The past decade has witnessed a paradigm shift in the initial treatment of multiple myeloma with the introduction of novel agents such as thalidomide, lenalidomide, and bortezomib, leading to improved outcomes. High-dose therapy and autologous stem cell transplantation remains an important therapeutic option for patients with multiple myeloma eligible for the procedure. Before the advent of the novel agents, patients underwent stem cell collection prior to significant alkylating agent exposure, given its potential deleterious effect on stem cell collection. With increasing use of the novel agents in the upfront setting, several reports have emerged raising concerns about their impact on the ability to collect stem cells. An expert panel of the International Myeloma Working Group (IMWG) was convened to examine the implications of these therapies on stem collection in patients with myeloma and to develop recommendations for addressing these issues. Here we summarize the currently available data and present our perspective on the problem and potential options to overcome this problem. Specifically, we recommend early mobilization of stem cells, preferably within the first 4 cycles of initial therapy, in patients treated with novel agents and encourage participation in clinical trials evaluating novel approaches to stem cell mobilization.

High-dose therapy and autologous stem cell transplantation (ASCT) remains an integral component of the myeloma treatment algorithm for patients considered eligible for the procedure. The majority of the randomized clinical trials have demonstrated a superior progression-free survival among patients undergoing ASCT compared with those treated with only conventional therapies and ASCT was associated with superior overall survival in 3 of those.1,,,,,7  Subsequent randomized trials have further defined the role of ASCT by demonstrating equivalent overall survival for delayed transplantation compared with upfront ASCT, albeit with some compromise in the quality-of-life parameters.8  Introduction of novel agents such as thalidomide, lenalidomide, and bortezomib have resulted in a paradigm change in the therapy of myeloma.9,,,,14  The high response rates with these agents, hitherto seen only in the context of high-dose therapy, have once again raised questions regarding the utility of ASCT in the setting of myeloma. Given the lack of long-term follow-up of patients treated with these new agents, the durability of these responses as well as their potential long-term adverse effects remain unknown and ASCT continues to be an important part of myeloma therapy.

Despite the increased use of the newer drugs for the initial treatment of myeloma, there is a continuous increase in the number of ASCTs reported to the Center for International Blood and Marrow Transplant Research (CIBMTR), highlighting their continued important role. Currently, the novel agents appear best suited to be used as first-line therapy, enhancing the quality of responses prior to proceeding to ASCT and diminishing early mortality from the disease, or in selected patients as primary therapy, moving ASCT to a second-line position or as adjuncts to transplant conditioning regimens or as maintenance therapy in patients undergoing ASCT.15,,,,,,,23  Furthermore, in a randomized trial evaluating single versus double ASCT a survival advantage with tandem ASCT was demonstrated in an unplanned subset analysis for those patients not obtaining at least a very good partial response after the first ASCT. This observation has increased the number of ASCTs being performed for patients not achieving very good partial response after the first ASCT.24  In addition, ASCT can also be used as part of second-line therapy after relapse, especially among patients who achieved a durable response after the first ASCT.6,23 

The traditional approach to patients with newly diagnosed myeloma, considered to be candidates for ASCT, has been to provide initial therapy with 4 to 6 cycles of a non–alkylator-containing regimen followed by collection of stem cells and high-dose therapy. The initial therapy for the disease allows time to obtain necessary insurance approvals as well as control disease-related symptoms, simultaneously controlling toxicity by limiting the number of cycles. In addition, adequate disease control provides an opportunity to reverse disease-related complications where feasible, and generally improve the functional status of the patient, allowing for a safer transplantation. Until the advent of the novel agents, the initial therapy regimens commonly used were vincristine, doxorubicin, and dexamethasone (VAD) or single-agent dexamethasone, both of which shared the advantage of having little impact stem cell mobilization and collection. Previous studies had shown that alkylating agents can potentially affect the stem cell pool and thus interfere with the ability to collect adequate numbers of stem cells.25,,28 

