1,152
Views
8
CrossRef citations to date
0
Altmetric
Editorial

Autologous stem cell transplantation in multiple myeloma is not dead but alive and well

, MD &

Abstract

In multiple myeloma (MM), the use of high-dose chemotherapy with autologous stem cell transplantation (ASCT) led to incremental advances in patient management in the 1990s. The clinical results for patients dramatically improved further in the 2000s with the introduction of immunomodulatory drugs and proteasome inhibitors. In the ‘modern’ era for MM treatment, transplant trials strongly support the use of upfront ASCT in the context of novel agents, and until proven otherwise, the old ASCT remains the standard of care for eligible patients. Nevertheless, some issues remain unresolved and this editorial aims to highlight the concerns to be addressed in the future.

1. Introduction

From the history of multiple myeloma (MM), we have learned that among the multiple combinations of chemotherapy available until 1996, only melphalan and prednisone could improve the overall survival (OS) of patients. In that year, a French group Citation[1] identified ‘something’ that could make a difference in the treatment and survival and that ‘something’ was high-dose chemotherapy (HDC) and autologous stem cell transplantation (ASCT). In the 2000s, a dramatic increase in improvement rates was observed with the advent of immunomodulatory drugs (IMiDs) and proteasome inhibitors, and if this trend continues, the 5-year survival for a patient diagnosed in 2014 should be ∼ 66% Citation[2].The 2010s are characterized by studying the optimal combination, sequence and duration of therapies; however, the specific role and timing for ASCT has not been established with modern induction regimens. In the scientific community, there has been much debate on whether to withdraw the procedure, but we believe that ASCT in MM is not dead but alive and well.

2. Evidence in favor of ASCT

Before the use of the new drugs in induction therapy, the complete remission (CR) rate did not exceed 10%. In this scenario, the rationale for proceeding to HDC with ASCT was to increase the depth of response. Over the past decade, this approach has been considered the standard of care for eligible patients with newly diagnosed MM, based on increased rate of CR, prolonged disease-free (DFS) Citation1, Citation3, Citation4, Citation5, Citation6, Citation7 and OS Citation1, Citation4,compared with conventional chemotherapy (CC).

The major limitation to these studies is that all randomized trials of ASCT after induction therapy were designed and implemented before the availability of novel agents, but we believe that the role of transplantation is equally as strong in the ‘modern era’ ().

Table 1. Multiple myeloma: the role of autologous stem cell transplantation in the era of immunomodulatory drugs and proteasome inhibitors.

The results of the Phase III PETHEMA trial Citation[8] support the use of upfront ASCT in the context of novel drugs. In this study, the response rates were evaluated after induction therapy and after ASCT. The CR rates increased from 35% pre-transplant to 57% post-transplant, in the group treated with bortezomib/thalidomide/dexamethasone (VTD) as induction therapy and from 14 to 40% in the group treated with thalidomide/dexamethasone (TD).

Recently, Palumbo et al. Citation[9]. published results of a randomized, Phase III study comparing melphalan 200 mg/mq (HDM) plus ASCT with melphalan–prednisone–lenalidomide (MPR). Both progression-free survival (PFS) and OS were significantly longer with HDM plus ASCT than with MPR (median PFS: 43.0 vs 22.4 months; hazard ratio [HR] for progression or death = 0.44; 95% CI: 0.32 – 0.61; p <0.001; and 4-year OS: 81.6 vs 65.3%; HR for death = 0.55; 95% CI: 0.32 – 0.93; p = 0.02).

Results from a French trial Citation[10], with patients receiving primary induction therapy with bortezomib/dexamethasone (Vel/Dex) versus vincristine/doxorubicin/dexamethasone (VAD) showed a marked improvement in overall responses with Vel/Dex over VAD. Responses were evaluated after induction treatment and post-ASCT. After the first ASCT, CR/near CR was 35% in the Vel/Dex arm, compared with 18.4% in the VAD arm. The very good partial remission (VGPR) was 54.3 versus 37.2%. PFS was 36 versus 29.7 months (p = 0.064) with Vel/Dex vs VAD. The PFS was longer in patients achieving greater than or equal VGPR after ASCT Citation[11].

