in thalidomide after it was incidentally discovered to be an effective treatment in patients with cutaneous leprosy presumably through inhibition of TNFα. Subsequent preclinical trials revealed that thalidomide, in fact, had several favorable properties that would optimize its use as an anti-cancer agent.

The IMiDs, were created with the intent to maximize the pleiotropic activity directed against myeloma cells that was demonstrated by thalidomide, and, in fact are 50,000 times more potent than thalidomide in their immunomodulatory properties, including CD4+ and CD8+ T-cell costimulation, Th1 cytokine production, NK and NK T-cell activation, and antibody-dependent cellular cytotoxicity. Furthermore, they also disrupt the interaction between myeloma cells and the tumor microenvironment through potent inhibition of angiogenesis and downregulation of inflammatory cytokines, specifically TNFα, from peripheral blood mononuclear cells. The IMiDs also directly exert anti-tumor proliferation effects. Additionally the IMiDs are more capable of stimulating T-cells with without incurring the same degree of toxicity as thalidomide. The manipulation of the immune system by IMiDs has established their efficacy in the management of multiple myeloma. Lenalidomide and thalidomide, in addition to the proteasome inhibitor, bortezomib, are considered the main novel agents, and, in light of their significant disease activity, are now routinely integrated into multiple myeloma management in ASCT eligible and ineligible patients.

Advertisement

4. The impact of novel agents on induction and stem cell mobilization

Prior to the widespread use and incorporation of novel agents, the standard induction regimen was vincristine, doxorubicin and dexamethasone (VAD). Dexamethasone was the most active drug in this regimen and has long since remained the cornerstone of upfront treatment for multiple myeloma,. The investigation and incorporation of novel agents into induction chemotherapy regimens was prompted by the discovery that the quality of disease response following induction therapy, preceding ASCT, corresponded to better clinical outcomes, including subsequent response to ASCT, PFS and OS. Novel agents were initially investigated to determine whether the rates of these responses could be improved. Table 3 summarizes the results of published studies using novel agents as part of induction therapy prior to ASCT.

Thalidomide-based induction regimens were initially compared to VAD and were found to produce higher VGPR, but not CR, rates prior to transplant.. However, the increased incidence of thromboembolic complications and drug toxicity rendered the overall benefit of thalidomide containing regimens somewhat modest. A 10-year clinical follow-up study of 169 myeloma with advanced or refractory disease who were initially treated with thalidomide demonstrated remarkably improved event-free survival and OS in patients with normal cytogenetics and non-lambda light chain isotype.

Lenalidomide and high-dose dexamethasone (RD) was compared to lenalidomide and low-dose dexamethasone (Rd) as initial therapy in transplant eligible and ineligible patients and, while improved response rates (≥ VGPR) were significantly improved in patients receiving RD, increased toxicities and mortality were also more pronounced with this regimen, especially in patients older than 65 years of age. Furthermore, ASCT in combination with RD or Rd improved 3-year OS rates compared to patients who did not undergo ASCT [92% vs 79%]. Three drug-combinations using lenalidomide, bortezomib and dexamethasone (RVD) have also been investigated in a few phase I/II studies and have shown even greater improvements in response rates pre and post-transplant.

The proteasome inhibitor, bortezomib, in combination with dexamethasone was initially discovered to significantly improve near complete remission (nCR) and CR rates in the landmark IFM2005-1 trial when it was compared to VAD, VAD and dexamethasone, cyclophosphamide, etoposide and cisplatin (DCEP) consolidation, and bortezomib and dexamethasone followed by DCEP consolidation followed by ASCT. Bortezomib-containing regimens resulted in higher CR/nCR rates irrespective of disease stage or cytogenetic risk. Post-transplant, these improved response rates were associated with improved CR, nCR and VGPR rates as well as improved PFS after a median follow-up of 32 months compared to patients treated with VAD alone (36 mos vs 30 mos). In the VISTA trial, the addition of bortezomib to melphalan and prednisone also produced longer OS, and was not found to incur more resistant relapses in a long term follow-up study. The IFM 2005-1 and VISTA trials were critical in establishing the role of bortezomib in induction therapy for myeloma. To further improve the depth of disease response several phase II and III clinical trials have evaluated the efficacy of adding a third novel agent, either lenalidomide or thalidomide, to the bortezomib and dexamethasone backbone, and have demonstrated improved responses following the addition of a third agent.

Although novel agents have vastly improved the quality of disease response as well as overall response rates in the pre- and post-transplant settings, the use of these agents as part of induction therapy has resulted in greater difficulties with stem cell collection prior to autologous transplant, particularly with the use of lenalidomide and, to a lesser extent, bortezomib although the exact mechanisms by which stem cell collection is hindered has not yet been fully elucidated. To address this issue, the International Myeloma Working Group has recommended early stem cell mobilization, following 3-4 cycles of induction therapy. Mobilization using G-CSF alone or in combination with cyclophosphamide is typically considered adequate; and while a large multi-center randomized phase III trial demonstrated a significant improvement in the number of CD34+ cells/kg collected in patients receiving G-CSF and the CXCR4 inhibitor, plerixafor (AMD3100) compared to G-CSF and placebo, the routine use of plerixafor upfront for mobilization remains controversial.

Advertisement

5. The importance of pre-transplant disease response

Complete remissions in the pre-ASCT era were rare, but have now become a very attainable and desirable treatment goal in the pre and post-transplant settings, especially as they are considered to be strong surrogate markers for progression-free and survival overall survival in several studies. The prognostic impact of CR was not fully appreciated until ASCT was adopted as frontline therapy in the management of multiple myeloma, and this is reflected in the International Myeloma Working Group response criteria by the introduction of stringent CRs to further qualify the depth of response [Table 2]. Furthermore, the duration of CR is also described as a favorable prognostic variable ; however, in several patient subgroups, including those with a prior history of monoclonal gammopathy of undetermined significance and smoldering myeloma or with low-risk disease achievement of CR appears to be of less importance.

sCR: Meets criteria for CR plus normal FLC ratio and no clonal cells bone marrow IHC or immunofluorescence
CR: Absence of M protein in serum and urine by immunofixation, < 5% bone marrow plasma cells, no increase of lytic bone lesions, disappearance of soft tissue plasmacytomas
VGPR: Serum and urine M protein detectable by immunofixation but not on electrophoresis OR ≤ 90% reduction in serum M-protein plus urine M-protein <100mg/24hr

Table 2.