The number of CD34+ cells collected for ASCT is dependent on a number of factors, most importantly the number of intended transplantations. The cell collection target usually depends on patient age, upfront versus delayed ASCT, patient preference, patient performance status, and presence of comorbidities, among others. Traditionally, the target for CD34 cell collection for a single ASCT has been 4 to 6 × 106 CD34+/kg, with studies showing a deleterious impact on engraftment characteristics once the number falls below 2 × 106 CD34+/kg.29,30  Use of more CD34 cells has not been consistently associated with any significant benefit in the parameters studied. Patient age is an important factor from several perspectives. Clearly there is decreasing use of stem cell transplantation (SCT) with increasing patient age, although selected patients with good performance status may undergo transplantation into their mid-70s. The target for stem cell collection is usually based on single transplantation in the United States, because Medicare reimburses only single SCT for myeloma. Finally, there is a clear impact of age on the ability to collect stem cells, with decreasing yield with advancing age.31  In the majority of patients undergoing an ASCT for myeloma, stem cells are collected from the peripheral blood following mobilization using growth factor administration with or with out preceding chemotherapy. A minority of patients undergo ASCT with stem cells collected through a bone marrow harvest. Use of cyclophosphamide or other chemotherapy regimens for myelosuppression to achieve rebound CD34+ cell spillover into the blood with enhanced effects of myeloid growth factors during the recovery phase of peripheral blood counts allows for a more rapid stem cell collection and higher numbers of collected CD34+ cells compared with myeloid growth factor alone.32,,35  Institutions differ in their standard approach for collecting stem cells, with pros and cons to either approach. Use of cyclophosphamide, although allowing better stem cell collection and less likelihood of a collection failure (less than the 2 × 106 CD34+/kg), prolongs the collection process while awaiting count recovery and increases the risk of febrile neutropenia and other infectious complications.

One of the recent advances in the field of stem cell mobilization strategies has been the introduction of AMD3100 (plerixafor), a receptor chemokine receptor 4 (CXCR4) inhibitor. Previous studies have highlighted the role of CXCR4 in the stem cell mobilization induced by granulocyte colony-stimulating factor (G-CSF) and cyclophosphamide. Lévesque et al showed that mobilization of stem cells by G-CSF coincides in vivo with the cleavage of the N-terminus of the chemokine receptor CXCR4 on the stem cells in the BM, leading to loss of chemotaxis in response to the CXCR4 ligand, the chemokine stromal cell–derived factor-1 (SDF-1/CXCL12).36  In addition, accumulation of serine proteases led to cleavage and inactivation of SDF-1. Originally developed as an anti-HIV drug, the ability of this drug to enhance peripheral mobilization of CD34+ cells was subsequently recognized. AMD3100, a reversible inhibitor of the binding of stromal cell–derived factor-1α (SDF-1α, also known as CXCL12) to its cognate CXCR4, has been shown to increase the number of circulating CD34+ cells in healthy volunteers when administered alone or with G-CSF prior to treatment.37,39  Stem cells express CXCR4 and are known to migrate to the bone marrow through a chemoattractant effect of SDF-1α that is produced locally by bone marrow stromal cells. Once in the marrow, it is also believed that stem cell CXCR4 can act to help “anchor” these cells to stromal cell surface SDF-1α. AMD3100-induced leukocytosis and elevations in circulating hematopoietic progenitor cell levels are thought to result from a disruption of the CXCR4/CXCL12 interaction and cell adhesion effects, resulting in the appearance of both mature and pluripotent cells in the systemic circulation. AMD3100 has been shown to exert an additive effect on the number of circulating hematopoietic stem and progenitor cells when administered with G-CSF. AMD3100-3102 was a multicenter randomized, double-blind, placebo-controlled comparative trial designed to examine the ability of 240 μg/kg AMD3100 plus G-CSF versus placebo plus G-CSF to mobilize CD34+ stem cells in patients with multiple myeloma (MM), who had not previously failed stem cell collections and had not received previous stem cell transplant. The primary end point, the percentage of patients who achieved 6 × 106 or more CD34+ cells/kg in 2 or fewer apheresis days, was met in 106 (72%) of 128 patients in the AMD plus G-CSF group and 53 (34%) of 154 patients in the placebo plus G-CSF group, (P < .001). Fifty-four percent of study patients reached target after 1 day of apheresis in the AMD + G-CSF group compared with 17.3% in the placebo plus G-CSF group. After 4 days of apheresis, these numbers were 86.8% and 56%, respectively.40 