In another study, patients were randomized to induction therapy with VTD or TD before double ASCT. The three-drug regimen yielded high response rates, with a CR of 19 versus 5% and VGPR of 62 versus 31%. After ASCT, improved incremental responses were still seen with VTD compared to TD Citation[12].

Taken together, these studies suggest that improved responses with the novel induction therapies result in improved outcomes after ASCT, and currently, the most common indication for ASCT in Europe remains MM Citation[13].

2.1 Toxicity, morbidity and quality of life

HDM has become substantially less toxic over time and it should be considered a safe procedure. The Center for the International Blood and Marrow Transplant Registry showed that the risk of death after ASCT in the 2000s has decreased and published data suggest that day 100 mortality of upfront ASCT is 1% Citation[14]. The introduction of myeloid growth factors after transplantation, the use of peripheral blood stem cells (SCs) in place of bone marrow SCs, modern, better-tolerated induction regimens and other improvements in supportive care have reduced the transplant-related mortality rate, and many transplant teams routinely perform ASCT in the outpatient setting Citation[15]. Also, in contrast to popular perception, mortality, morbidity and societal cost from ongoing non-transplant therapy are not negligible.

3. Questions for the future

In the ‘modern’ era for MM treatment, transplant trials strongly support the use of upfront ASCT in the context of novel agents, and until proven otherwise, ASCT remains the standard of care for eligible patients.

However, there are some highly debated issues that the scientific community needs to resolve. First, the type and duration of induction therapy has not been well defined and there is little consensus on the optimal non-melphalan-containing induction regimens. Although randomized trials have been conducted comparing older regimens with newer regimens, containing IMiDs or proteasome inhibitors, there are few randomized trials that have compared these modern combinations Citation[16]. Thus, the best regimen can only be extrapolated indirectly from Phase III trials showing improved responses before transplant Citation8, Citation9, Citation10, Citation17. However, in the context of evidence-based medicine, bortezomib-based induction seems to be most appropriate for patients who are on their way to an ASCT Citation[18].

The second issue is to better clarify the definition of CR. The International Myeloma Working Group response criteria were developed from the European Society for Blood and Marrow Transplantation, International Bone Marrow Transplant Registry and Autologous Blood & Marrow Transplant registry response criteria, with revisions and improvements to help uniform reporting Citation19, Citation20. However, we believe that the current electrophoresis-based response definitions are inadequate because improvements in sensibility of molecular and flow-cytometry techniques have clearly demonstrated the presence of significant residual MM disease using the current CR definition Citation[21]. In this context, one needs to reconsider the criteria for defining CR (i.e., whether based on flow-cytometry data, immunohistochemistry, positron emission tomography, free light chain levels, etc.).

The third open question is to understand the best SC mobilization procedure to be administered. Mobilization with G-CSF alone is well tolerated but its use can be limited by suboptimal SC yields Citation[22]. Adding chemotherapeutic agents to G-CSF may increase SC yields. The most commonly used chemotherapy-based mobilization in MM includes high-dose cyclophosphamide Citation[23]. There is indication that chemo-mobilization reduces the tumor load in vivo and tumor cell contamination in the apheresis product. The benefit of higher cell yields (compared with G-CSF alone) may be offset by less predictability of timing and an increased risk for the patient Citation[24]. In the era of novel agents, chemotherapy mobilization is clearly not superior to mobilization by growth factors alone. The impressive results achieved by incorporating newer novel agents into frontline therapy are observed not only in response rates but are also correlated with the potential for obtaining minimal residual disease-negative, tumor-free hematopoietic progenitor cell grafts Citation[25]. In this way, it becomes unnecessary to expose the patient to the risks of chemotherapy, the effectiveness of which on the disease is questioned Citation[26].