IMWG Complete Response Criteria (Durie et al, Leukemia 2006)

Abbreviations: IMWG: International Myeloma Working Group, sCR: stringent complete remission, CR: Complete Remission, VGPR: Very Good Partial Response, FLC: Free Light Chains, IHC: Immunohistochemistry


Author, date of publication No. of patients Treatment regimen Median follow-up RR after induction (%) RR after transplant (%) PFS, median OS
Rajkumar, 2006 207 TD vs Dex 207 CR: 4 vs 0
≥ VGPR: ---
≥ PR: 63 vs 41
CR: ---
≥ VGPR: ---
≥ PR: ---
--- ---
Lokhorst, 2010 536 VAD vs TAD 52 mos CR: 2 vs 3
≥ VGPR: 18 vs 37
≥ PR: 57 vs 71
CR: 12 vs 14
≥ VGPR: 44 vs 54
≥ PR: 76 vs 84
22 vs 34 mos 60 vs 73 mos
Harousseau, 2010 482 VAD vs VD 31.2 mos CR/nCR: 6.4 vs 14.8
≥ VGPR: 15.1 vs 37.7
≥ PR: 62.8 vs 72.5
CR/nCR: 18.4 vs 35
≥ VGPR: 37.2 vs 54.3
≥ PR: 77.1 vs 80.3
29.7 vs 36 mos 3 yr OS 77.4% vs 81.4%
Cavo, 2010 480 VTD vs TD 36 mos CR/nCR: 31 vs 11
≥ VGPR: 62 vs 28
≥ PR: 93 vs 79
CR/nCR: 55 vs 41
≥ VGPR: 82 vs 64
≥ PR: 93 vs 84
68 % vs 56%* 86% vs 84%*
Rajkumar, 2010 445 RD vs Rd 35.8 mos CR: 5 vs 4
≥ VGPR: 71vs 26
≥ PR: 81 vs 70
----
----
----
19 vs 25 mos 2yr OS 87% vs 75%
Moreau, 2011 199 VD vs vtD 32 mos CR: 22 vs 31
≥ VGPR: 36 vs 49
≥ PR: 81 vs 88
CR: 52 vs 61
≥ VGPR: 58 vs 74
≥ PR: 86 vs 89
30 vs 26 mos ---
Rosinol, 2012 386 VTD vs TD vs VBMCP/VBAD/B 35.2 mos CR: 35 vs 14 vs 21
≥ VGPR: 25 vs 15 vs 15
≥ PR: 25 vs 33 vs 39
CR: 46 vs 24 vs 38
≥ VGPR: ---
≥ PR: ---
56.2 vs 28.2 vs 35.3 mos 4yr OS 74% vs 65% vs 70%
Sonneveld, 2012 833 VAD vs PAD 41 mos CR/nCR: 15 vs 11
≥ VGPR: 14 vs 42
≥ PR: 54 vs 78
CR/nCR: 15 vs 31
≥ VGPR: 36 vs 62
≥ PR: 75 vs 88
28 vs 35 mos 5 yr OS, 55% vs 65%

Table 3.

Published phase III studies using novel agents as part of induction therapy prior to ASCT

Abbreviations: TD: Thalidomide and Dexamethasone,Dex: Dexamethasone VAD: Vincristine, Adriamycin and Dexamethasone, TAD: Thalidomide, Adriamycin and Dexamethasone, VD: Bortezomib and Dexamethasone, VTD: Bortezomib, Thalidomide and Dexamethasone, RD: Lenalidomide and high-dose dexamethasone, Rd: Lenalidomide and low-dose dexamethasone, PAD: Bortezomib, Adriamycin and Dexamethasone, vtD: reduced dose bortezomib, thalidomide and Dexamethasone


Advertisement

6. Early versus late transplant

Only one randomized trial has compared upfront ASCT to late ASCT at the time of relapse. Upfront ASCT improved event-free survival and quality of life compared to patients treated with conventional chemotherapy and who underwent ASCT as rescue treatment at the time of relapse; interestingly there was no appreciable difference in 5-year overall survival between the arms . However, in the era of novel agents and resultant improvements in complete remission rates, the question as to whether ASCT could potentially be delayed until disease relapse or progression has, again, resurfaced. The International Myeloma Working Group recommends that all eligible patients be offered ASCT at some point in their disease course and while there are no published randomized phase III trials incorporating the use of novel agents in induction therapy to support the use of ASCT upon disease relapse, many clinicians opt to collect stem cells early and preserve them for use following disease relapse. We believe that upfront ASCT should be the standard of care until ongoing trials establish that delayed ASCT after novel agents has a role.

Advertisement

7. Single ASCT versus tandem transplant

Tandem ASCT, as part of a more intensified treatment strategy (“total therapy”) was initially shown to improve CR rates, EFS and OS in comparison to standard therapy. The superiority of double ASCT was later appreciated in a landmark randomized clinical trial which demonstrated significantly improved OS, particularly in patients who had not achieved VGPR following transplant. Several other randomized trials have also attempted to compare single versus double ASCT and have reported conflicting results regarding the superiority of one approach over the other. A recent meta-analysis attempted to answer this question and concluded that tandem transplant offered no benefit in terms of disease outcomes and was, in fact, associated with greater morbidity; however, this analysis has received criticism due to the heterogeneity of the selected studies which were evaluated as well as variability in treatment methodology. A more recent analysis suggests that tandem ASCT does offer a survival benefit. Most clinicians speculate that tandem and single transplants are equivocal, however, there have been no definitive trials evaluating this issue and it remains an area of considerable debate.

Advertisement

8. Methods to improve conditioning regimens: The addition of total body irradiation or other agents to high-dose melphalan

The quality of disease response following pre-transplant conditioning is critical to the success of ASCT. High dose melphalan 200mg/m2 is the standard conditioning chemotherapy regimen prior to autologous HSCT in multiple myeloma as this approach has demonstrated superior overall survival rates in comparison to chemotherapy alone. Various approaches to improve responses to this conditioning regimen while minimizing toxicities have been evaluated in a number of studies.

Total body irradiation (TBI) in combination with melphalan demonstrated improved CR rates, relapse and progression rates and five year OS rates when compared to TBI and cyclophosphamide as a myeloablative conditioning regimen in myeloma patients undergoing allogeneic HSCT. A landmark study, however, in which melphalan and TBI was compared with high-dose melphalan 200mg/m2 demonstrated more rapid hematologic recovery, reduced transfusion requirements, shorter hospitalization and improved survival in patients receiving high-dose melphalan alone. As such, the routine use of TBI in conjunction with melphalan is not widely used.

The alkylating agent, busulfan, has been used in several studies in combination melphalan with promising outcomes, particularly in patients with non-remission disease at the time of transplant. A recent analysis of multiple myeloma patients treated with oral busulfan and melphalan 140mg/m2 compared to standard melphalan 200mg/m2 did demonstrate improved median PFS (41 mos vs 31 mos, p=0.009), however, the increased incidence of veno-occlusive disease and transplant related mortality counteracted the benefits; additionally, patients who received busulfan had less access to salvage therapies using novel agents than patients who had relapsed following treatment with melphalan 200mg/m2.

The Intergroupe Francophone du Myelome study group also evaluated the efficacy of adding bortezomib to high-dose melphalan in a recent phase II study and were able to demonstrate, that, in comparison to historical controls, patients treated with the bortezomib and melphalan achieved higher CR rates (35% vs 11%, p=.001) with no increase in hematologic toxicity.

Advertisement

9. Novel agents as post-transplant maintenance therapy

Maintenance with interferon, steroids, and chemotherapy has been tried in many centers for over 30 years with no clear benefit. Maintenance interferon frequently resulted in worsened quality of life; furthermore, future development of therapy-related myelodysplastic syndrome following chemotherapy led to these maintenance treatments to fall out of favor. The availability of novel agents and their tolerable toxicity profiles has renewed interest in post-transplant maintenance treatment. The results of this approach have, thus far, been encouraging, including upgrades in disease responses and improvements in PFS/EFS, and OS in many studies; however, none of these agents are currently approved in the post-transplant setting. The recently released consensus statement from the International Myeloma Working Group does not advocate for or against maintenance therapy and recommends that the decision to use maintenance therapy be made on an individualized basis. In the following paragraph we will review various agents with a brief summary of the randomized trials data.