Introduction of thalidomide represented the first major therapeutic advance in myeloma in several decades. After the initial trials in relapsed myeloma, several randomized phase 3 trials of thalidomide and dexamethasone in patients with previously untreated myeloma were performed.15,18,41,42  Thalidomide combinations were associated with superior response rates and improved response duration with no definite impact on the overall survival compared with dexamethasone alone or VAD. This was followed by introduction of the thalidomide analog, lenalidomide, that in phase 2 trials resulted in very high response rates as well as deeper responses than were seen with previous approaches.11,43  Subsequent phase 3 trials of lenalidomide and dexamethasone demonstrated its superiority compared with dexamethasone alone as well as its ability to spare high doses of steroids and simultaneously improve survival.12,19  More recent clinical trials have examined the efficacy and tolerability of lenalidomide combined with cyclophosphamide (CTX), bortezomib, and clarithromycin, as well as other combinations.44,46  Another major advance in the field had been the introduction of the proteasome inhibitor bortezomib, which along with dexamethasone or in combination with conventional chemotherapy agents is increasingly being used in the setting of newly diagnosed disease with high efficacy. Phase 3 trials of bortezomib in combination with dexamethasone with or without doxorubicin has shown excellent tolerability and improved response rates and progression-free survival compared with traditional VAD in the setting of initial therapy prior to SCT.17,20,47  Both lenalidomide and bortezomib have been combined with cyclophosphamide in the setting of transplantation-eligible patients in phase 2 studies with excellent response rates.48,49  Recently reported phase 2 trials have examined the efficacy of combining bortezomib with lenalidomide or thalidomide with or without cyclophosphamide in the setting of newly diagnosed MM.50,,53  These combinations have led to very high complete and very good partial response rates and will undoubtedly become integral components of the initial treatment choices in the future. This in turn has led to renewed interest in the potential impact of initial therapy on the ability to collect adequate numbers of stem cells for one or more transplantations.

A large volume of data, albeit limited to single institution reports and less detailed data from phase 3 trials, has appeared in the past few years evaluating the effect of these new drugs on the stem cell collection process (Table 1). Although there are contradictory data on the impact of thalidomide on stem cell mobilization and collection, the effect if any appears to be relatively small with limited impact on the ability to proceed with SCT.15,18,33,54  In addition, there is no evidence to suggest that initial therapy with thalidomide-containing regimens prior to stem cell collection adversely impacts the engraftment potential of the collected stem cells.

In contrast to thalidomide, one of the common adverse effects of lenalidomide has been hematologic toxicity, especially myelosuppression. This finding raised concern that use of lenalidomide could adversely affect the ability to mobilize and collect adequate numbers of CD34+ cells for ASCT. In 2 large studies from Mayo Clinic and M. D. Anderson, the most significant factor influencing the ability to collect adequate numbers of stem cells appeared to be initial therapy with lenalidomide (Table 1).33,60  In addition to lenalidomide therapy, other important factors impacting the stem cell collection appeared to be the patient age and the duration of lenalidomide therapy.33,60  The failure rate of mobilization following lenalidomide therapy has varied significantly between the different studies, likely a reflection of the lenalidomide treatment duration, age of the patient population undergoing stem cell collection, mobilization regimens, and collection targets used. However, data so far do not indicate any impact on the quality of stem cells collected, as reflected in the engraftment kinetics as well as success.12,55,56 