The fourth issue to be clarified is the number of transplants and the timing of transplantation. ASCT may be single or tandem (a planned second course of HDM within 6 months of the first). Although the studies were performed in the old era, the tandem ASCT approach achieved improvement in OS Citation[27], even though a survival benefit was mainly seen in those patients who failed to achieve at least a VGPR Citation28, Citation29. We underline that patients who achieve a CR and stop treatment is a clinical error, because what we may define as an immunofixation-negative CR may still have tumor cells present. The improved depth of response with newer induction strategies and the efficacy of continuous treatment with IMiDs and proteasome inhibitors is a driving force behind the shift in the timing of ASCT. We believe that clinicians should deliver the best treatment to all patients, particularly when faced with a patient with a potentially good prognosis. From this stand point, it makes much more sense to intensify treatment in patients with a sensitive disease, instead of intensifying treatment in patients with aggressive disease.

Recent results with the new induction regimens suggest that there is a role for tandem ASCT in MM in the presence of adverse cytogenetic abnormalities. In the Italian trial Citation[19] investigating VTD versus TD as induction and consolidation, PFS, DFS or OS at 3 years was comparable for patients with or without the t(4;14) translocation, indicating that the double procedure is efficacious in the presence of this high-risk marker. In another trial comparing bortezomib/doxorubicin/dexamethasone with VAD as induction therapy, the impact of del(17p13) on PFS and OS can be reduced by a novel agent induction regimen followed by a double ASCT Citation[30].

In 2012, the Cochrane review compared tandem ASCT with single ASCT as first-line treatment in patients with symptomatic MM with respect to OS, event-free survival, quality of life and treatment- or transplantation-related mortality Citation[31]. The authors did not consider any study to be sufficiently informative for contemporary treatment decisions in terms of single versus tandem ASCT in view of inherent biases in these studies. In addition, none of the trials integrated the so-called novel agents, which are now considered standard treatment for MM.

A second transplant need not always be performed in tandem. The second transplant can be delayed until progression and becomes a useful salvage strategy. The second ASCT in relapsed MM can be associated with superior OS and PFS compared with CC if the response duration from the first transplant is at least 12 – 24 months Citation[32]. A single trial conducted prior to the availability of novel agents provides the only Phase III data comparing ‘early’ upfront ASCT versus ‘late’ ASCT at disease progression/relapse, with comparable OS noted in both groups Citation[33]. Analyses of the randomized trial, conducted by Rajkumar et al., which compared lenalidomide in combination with high-dose or low-dose dexamethasone as initial therapy, showed that patients choosing ASCT after 4 cycles of therapy had a better 3-year OS than patients choosing to continue initial therapy Citation[34]. In the recently published randomized trial by Palumbo et al. Citation[9], the results suggest that upfront ASCT improves both PFS and OS compared to the arm without HDM.

Further ongoing randomized trials comparing upfront ASCT with new drugs without high-dose therapy will provide a definitive answer to the question if, with modern induction regimens, ASCT can add benefit. It is possible that some subgroups of patients may need early frontline ASCT, whereas in some other patients the procedure may be postponed. It is also true that an appropriate follow up is needed before any conclusion.

The fifth point is the definition of ‘transplantation eligible’. Patients treated are often older than the ones included in the published trials, and based on the increasing number of patients aged over 65 years who undergo ASCT, the performance status and comorbidities should be considered as factors defining transplantation eligibility rather than age Citation14, Citation35.

The sixth issue of debate is the role of consolidation and maintenance in the treatment of MM. Consolidation/maintenance therapy with IMiDs and proteasome inhibitors is a reasonable option for long-term disease control and improvement in OS, although no agent has been approved by the European Medicines Agency for this use. Regardless of the agent that we choose, there must be a risk–benefit discussion. The risk of side effects must be considered with the use of lenalidomide, and an increased risk of second primary malignancies has been reported with long-term exposure, which appears associated with its use in conjunction with melphalan Citation[36]. Toxicities must be considered with long-term use of bortezomib, but the risk of peripheral neuropathy may be lessened with the use of subcutaneous administration. Peripheral neuropathy, thromboembolic and hematologic side effects must be considered with the use of thalidomide, and, in patients with selective adverse molecular cytogenetic lesions determined by interphase fluorescence in situ hybridization (FISH), thalidomide should not be used as maintenance therapy Citation[37].