9.1. Thalidomide

Thalidomide maintenance therapy following ASCT has been evaluated in six randomized clinical trials all of which have reported a significant improvement in progression free survival in patients receiving thalidomide maintenance versus the comparator arm, but only 3 had shown improvement in OS by 6-9 months. Two meta-analyses have confirmed improved OS with thalidomide maintenance. However, most patients (> 50%) eventually discontinued thalidomide, between 7 months and 2 years of treatment, due to side effects, particularly development of peripheral neuropathy. Interestingly, thalidomide maintenance does not benefit patients with poor-risk cytogenetics, and, in fact, this patient subset was shown to have a shorter survival duration. Similar results from the Total Therapy 2 study were reported although a longer follow up showed improvement in long-term survival in high risk disease, although the main impact was most appreciable in patients with favorable cytogenetics.

9.2. Lenalidomide

Lenalidomide has a favorable toxicity profile, and its efficacy extends beyond inhibition of the growth of myeloma cells as the drug also causes alterations within the bone marrow microenvironment leading to an enhancement of immune responses, thereby making it an ideal drug for post-transplant maintenance. Two very recently published trials from the Cancer and Leukemia Group B (CALGB) and IFM evaluated the efficacy of lenalidomide following transplant and demonstrated that lenalidomide maintenance therapy was associated with a significant improvement in PFS compared to placebo (48 vs 30.9 mos, and 41 vs 24 mos in the CALGB and IFM studies, respectively). The benefits of lenalidomide maintenance were appreciated across all patient subgroups, including those with high-risk cytogenetics, although the data is limited to a small number pf patients in the IFM study, β2-microglobulin and response following transplant. In the IFM-2005-02 trial, patients were given two courses of lenalidomide as consolidation treatment which led to an upgrade in the number of disease response with rates of CR increasing from 14% to 20% and responses higher than or equal to VGPR from 58% to 67%. The side effects were tolerable, mostly hematologic, and responded well to dose adjustments, supportive growth factor injections and transfusion support. A meta-analysis by the International Myeloma Working Group, which included a total of 1380 patients demonstrated a 65% reduction in risk of disease progression for patients treated with lenalidomide maintenance therapy. There is a notable increased risk of second cancers in association with this drug as noted by both IFM and CALGB. The IFM reported the incidence of second cancers as 3.1 per 100 patient years, compared to 1.2 per 100 patient years in the placebo group (p = 0.002). In the CALGB study, 8% of patients treated with lenalidomide developed second cancers, compared to 3% in the control arm.

9.3. Bortezomib

An interesting study to evaluate the effect of minimal residual disease, by qualitative and real-time quantitative polymerase chain reaction (RQ-PCR) after ASCT showed that a consolidation regimen comprised of bortezomib, thalidomide, and dexamethasone (VTD) increased CRs from 15% after ASCT to 49% after VTD. Most importantly, molecular remissions increased from 3% after ASCT to 18% after VTD. No patients had relapsed at the time of reporting (median follow-up, 42 months). These unprecedented levels of tumor cell reduction are very encouraging and have laid the foundation for a new area of investigation to better evaluate the depth of treatment response in myeloma.

A subsequent randomized phase 3 study specifically assessed the efficacy and safety of consolidation therapy using bortezomib, thalidomide and dexamethasone (VTD) versus thalidomide and dexamethasone (TD). Before starting consolidation, CR/nCR rates were not significantly different in the VTD and TD arms (63.1% 54.7%, respectively). However, after consolidation, CR (60.6% vs 46.6%) and CR/nCR (73.1% vs 60.9%) rates were significantly higher for VTD-treated versus TD-treated patients. With a median follow-up of 30.4 months from start of consolidation, 3-year PFS was significantly longer for the VTD group compared to TD (60% vs 48%). The VTD consolidation therapy was shown to significantly improve clinical outcomes after ASCT.

The evaluation of novel agents in the post-transplant setting has resulted in significant improvements in disease responses and survival endpoints. Moreover combination regimens in the form of consolidation and/or long term maintenance are well tolerated with further improvements and achievement of molecular remissions. Future studies to determine the optimal duration of maintenance therapy are urgently needed.

Advertisement

10. Combined ASCT/Allogeneic Hematopoeitic Stem Cell Transplant approaches

Early trials evaluating myeloablative allogeneic stem cell transplantation in the treatment of multiple myeloma demonstrated improvements in relapse and progression rates attributed to graft versus myeloma effects; however, development of graft versus host disease and infectious complications resulted in high transplant-related mortality [TRM]. A critical advantage of allogeneic transplantation was the development of reduced intensity conditioning (RIC) regimens that were associated with decreased toxicities and profound graft versus tumor effects as demonstrated in early trials evaluating the efficacy of RIC in relapsed and refractory myeloma patients. However, higher rates of disease progression and relapse, were noted and attributed to the late use of this modality underscoring the importance of using effective regimens early before the disease becomes refractory especially since the goals of allogeneic transplant are curative in intent.

Combined sequential therapy utilizing ASCT for cytoreduction followed RIC allogeneic transplant (i.e. the auto-allo approach) to exploit the graft versus myeloma effect has been compared to tandem ASCT in several studies; randomization in these trials was biological; i.e. patients with an HLA-matched sibling received RIC allogeneic transplant and all others underwent tandem ASCT. The first published study from the IFM compared tandem ASCT in 219 patients to auto-allo in 65 patients with high-risk multiple myeloma and reported no significant difference in response rates or event-free survival between groups; however, there was an observed trend toward better overall survival in patients treated with tandem ASCT; these findings remained unchanged in a long-term follow-analysis from the same group. Subsequent comparisons have reported improved CR rates and PFS durations and only one has shown superior OS in auto-allo treated patients. However a recently published large multi-center phase 3 study reported that the auto-allo approach was not superior to auto-auto in terms of progression-free survival (43 % vs 46% at 3 years) or 3-year OS (77% vs 80%). Additionally, there was no significant difference in the development of grade 3-5 adverse events between groups by three years (46% vs 42%). Further modifications to allogeneic transplantation would be needed to offset the graft versus myeloma effect as well as the increase in transplant-related mortality. The Eastern Cooperative Oncology Group conducted a small trial in which 32 patients received non-myeloablative matched sibling donor transplant following ASCT and reported a 78% ORR (30% CR and 48% PR) with low TRM; however over half of patients developed chronic GVHD. A recent Swedish study compared auto-allo approach to single ASCT in 357 previously untreated multiple myeloma patients and was demonstrated that the auto-allo approach was superior in terms of PFS, OS and relapse rate with a 12% nonrelapse mortality rate. The data remain conflicting; however, a meta-analysis reviewing outcomes on 7 published and unpublished studies concluded that the auto-allo approach offers no benefit compared to autologous transplant approaches and is associated with higher TRM. The International Myeloma Working Group does not recommend the routine use of allogeneic transplantation, and, in fact, recommends consideration of RIC transplant only in the setting of a clinical trial.