The effect of bortezomib on the ability to collect stem cells has also been examined in the context of phase 2 and 3 trials examining the combinations (Table 1).17,21,49,61  Although no definite impact of initial therapy on stem cell harvest was demonstrated in the smaller phase 2 studies of bortezomib and dexamethasone,21  in the IFM 2005/01 trial comparing bortezomib/dexamethasone to VAD there was a trend toward lower CD34 numbers among those receiving bortezomib.17  In contrast, in the HOVON-65/GMMG-HD4 randomized phase 3 trial comparing bortezomib, adriamycin, dexamethasone (PAD) versus VAD as induction treatment, no impact of the regimen was seen on the ability to collect stem cells.62  No significant impact of initial therapy has been seen in other trials that have combined the novel agents, bortezomib in combination with either lenalidomide or thalidomide.51,63  Addition of alkylating agents to the initial therapy, especially in combination, may increase the risk of collection failures, but no comparative data are available.48,49  In a phase 2 study looking at the combination of lenalidomide with cytoxan and dexamethasone for newly diagnosed myeloma, we observed 8 of 30 failures at mobilization.48  In contrast, in a phase 2 study of cytoxan, bortezomib, and dexamethasone by Reeder et al, all patients who attempted stem cell collection were able to get enough cells for at least one transplantation.49 

Although there has been a wide spectrum of reported data on the initial therapy with a novel agent and the ability to collect stem cells, some common themes have emerged. In the 2 larger experiences published to date of lenalidomide therapy prior to harvest, the number of cycles of therapy appears to be important.33,60  Although none of the patients with fewer than 6 cycles of lenalidomide failed to collect stem cells in the Mayo Clinic series, more than 3 cycles of lenalidomide were associated with a higher risk in the M. D. Anderson series. However the smaller studies have not demonstrated such a relationship, and in the absence of detailed data from the larger prospective studies, it would be reasonable to assume that longer duration of therapy will increase the risk of failure. Another common finding has been the age of the patients, with more than one study demonstrating increased likelihood of failure in the older patients.33,60  In these 2 studies, no relationship was noted between the time off lenalidomide prior to stem cell harvest. Another important finding across the studies has been the low incidence of collection failure among patients mobilized with chemotherapy, typically cyclophosphamide, and G-CSF.58,59  Among 28 treatment-naive patients treated with the combination of clarithromycin, lenalidomide, and dexamethasone (BiRD) reported by Mark et al, sufficient stem cells for 2 autologous stem cell transplantations were collected from all patients mobilized with CTX plus G-CSF, versus 33% mobilized with G-CSF alone, demonstrating that this approach can potentially overcome the impairment in stem cell mobilization associated with lenalidomide.58  For patients failing initial attempts at stem cell mobilization with G-CSF alone, chemotherapy + G-CSF approach appears to have a reasonable efficacy. Five of 7 patients failing G-CSF alone were successfully mobilized with CTX + G-CSF in one study57  and 18 of 21 patients were remobilized successfully with a chemotherapy + G-CSF approach in another study.60  Mazumder et al also reported 3 patients who failed to collect successfully despite undergoing mobilization with the CXCR4 inhibitor plerixafor, but were subsequently collected using a combination of cyclophosphamide and G-CSF.55 