The last issue to consider is that not all MM are the same. We know that we can risk-stratify the malignancy based on very basic demographics and variables such as the free light chain assay, cytogenetic or FISH markers that can help us identify differences in the structural and functional manifestations of the disease and those indices translate into differences in clinical outcomes. Induction and HDC are good approaches for achieving CR, and in high-risk patients we need to consider maintaining these CRs. In standard-risk patients, maintenance therapy may also improve not only the duration of a response but also OS.

In conclusion, new agents and new drug combinations with ASCT have improved our ability to obtain rapid control of a patient’s disease. The critical issues associated with ASCT in MM are summarized in . Using our best strategies upfront may convert MM into a much more indolent and chronic disease.

Table 2. High-dose chemotherapy with autologous stem cell transplantation strategy in multiple myeloma: open questions.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents, received or pending, or royalties.

Bibliography

  • Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med 1996;335:91-7
  • Bergsagel PL. Where we were, where we are, where we are going: progress in multiple myeloma. Am Soc Clin Oncol Educ Book 2014;199-203
  • Fermand JP, Ravaud P, Chevret S, et al. High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood 1998;92:3131-6
  • Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 2003;348:1875-83
  • Blade J, Rosinol L, Sureda A, et al. High-dose therapy intensification compared with continued standard chemotherapy in multiple myeloma patients responding to the initial chemotherapy: long-term results from a prospective randomized trial from the Spanish cooperative group PETHEMA. Blood 2005;106:3755-9
  • Fermand JP, Katsahian S, Divine M, et al. High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. J Clin Oncol 2005;23:9227-33
  • Barlogie B, Kyle RA, Anderson KC, et al. Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321. J Clin Oncol 2006;24:929-36
  • Rosinol L, Oriol A, Teruel AI, 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;120:1589-96
  • Palumbo A, Cavallo F, Gay F, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med 2014;371(10):895-905
  • Harousseau JL, Attal M, Avet-Loiseau H, et al. Bortezomib plus dexamethasone is superior to vincristine plus doxorubicin plus dexamethasone as induction treatment prior to autologous stem-cell transplantation in newly diagnosed multiple myeloma: results of the IFM 2005-01 phase III trial. J Clin Oncol 2010;28(30):4621-9
  • Harousseau JL, Mathiot C, Attal M, et al. Bortezomib/dexamethasone versus VAD as induction prior to autologous stem cell transplantion (ASCT) in previously untreated multiple myeloma (MM): Updated data from IFM 2005/01 trial. J Clin Oncol (2008 ASCO Annual Meeting Proceedings (Post-Meeting Edition)). 2008. 26: 15S(20 Suppl): 8505
  • Cavo M, Tacchetti P, Patriarca F, et al. A Phase III Study of Double Autotransplantation Incorporating Bortezomib-Thalidomide-Dexamethasone (VTD) or Thalidomide-Dexamethasone (TD) for Multiple Myeloma: Superior Clinical Outcomes with VTD Compared to TD. Blood (ASH Annual Meeting Abstracts) 2009;114:abstract 351
  • Passweg JR, Baldomero H, Peters C, et al. Hematopoietic SCT in Europe: data and trends in 2012 with special consideration of pediatric transplantation. Bone Marrow Transplant 2014;49(6):744-50
  • Costa LJ, Zhang MJ, Zhong X, et al. Trends in utilization and outcomes of autologous transplantation as early therapy for multiple myeloma. Biol Blood Marrow Transplant 2013;19(11):1615-24
  • Martino M, Montanari M, Ferrara F, et al. Very low rate of readmission after an early discharge outpatient model for autografting in multiple myeloma patients: an Italian multicenter retrospective study. Biol Blood Marrow Transplant 2014;20(7):1026-32