11. Novel immunotherapy strategies

The post-transplant period is the ideal time point for immunotherapy as the disease burden is, theoretically, low. Immune function remains depressed following high-dose therapy for many months. Ex vivo expansion and subsequent transfer of autologous stimulated T cells may enhance host antitumor immunity and may also allow for enhancement of a post-transplant vaccination strategy against tumor-directed antigens. Early trials focused on the generation of antibodies against myeloma specific antigens. The idiotype [Id] protein has, in a number of pre-clinical studies, demonstrated powerful antibody responses that, in vitro, resulted in apoptosis of myeloma cells. However, durable clinical responses were not seen in subsequent clinical studies. Idiotype-pulsed dendritic cell vaccinations following ASCT have also demonstrated that cellular immune responses can be elicited in the context of minimal residual disease following transplantation; however, again, there is no definitive evidence that these vaccination strategies alter the course of disease. It has been suggested that the immune dysfunction in myeloma patients is the primary barrier to successful vaccination strategies. A low number of T-cells with activity against myeloma have been detected in multiple myeloma patients . Several attempts to expand T cells, collected from the peripheral blood of affected patients, and infused after ASCT, have shown that rapid recovery of T-cell numbers can be achieved but, unfortunately, with no clear anti-myeloma benefits. The results of one interesting study in which myeloma patients received the conjugated pneumococcal vaccine before T-cell collection and after ASCT showed profound antibody responses, suggesting that T cells may improve immune responses to vaccination. A subsequent study in which adoptive transfer of vaccine primed autologous T-cells to the htert/survivin multipeptide vaccine, a target in myeloma cells, corroborated these findings and demonstrated that vaccination was associated with robust antibody responses in most patients; however, again, there was no definitive activity directed against myeloma cells specifically. Clinical trials building on the expansion of T cells and targeting various myeloma antigen such as MAGE A3 and NYESO1 are ongoing. Of important note, several studies focusing on expansion of marrow infiltrating lymphocytes (MILs) had yielded interesting results with regards to antimyeloma activities, but, again, the clinical benefit was quite limited.

Several antibody trials in myeloma are ongoing. A recently published phase 1 study has provided encouraging evidence that elotuzumab in combination with lenalidomide yields impressive responses in relapsed and refractory myeloma; whether the responses seen in the relapsed setting can be confirmed and implemented in patients with minimal disease states would require further investigation. In light of these findings, it is suggested that enhancement of T-cell function could potentially lay the groundwork for subsequent trials aimed to improve immune function, and by extension, clinical outcomes following ASCT in myeloma patients.

12. Salvage ASCT for relapsed disease

At present, the optimal treatment approach for patients with relapsed disease following initial ASCT has not yet been defined. Potential options include treatment with novel agents, conventional chemotherapy or a second salvage ASCT. While the data evaluating the role of a second ASCT are limited, several small retrospective analyses have demonstrated that it is an effective and well tolerated treatment option with overall response rates reported between 55-90%. Overall survival and progression free survival is significantly improved for patients who have received fewer lines of therapy prior to transplant and for those who have experienced a late disease relapse. However, the length of time which constitutes a late relapse has varied between studies, ranging between 12 months and > 36 months. A recently published retrospective review suggested a time-dependent association between remission duration following initial ASCT and PFS following transplant. Patients who relapsed within 18 months of initial ASCT had significantly shorter PFS compared to those who relapsed between 18 and 36 months and those who relapsed 36 months or more (4.2 mos vs 13.8 mos vs 49.1 mos) [111]. Although larger studies would provide greater insight regarding the optimal timing of a second transplant, consideration of salvage ASCT is generally regarded as feasible approach which offers the greatest benefit in select patients who have relapsed at least more than 12 months after their initial ASCT. Salvage allogeneic transplant following failure of initial autografting has also been compared to salvage ASCT in a limited number of studies and has been reported to have comparable PFS due to lower rates of disease progression following allogeneic transplant, but superior OS in autografted patients; furthermore, the increased incidence of graft versus host disease in allografted patients has rendered this approach less preferable. Refinements in allogeneic transplant techniques may potentially generate renewed interest in this treatment approach.

13. Conclusion

The widespread implementation of autologous stem cell transplantation, in conjunction with novel agents, has revolutionized the management of multiple myeloma and has markedly altered the natural history of the disease by improving disease responses and response duration, which, by extension, have led to significant improvements in overall survival. While treatment options for multiple myeloma have expanded considerably over the past several decades, long-term survivorship remains low. Continued investigative efforts are targeted towards refining our current treatment modalities with the hope of ultimately developing a treatment approach which results in cure.

References

  1. 1. Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma: an overview of 6,633 patients from 27 randomized trials. Myeloma Trialists' Collaborative Group. J Clin Oncol. 1998 Dec;16[12]:3832-42. PubMed PMID: 9850028.
  2. 2. Corso A, Nozza A, Lazzarino M, Klersy C, Zappasodi P, Arcaini L, et al. Plateau phase in multiple myeloma: an end-point of conventional-dose chemotherapy. Haematologica. 1999;84[4]:336-41.
  3. 3. McElwain TJ, Powles RL. High-dose intravenous melphalan for plasma-cell leukaemia and myeloma. Lancet. 1983;2[8354]:822-4.
  4. 4. Barlogie B, Hall R, Zander A, Dicke K, Alexanian R. High-dose melphalan with autologous bone marrow transplantation for multiple myeloma. Blood. 1986;67[5]:1298-301.
  5. 5. Attal M, Harousseau JL, Stoppa AM, Sotto JJ, Fuzibet JG, Rossi JF, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. The New England journal of medicine. 1996;335[2]:91-7.
  6. 6. Child JA, Morgan GJ, Davies FE, Owen RG, Bell SE, Hawkins K, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. The New England journal of medicine. 2003;348[19]:1875-83.
  7. 7. Durie BG, Harousseau JL, Miguel JS, Blade J, Barlogie B, Anderson K, et al. International uniform response criteria for multiple myeloma. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2006;20[9]:1467-73.
  8. 8. Barlogie B, Kyle RA, Anderson KC, Greipp PR, Lazarus HM, Hurd DD, et al. Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24[6]:929-36.
  9. 9. Blade J, Rosinol L, Sureda A, Ribera JM, Diaz-Mediavilla J, Garcia-Larana J, et al. High-dose therapy intensification compared with continued standard chemotherapy in multiple myeloma patients responding to the initial chemotherapy: long-term results from a prospective randomized trial from the Spanish cooperative group PETHEMA. Blood. 2005;106[12]:3755-9.
  10. 10. Fermand JP, Katsahian S, Divine M, Leblond V, Dreyfus F, Macro M, et al. High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23[36]:9227-33.
  11. 11. Fermand JP, Ravaud P, Chevret S, Divine M, Leblond V, Belanger C, et al. High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood. 1998;92[9]:3131-6.
  12. 12. Palumbo A, Bringhen S, Petrucci MT, Musto P, Rossini F, Nunzi M, et al. Intermediate-dose melphalan improves survival of myeloma patients aged 50 to 70: results of a randomized controlled trial. Blood. 2004;104[10]:3052-7.
  13. 13. Koreth J, Cutler CS, Djulbegovic B, Behl R, Schlossman RL, Munshi NC, et al. High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: A systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant. 2007 Feb;13[2]:183-96. PubMed PMID: 17241924.
  14. 14. Harousseau JL, Moreau P. Autologous hematopoietic stem-cell transplantation for multiple myeloma. The New England journal of medicine. 2009;360[25]:2645-54.
  15. 15. Ludwig H, Durie BG, Bolejack V, Turesson I, Kyle RA, Blade J, et al. Myeloma in patients younger than age 50 years presents with more favorable features and shows better survival: an analysis of 10 549 patients from the International Myeloma Working Group. Blood. 2008 Apr 15;111[8]:4039-47. PubMed PMID: 18268097.
  16. 16. Avet-Loiseau H, Attal M, Moreau P, Charbonnel C, Garban F, Hulin C, et al. Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myelome. Blood. 2007;109[8]:3489-95.
  17. 17. Rossi D, Fangazio M, De Paoli L, Puma A, Riccomagno P, Pinto V, et al. Beta-2-microglobulin is an independent predictor of progression in asymptomatic multiple myeloma. Cancer. 2010;116[9]:2188-200.
  18. 18. Stella-Holowiecka B, Czerw T, Holowiecka-Goral A, Giebel S, Wojnar J, Holowiecki J. Beta-2-microglobulin level predicts outcome following autologous hematopoietic stem cell transplantation in patients with multiple myeloma. Transplantation proceedings. 2007 Nov;39[9]:2893-7. PubMed PMID: 18022010.
  19. 19. Maltezas D, Dimopoulos MA, Katodritou I, Repousis P, Pouli A, Terpos E, et al. Re-evaluation of prognostic markers including staging, serum free light chains or their ratio and serum lactate dehydrogenase in multiple myeloma patients receiving novel agents. Hematological oncology. 2012.
  20. 20. Fonseca R, Blood E, Rue M, Harrington D, Oken MM, Kyle RA, et al. Clinical and biologic implications of recurrent genomic aberrations in myeloma. Blood. 2003;101[11]:4569-75.
  21. 21. Gertz MA, Lacy MQ, Dispenzieri A, Greipp PR, Litzow MR, Henderson KJ, et al. Clinical implications of t[11;14][q13;q32], t[4;14][p16.3;q32], and -17p13 in myeloma patients treated with high-dose therapy. Blood. 2005;106[8]:2837-40.
  22. 22. Avet-Loiseau H, Attal M, Campion L, Caillot D, Hulin C, Marit G, et al. Long-term analysis of the IFM 99 trials for myeloma: cytogenetic abnormalities [t[4;14], del[17p], 1q gains] play a major role in defining long-term survival. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30[16]:1949-52.
  23. 23. Brown RD, Pope B, Murray A, Esdale W, Sze DM, Gibson J, et al. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 [B7-1] expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood. 2001;98[10]:2992-8.
  24. 24. Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, Smyth MJ, et al. Mechanism of action of immunomodulatory drugs [IMiDS] in multiple myeloma. Leukemia. 2010 Jan;24[1]:22-32. PubMed PMID: 19907437.
  25. 25. Sampaio EP, Sarno EN, Galilly R, Cohn ZA, Kaplan G. Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes. The Journal of experimental medicine. 1991;173[3]:699-703.
  26. 26. D'Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of angiogenesis. Proceedings of the National Academy of Sciences of the United States of America. 1994;91[9]:4082-5.
  27. 27. Davies FE, Raje N, Hideshima T, Lentzsch S, Young G, Tai YT, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood. 2001;98[1]:210-6.
  28. 28. Haslett PA, Corral LG, Albert M, Kaplan G. Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production, and cytotoxic responses in the CD8+ subset. The Journal of experimental medicine. 1998;187[11]:1885-92.
  29. 29. Gorgun G, Calabrese E, Soydan E, Hideshima T, Perrone G, Bandi M, et al. Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood. 2010;116[17]:3227-37.
  30. 30. Corral LG, Haslett PA, Muller GW, Chen R, Wong LM, Ocampo CJ, et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. Journal of immunology [Baltimore, Md: 1950]. 1999;163[1]:380-6.
  31. 31. Muller GW, Corral LG, Shire MG, Wang H, Moreira A, Kaplan G, et al. Structural modifications of thalidomide produce analogs with enhanced tumor necrosis factor inhibitory activity. Journal of medicinal chemistry. 1996;39[17]:3238-40.
  32. 32. Barlogie B, Smith L, Alexanian R. Effective treatment of advanced multiple myeloma refractory to alkylating agents. N Engl J Med. 1984 May 24;310[21]:1353-6. PubMed PMID: 6546971.
  33. 33. Alexanian R, Barlogie B, Tucker S. VAD-based regimens as primary treatment for multiple myeloma. Am J Hematol. 1990 Feb;33[2]:86-9. PubMed PMID: 2301376.
  34. 34. Alexanian R, Dimopoulos MA, Delasalle K, Barlogie B. Primary dexamethasone treatment of multiple myeloma. Blood. 1992 Aug 15;80[4]:887-90. PubMed PMID: 1498331.