  • Rajkumar SV. Doublets, triplets, or quadruplets of novel agents in newly diagnosed myeloma? Hematology Am Soc Hematol Educ Program 2012;2012:354-61
  • Cavo M, Tacchetti P, Patriarca F, 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;376:2075-85
  • Sonneveld P, Goldschmidt H, Rosinol L, et al. Bortezomib-based versus nonbortezomib-based induction treatment before autologous stem-cell transplantation in patients with previously untreated multiple myeloma: a meta-analysis of phase III randomized, controlled trials. J Clin Oncol 2013;31(26):3279-87
  • Bladé J, Samson D, Reece D, et al. Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation. Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant. Br J Haematol 1998;102(5):1115-23
  • Rajkumar SV. Multiple myeloma: 2011 update on diagnosis, risk-stratification, and management. Am J Hematol 2011;86(1):57-65
  • Paiva B, Gutierrez NC, Rosinol L, et al. High-risk cytogenetics and persistent minimal residual disease by multiparameter flow cytometry predict unsustained complete response after autologous stem cell transplantation in multiple myeloma. Blood 2012;119:687-91
  • Mohty M, Ho AD. In and out of the niche: perspectives in mobilization of hematopoietic stem cells. Exp Hematol 2011;39(7):723-9
  • Bensinger W, DiPersio JF, McCarty JM. Improving stem cell mobilization strategies: future directions. Bone Marrow Transplant 2009;43(3):181-95
  • Lemoli RM. New strategies for stem cell mobilization. Mediterr J Hematol Infect Dis 2012;4(1):e2012066
  • Tageja N, et al. Carfilzomib and bortezomib containing regimens yield high rates of MRD negative autologous hematopoietic progenitor cell (HPC) grafts in multiple myeloma (MM) and high risk smoldering myeloma (SMM). Blood [Annual Meeting Abstracts]. 2013. 122(21):Abstract 2030
  • Fitoussi O, Perreau V, Boiron JM, et al. A comparison of toxicity following two different doses of cyclophosphamide for mobilization of peripheral blood progenitor cells in 116 multiple myeloma patients. Bone Marrow Transplant 2001;27:837-42
  • Attal M, Harousseau JL, Facon T, et al. Single versus double autologous stem-cell transplantation for multiple myeloma. N Engl J Med 2003;349(26):2495-502
  • Harousseau JL. Hematopoietic stem cell transplantation in multiple myeloma. J Natl Compr Canc Netw 2009;7:961-70
  • Cavo M, Tosi P, Zamagni E, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: bologna 96 clinical study. J Clin Oncol 2007;25(17):2434-41
  • Neben K, Lokhorst HM, Jauch A, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood 2012;119(4):940-8
  • Naumann-Winter F, Greb A, Borchmann P, et al. First-line tandem high dose chemotherapy and autologous stem cell transplantation versus single high-dosechemotherapy and autologous stem cell transplantation in multiple myeloma, a systematic review of controlledstudies. Cochrane Database Syst Rev 2012;10:CD004626
  • Atanackovic D, Schilling G. Second autologous transplant as salvage therapy in multiple myeloma. Br J Haematol 2013;163(5):565-72
  • Fermand JP, Ravaud P, Chevret S, et al. High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood 1998;92:3131-6
  • Rajkumar SV, Jacobus S, Callander NS, et al. Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol 2010;11:29-37
  • McCarthy PL. Where are we going with autologous transplantation for multiple myeloma? Biol Blood Marrow Transplant 2013;19(11):1532-3
  • Palumbo A, Bringhen S, Kumar SK, et al. Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: a meta-analysis of individual patient data. Lancet Oncol 2014;15:333-42
  • Morgan GJ, Gregory WM, Davies FE, et al. National Cancer Research Institute Haematological Oncology Clinical Studies Group. The role of maintenance thalidomide therapy in multiple myeloma: MRC Myeloma IX results and meta-analysis. Blood 2012;119(1):7-15

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.