  35. 35. Harousseau JL, Attal M, Avet-Loiseau H. The role of complete response in multiple myeloma. Blood. 2009;114[15]:3139-46.
  36. 36. Harousseau JL, Avet-Loiseau H, Attal M, Charbonnel C, Garban F, Hulin C, et al. Achievement of at least very good partial response is a simple and robust prognostic factor in patients with multiple myeloma treated with high-dose therapy: long-term analysis of the IFM 99-02 and 99-04 Trials. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27[34]:5720-6.
  37. 37. Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet. 2010 Dec 18;376[9758]:2075-85. PubMed PMID: 21146205.
  38. 38. Harousseau JL, Attal M, Avet-Loiseau H, Marit G, Caillot D, Mohty M, et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28[30]:4621-9.
  39. 39. Lokhorst HM, van der Holt B, Zweegman S, Vellenga E, Croockewit S, van Oers MH, et al. A randomized phase 3 study on the effect of thalidomide combined with adriamycin, dexamethasone, and high-dose melphalan, followed by thalidomide maintenance in patients with multiple myeloma. Blood. 2010 Feb 11;115[6]:1113-20. PubMed PMID: 19880501.
  40. 40. Moreau P, Avet-Loiseau H, Facon T, Attal M, Tiab M, Hulin C, et al. Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood. 2011;118[22]:5752-8; quiz 982.
  41. 41. Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR. Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol. 2006 Jan 20;24[3]:431-6. PubMed PMID: 16365178. Epub 2005/12/21. eng.
  42. 42. Rajkumar SV, Jacobus S, Callander NS, Fonseca R, Vesole DH, Williams ME, et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. The lancet oncology. 2010;11[1]:29-37.
  43. 43. Rosinol L, Oriol A, Teruel AI, Hernandez D, Lopez-Jimenez J, de la Rubia J, et al. Superiority of bortezomib, thalidomide, and dexamethasone [VTD] as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood. 2012;120[8]:1589-96.
  44. 44. Sonneveld P, Schmidt-Wolf IG, van der Holt B, El Jarari L, Bertsch U, Salwender H, et al. Bortezomib Induction and Maintenance Treatment in Patients With Newly Diagnosed Multiple Myeloma: Results of the Randomized Phase III HOVON-65/ GMMG-HD4 Trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30[24]:2946-55.
  45. 45. Cavo M, Zamagni E, Tosi P, Tacchetti P, Cellini C, Cangini D, et al. Superiority of thalidomide and dexamethasone over vincristine-doxorubicindexamethasone [VAD] as primary therapy in preparation for autologous transplantation for multiple myeloma. Blood. 2005;106[1]:35-9.
  46. 46. Goldschmidt H, Sonneveld P, Cremer FW, van der Holt B, Westveer P, Breitkreutz I, et al. Joint HOVON-50/GMMG-HD3 randomized trial on the effect of thalidomide as part of a high-dose therapy regimen and as maintenance treatment for newly diagnosed myeloma patients. Annals of hematology. 2003 Oct;82[10]:654-9. PubMed PMID: 12845480.
  47. 47. van Rhee F, Dhodapkar M, Shaughnessy JD, Jr., Anaissie E, Siegel D, Hoering A, et al. First thalidomide clinical trial in multiple myeloma: a decade. Blood. 2008;112[4]:1035-8.
  48. 48. Richardson PG, Weller E, Lonial S, Jakubowiak AJ, Jagannath S, Raje NS, et al. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. 2010;116[5]:679-86.
  49. 49. Wang M, Delasalle K, Giralt S, Alexanian R. Rapid control of previously untreated multiple myeloma with bortezomib-lenalidomide-dexamethasone [BLD]. Hematology [Amsterdam, Netherlands]. 2010;15[2]:70-3.
  50. 50. Mateos MV, Richardson PG, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, et al. Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow-up and impact of subsequent therapy in the phase III VISTA trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28[13]:2259-66.
  51. 51. Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet. 2010;376[9758]:2075-85.
  52. 52. Popat U, Saliba R, Thandi R, Hosing C, Qazilbash M, Anderlini P, et al. Impairment of filgrastim-induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma. Biol Blood Marrow Transplant. 2009 Jun;15[6]:718-23. PubMed PMID: 19450756.
  53. 53. Kumar S, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, Gastineau DA, et al. Impact of lenalidomide therapy on stem cell mobilization and engraftment post-peripheral blood stem cell transplantation in patients with newly diagnosed myeloma. Leukemia. 2007 Sep;21[9]:2035-42. PubMed PMID: 17581613.
  54. 54. DiPersio JF, Micallef IN, Stiff PJ, Bolwell BJ, Maziarz RT, Jacobsen E, et al. Phase III prospective randomized double-blind placebo-controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-Hodgkin's lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27[28]:4767-73.
  55. 55. Kumar S, Giralt S, Stadtmauer EA, Harousseau JL, Palumbo A, Bensinger W, et al. Mobilization in myeloma revisited: IMWG consensus perspectives on stem cell collection following initial therapy with thalidomide-, lenalidomide-, or bortezomib-containing regimens. Blood. 2009 Aug 27;114[9]:1729-35. PubMed PMID: 19561323.
  56. 56. Lahuerta JJ, Martinez-Lopez J, Serna JD, Blade J, Grande C, Alegre A, et al. Remission status defined by immunofixation vs. electrophoresis after autologous transplantation has a major impact on the outcome of multiple myeloma patients. British journal of haematology. 2000;109[2]:438-46.
  57. 57. van de Velde HJ, Liu X, Chen G, Cakana A, Deraedt W, Bayssas M. Complete response correlates with long-term survival and progression-free survival in high-dose therapy in multiple myeloma. Haematologica. 2007;92[10]:1399-406.
  58. 58. Barlogie B, Anaissie E, Haessler J, van Rhee F, Pineda-Roman M, Hollmig K, et al. Complete remission sustained 3 years from treatment initiation is a powerful surrogate for extended survival in multiple myeloma. Cancer. 2008;113[2]:355-9.
  59. 59. Pineda-Roman M, Bolejack V, Arzoumanian V, Anaissie E, van Rhee F, Zangari M, et al. Complete response in myeloma extends survival without, but not with history of prior monoclonal gammopathy of undetermined significance or smouldering disease. British journal of haematology. 2007;136[3]:393-9.
  60. 60. Cavo M, Rajkumar SV, Palumbo A, Moreau P, Orlowski R, Blade J, et al. International Myeloma Working Group consensus approach to the treatment of multiple myeloma patients who are candidates for autologous stem cell transplantation. Blood. 2011;117[23]:6063-73.
  61. 61. Barlogie B, Jagannath S, Vesole DH, Naucke S, Cheson B, Mattox S, et al. Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood. 1997;89[3]:789-93.
  62. 62. Attal M, Harousseau JL, Facon T, Guilhot F, Doyen C, Fuzibet JG, et al. Single versus double autologous stem-cell transplantation for multiple myeloma. The New England journal of medicine. 2003;349[26]:2495-502.
  63. 63. Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25[17]:2434-41.
  64. 64. Goldschmidt H. Single vs double high-dose therapy in multiple myeloma: second analysis of the GMMG-HD2 trial. Haematologica2005.
  65. 65. Segeren CM, Sonneveld P, van der Holt B, Vellenga E, Croockewit AJ, Verhoef GE, et al. Overall and event-free survival are not improved by the use of myeloablative therapy following intensified chemotherapy in previously untreated patients with multiple myeloma: a prospective randomized phase 3 study. Blood. 2003;101[6]:2144-51.
  66. 66. Kumar A, Kharfan-Dabaja MA, Glasmacher A, Djulbegovic B. Tandem versus single autologous hematopoietic cell transplantation for the treatment of multiple myeloma: a systematic review and meta-analysis. J Natl Cancer Inst. 2009 Jan 21;101[2]:100-6. PubMed PMID: 19141779. Epub 2009/01/15. eng.
  67. 67. Barlogie B, Attal M, Crowley J, van Rhee F, Szymonifka J, Moreau P, et al. Long-term follow-up of autotransplantation trials for multiple myeloma: update of protocols conducted by the intergroupe francophone du myelome, southwest oncology group, and university of arkansas for medical sciences. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28[7]:1209-14.
  68. 68. Palumbo A, Bringhen S, Bruno B, Falcone AP, Liberati AM, Grasso M, et al. Melphalan 200 mg/m[2] versus melphalan 100 mg/m[2] in newly diagnosed myeloma patients: a prospective, multicenter phase 3 study. Blood. 2010;115[10]:1873-9.
  69. 69. Hunter HM, Peggs K, Powles R, Rahemtulla A, Mahendra P, Cavenagh J, et al. Analysis of outcome following allogeneic haemopoietic stem cell transplantation for myeloma using myeloablative conditioning--evidence for a superior outcome using melphalan combined with total body irradiation. British journal of haematology. 2005;128[4]:496-502.
  70. 70. Moreau P, Facon T, Attal M, Hulin C, Michallet M, Maloisel F, et al. 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. 2002;99[3]:731-5.
  71. 71. Lahuerta JJ, Grande C, Blade J, Martinez-Lopez J, de la Serna J, Alegre A, et al. Myeloablative treatments for multiple myeloma: update of a comparative study of different regimens used in patients from the Spanish registry for transplantation in multiple myeloma. Leukemia & lymphoma. 2002;43[1]:67-74.
  72. 72. Ria R, Falzetti F, Ballanti S, Minelli O, Di Ianni M, Cimminiello M, et al. Melphalan versus melphalan plus busulphan in conditioning to autologous stem cell transplantation for low-risk multiple myeloma. The hematology journal : the official journal of the European Haematology Association / EHA. 2004;5[2]:118-22.
  73. 73. Blanes M, Lahuerta JJ, Gonzalez JD, Ribas P, Solano C, Alegre A, et al. Intravenous Busulfan and Melphalan as Conditioning Regimen for Autologous Stem Cell Transplantation in Patients with Newly Diagnosed Multiple Myeloma: A Matched Comparison to a Melphalan-only Approach. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2012.
  74. 74. Carreras E, Rosinol L, Terol MJ, Alegre A, de Arriba F, Garcia-Larana J, et al. Veno-occlusive disease of the liver after high-dose cytoreductive therapy with busulfan and melphalan for autologous blood stem cell transplantation in multiple myeloma patients. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2007;13[12]:1448-54.
  75. 75. Lahuerta JJ, Mateos MV, Martinez-Lopez J, Grande C, de la Rubia J, Rosinol L, et al. Busulfan 12 mg/kg plus melphalan 140 mg/m2 versus melphalan 200 mg/m2 as conditioning regimens for autologous transplantation in newly diagnosed multiple myeloma patients included in the PETHEMA/GEM2000 study. Haematologica. 2010;95[11]:1913-20.
  76. 76. Roussel M, Moreau P, Huynh A, Mary JY, Danho C, Caillot D, et al. Bortezomib and high-dose melphalan as conditioning regimen before autologous stem cell transplantation in patients with de novo multiple myeloma: a phase 2 study of the Intergroupe Francophone du Myelome [IFM]. Blood. 2010 Jan 7;115[1]:32-7. PubMed PMID: 19884643.
  77. 77. Ludwig H, Durie BG, McCarthy P, Palumbo A, San Miguel J, Barlogie B, et al. IMWG consensus on maintenance therapy in multiple myeloma. Blood. 2012;119[13]:3003-15.
  78. 78. Attal M, Harousseau JL, Leyvraz S, Doyen C, Hulin C, Benboubker L, et al. Maintenance therapy with thalidomide improves survival in patients with multiple myeloma. Blood. 2006;108[10]:3289-94.
  79. 79. Barlogie B, Pineda-Roman M, van Rhee F, Haessler J, Anaissie E, Hollmig K, et al. Thalidomide arm of Total Therapy 2 improves complete remission duration and survival in myeloma patients with metaphase cytogenetic abnormalities. Blood. 2008;112[8]:3115-21.
  80. 80. Morgan GJ, Gregory WM, Davies FE, Bell SE, Szubert AJ, Brown JM, et al. The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis. Blood. 2012;119[1]:7-15.
  81. 81. Spencer A, Prince HM, Roberts AW, Prosser IW, Bradstock KF, Coyle L, et al. Consolidation therapy with low-dose thalidomide and prednisolone prolongs the survival of multiple myeloma patients undergoing a single autologous stem-cell transplantation procedure. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27[11]:1788-93.
  82. 82. Hahn-Ast C. Improved progression-free and overall survival with thalidomide maintenance therapy after autologous transplantion in multiple myeloma: a meta-analysis of five randomized trials. Haematologica. 2011;96:suppl 2, abstract 884.
  83. 83. Barlogie B, Tricot G, Anaissie E, Shaughnessy J, Rasmussen E, van Rhee F, et al. Thalidomide and hematopoietic-cell transplantation for multiple myeloma. The New England journal of medicine. 2006;354[10]:1021-30.
  84. 84. Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. The New England journal of medicine. 2012;366[19]:1782-91.
  85. 85. McCarthy PL, Owzar K, Hofmeister CC, Hurd DD, Hassoun H, Richardson PG, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. The New England journal of medicine. 2012;366[19]:1770-81.
  86. 86. Ladetto M, Pagliano G, Ferrero S, Cavallo F, Drandi D, Santo L, et al. Major tumor shrinking and persistent molecular remissions after consolidation with bortezomib, thalidomide, and dexamethasone in patients with autografted myeloma. J Clin Oncol. 2010 Apr 20;28[12]:2077-84. PubMed PMID: 20308672. Epub 2010/03/24. eng.
  87. 87. Cavo M, Pantani L, Petrucci MT, Patriarca F, Zamagni E, Donnarumma D, et al. 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 Jul 5;120[1]:9-19. PubMed PMID: 22498745.
  88. 88. Gahrton G, Tura S, Flesch M, Gratwohl A, Gravett P, Lucarelli G, et al. Bone marrow transplantation in multiple myeloma: report from the European Cooperative Group for Bone Marrow Transplantation. Blood. 1987;69[4]:1262-4.
  89. 89. Aschan J, Lonnqvist B, Ringden O, Kumlien G, Gahrton G. Graft-versus-myeloma effect. Lancet. 1996;348[9023]:346.
  90. 90. Tricot G, Vesole DH, Jagannath S, Hilton J, Munshi N, Barlogie B. Graft-versus-myeloma effect: proof of principle. Blood. 1996;87[3]:1196-8.
  91. 91. Badros A, Barlogie B, Morris C, Desikan R, Martin SR, Munshi N, et al. High response rate in refractory and poor-risk multiple myeloma after allotransplantation using a nonmyeloablative conditioning regimen and donor lymphocyte infusions. Blood. 2001 May 1;97[9]:2574-9. PubMed PMID: 11313244.
  92. 92. Crawley C, Iacobelli S, Bjorkstrand B, Apperley JF, Niederwieser D, Gahrton G. Reduced-intensity conditioning for myeloma: lower nonrelapse mortality but higher relapse rates compared with myeloablative conditioning. Blood. 2007;109[8]:3588-94.
  93. 93. Garban F, Attal M, Michallet M, Hulin C, Bourhis JH, Yakoub-Agha I, et al. Prospective comparison of autologous stem cell transplantation followed by dose-reduced allograft [IFM99-03 trial] with tandem autologous stem cell transplantation [IFM99-04 trial] in high-risk de novo multiple myeloma. Blood. 2006;107[9]:3474-80.
  94. 94. Moreau P, Garban F, Attal M, Michallet M, Marit G, Hulin C, et al. 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 Nov 1;112[9]:3914-5. PubMed PMID: 18948589.
  95. 95. Bruno B, Rotta M, Patriarca F, Mordini N, Allione B, Carnevale-Schianca F, et al. A comparison of allografting with autografting for newly diagnosed myeloma. The New England journal of medicine. 2007;356[11]:1110-20.
  96. 96. Rosinol L, Perez-Simon JA, Sureda A, de la Rubia J, de Arriba F, Lahuerta JJ, et al. A prospective PETHEMA study of tandem autologous transplantation versus autograft followed by reduced-intensity conditioning allogeneic transplantation in newly diagnosed multiple myeloma. Blood. 2008;112[9]:3591-3.
  97. 97. Krishnan A, Pasquini MC, Logan B, Stadtmauer EA, Vesole DH, Alyea E, 3rd, et al. 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. The lancet oncology. 2011;12[13]:1195-203.
  98. 98. Vesole DH, Zhang L, Flomenberg N, Greipp PR, Lazarus HM, Huff CA, et al. A Phase II trial of autologous stem cell transplantation followed by mini-allogeneic stem cell transplantation for the treatment of multiple myeloma: an analysis of Eastern Cooperative Oncology Group ECOG E4A98 and E1A97. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2009;15[1]:83-91.
  99. 99. Bjorkstrand B, Iacobelli S, Hegenbart U, Gruber A, Greinix H, Volin L, et al. Tandem autologous/reduced-intensity conditioning allogeneic stem-cell transplantation versus autologous transplantation in myeloma: long-term follow-up. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011;29[22]:3016-22.
  100. 100. Armeson KE, Hill EG, Costa LJ. 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 transplantation. 2012.
  101. 101. Lokhorst H, Einsele H, Vesole D, Bruno B, San Miguel J, Perez-Simon JA, et al. International Myeloma Working Group consensus statement regarding the current status of allogeneic stem-cell transplantation for multiple myeloma. J Clin Oncol. 2010 Oct 10;28[29]:4521-30. PubMed PMID: 20697091.
  102. 102. Froyland M, Ruffini PA, Thompson KM, Gedde-Dahl T, Fredriksen AB, Bogen B. Targeted idiotype-fusion DNA vaccines for human multiple myeloma: preclinical testing. European journal of haematology. 2011;86[5]:385-95.
  103. 103. Houet L, Veelken H. Active immunotherapy of multiple myeloma. European journal of cancer [Oxford, England : 1990]. 2006;42[11]:1653-60.
  104. 104. Massaia M, Borrione P, Battaglio S, Mariani S, Beggiato E, Napoli P, et al. Idiotype vaccination in human myeloma: generation of tumor-specific immune responses after high-dose chemotherapy. Blood. 1999 Jul 15;94[2]:673-83. PubMed PMID: 10397734.
  105. 105. Osterborg A, Yi Q, Henriksson L, Fagerberg J, Bergenbrant S, Jeddi-Tehrani M, et al. Idiotype immunization combined with granulocyte-macrophage colony-stimulating factor in myeloma patients induced type I, major histocompatibility complex-restricted, CD8- and CD4-specific T-cell responses. Blood. 1998;91[7]:2459-66.
  106. 106. Lim SH, Bailey-Wood R. Idiotypic protein-pulsed dendritic cell vaccination in multiple myeloma. International journal of cancerJournal international du cancer. 1999;83[2]:215-22.
  107. 107. Liso A, Stockerl-Goldstein KE, Auffermann-Gretzinger S, Benike CJ, Reichardt V, van Beckhoven A, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood progenitor cell transplantation for multiple myeloma. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2000;6[6]:621-7.
  108. 108. Reichardt VL, Okada CY, Liso A, Benike CJ, Stockerl-Goldstein KE, Engleman EG, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma--a feasibility study. Blood. 1999;93[7]:2411-9.
  109. 109. Titzer S, Christensen O, Manzke O, Tesch H, Wolf J, Emmerich B, et al. Vaccination of multiple myeloma patients with idiotype-pulsed dendritic cells: immunological and clinical aspects. British journal of haematology. 2000;108[4]:805-16.
  110. 110. Dhodapkar MV, Krasovsky J, Olson K. T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells. Proceedings of the National Academy of Sciences of the United States of America. 2002;99[20]:13009-13.
  111. 111. Rapoport AP, Stadtmauer EA, Aqui N, Badros A, Cotte J, Chrisley L, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nature medicine. 2005;11[11]:1230-7.
  112. 112. Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011;117[3]:788-97.
  113. 113. Noonan K, Huff CA, Sproul JMD, Lemas MVM, Rudraraju L, Luznik L, et al. Phase I/II Study of Marrow Infiltrating Lymphocytes [MILs] Generates Measurable Myeloma-Specific Immunity in the Autologous Stem Cell Transplant [SCT] Setting. ASH Annual Meeting Abstracts. 2011 November 18, 2011;118[21]:997-.
  114. 114. Noonan KA, Mgebroff S, Serafini P, Meckel K, Bonyhadi M, Borrello I. CD4+CD25+ Marrow Infiltrating Lymphocytes in Myeloma Patients Display an Activated Phenotype and Lack Suppressive Function. ASH Annual Meeting Abstracts. 2006 November 16, 2006;108[11]:1741-.
  115. 115. Lonial S, Vij R, Harousseau JL, Facon T, Moreau P, Mazumder A, et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30[16]:1953-9.
  116. 116. Chow AW, Lee CH, Hiwase DK, To LB, Horvath N. Relapsed Multiple Myeloma: Who Benefits from Salvage Autografts? Internal Medicine Journal. 2012.
  117. 117. Jimenez-Zepeda VH, Mikhael J, Winter A, Franke N, Masih-Khan E, Trudel S, et al. Second autologous stem cell transplantation as salvage therapy for multiple myeloma: impact on progression-free and overall survival. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2012;18[5]:773-9.
  118. 118. Olin RL, Vogl DT, Porter DL, Luger SM, Schuster SJ, Tsai DE, et al. Second auto-SCT is safe and effective salvage therapy for relapsed multiple myeloma. Bone marrow transplantation. 2009;43[5]:417-22.
  119. 119. Qazilbash MH, Saliba R, De Lima M, Hosing C, Couriel D, Aleman A, et al. Second autologous or allogeneic transplantation after the failure of first autograft in patients with multiple myeloma. Cancer. 2006;106[5]:1084-9.
  120. 120. Shah N, Ahmed F, Bashir Q, Qureshi S, Dinh Y, Rondon G, et al. Durable remission with salvage second autotransplants in patients with multiple myeloma. Cancer. 2012;118[14]:3549-55.
  121. 121. Gonsalves WI, Gertz MA, Lacy MQ, Dispenzieri A, Hayman SR, Buadi FK, et al. Second auto-SCT for treatment of relapsed multiple myeloma. Bone marrow transplantation. 2012.
  122. 122. Fenk R, Liese V, Neubauer F, Bruns I, Kondakci M, Balleisen S, et al. Predictive factors for successful salvage high-dose therapy in patients with multiple myeloma relapsing after autologous blood stem cell transplantation. Leuk Lymphoma. 2011 Aug;52[8]:1455-62. PubMed PMID: 21657961.
  123. 123. Mehta J, Tricot G, Jagannath S, Ayers D, Singhal S, Siegel D, et al. Salvage autologous or allogeneic transplantation for multiple myeloma refractory to or relapsing after a first-line autograft? Bone marrow transplantation. 1998;21[9]:887-92.

Written By

Bhavana Bhatnagar and Ashraf Z. Badros

Submitted: 22 May 2012 Published: 13 February 2013