The exact mechanism why lenalidomide inhibits stem cell mobilization is not clear. Koh et al investigated the effects of lenalidomide, pomalidomide (CC4047) (IMiDs), and thalidomide on CD34+ hematopoietic progenitors. They showed in human colony formation assays and long-term culture-initiating cell tests that IMiDs and thalidomide are not toxic to hematopoietic stem cells and do not inhibit self-renewal capacity of stem cells.64  This makes it less likely that a direct toxic effect of lenalidomide on hematopoietic progenitors explains the limitation in stem cell collection. The group further showed that IMiDs promote myelopoiesis with a concomitant maturation stop of neutrophil granulocytes by down-regulation of critical transcription factors such as PU.1. This leads to an accumulation of immature granulocytes within the bone marrow compartment and neutropenia in the peripheral blood.65  Interestingly, the group also observed that the G-CSF secretion is highly up-regulated in cultures of hematopoietic progenitors treated with IMiDs (day 3 of treatment: control 140 pg/mL, lenalidomide 800 pg/mL, CC4047 1500 pg/mL)64  (S.L., written communication, September 2008). The biologic reason for the strong up-regulation of G-CSF is unknown. It is likely that self-regulatory mechanisms up-regulate G-CSF to overcome the maturation stop of granulocytes. Higher levels of G-CSF might lead to a tachyphylactic response, resulting in resistance to G-CSF mobilization. These findings are also supported by our observation that all other “non–G-CSF–based” mobilization approaches such as CTX and AMD3100 are successful in mobilizing a sufficient CD34+ cell number.

In June 2008, a panel of experts was convened by the International Myeloma Foundation to address issues regarding stem cell collection for autologous transplantation in patients receiving therapy with lenalidomide. The following statements reflect the considerations of the panel and the consensus recommendations formulated by the panel. The recommendations take into account the existing data suggesting compromised collection with the newer agents in some of the patients as well as the data, although limited, examining alternate approaches to stem cell mobilization. These recommendations will be revised when additional data become available, enabling us to make more specific recommendations.

First attempt: Given the potential for the novel agents to impact the ability of the stem cell collection, we recommend early stem cell mobilization when SCT is being contemplated immediately or later in the course of disease. Such an approach, after 3 to 4 cycles of initial therapy, is quite feasible given the rapid response seen with the new combinations. However, there exists considerable confusion at this point in terms of the mechanisms mediating the decreased collection as well as the best approaches to prevent this problem, and every effort should be made to enroll these patients in clinical trials evaluating these questions.

Among patients undergoing initial therapy with thalidomide or bortezomib in combination with dexamethasone or among those treated with lenalidomide and dexamethasone who have received fewer than 4 cycles of therapy and are younger than 65 years, G-CSF alone is considered adequate for the initial attempt at mobilizing stem cells, although many centers will continue to use cyclophosphamide and G-CSF as their standard protocol. Among those who have received more than 4 cycles of lenalidomide therapy, one should consider the initial use of cyclophosphamide and G-CSF for mobilization. This suggestion is based on the findings of increased failure risk in this population as well as the reduced risk of failure associated with the use of cyclophosphamide and G-CSF. Although the use of cyclophosphamide in all patients is likely to decrease the risk of failure at first attempt, the recommendation to use G-CSF alone in the former group is driven by the low risk of failure in that group, the increased risks and delay associated with use of cyclophosphamide, and finally the ability to successfully collect with cyclophosphamide and G-CSF in the few patients who fail the initial attempt with G-CSF alone. In patients older than 65 years old, we recommend consideration of reduced-dose cyclophosphamide with G-CSF or G-CSF alone with addition of AMD-3100 before the second leukapheresis if the first leukapheresis results in less than 2 million CD34+ cells/kg. In patients receiving other myelosuppressive drugs in combination with lenalidomide, cyclophosphamide and G-CSF should be considered for the initial attempt as the rate of failure increases in these situations. There are no data supporting additional time off therapy prior to mobilization enhancing the likelihood of a successful mobilization. We do not recommend a minimum period that patients have to be off lenalidomide prior to starting G-CSF for mobilization.

Failed stem cell collection

Among patients receiving initial therapy with lenalidomide-containing regimens failing to collect with G-CSF alone, there are 3 options for the subsequent attempt. The majority of the patients can be collected with cyclophosphamide priming and G-CSF. These patients will be candidates for use of AMD3100, which in combination with G-CSF has been very successful in mobilizing stem cells. Another approach includes the use of a combination of G-CSF and granulocyte-macrophage colony-stimulating factor (10 μg/kg per day subcutaneously for 2 days, followed by G-CSF 16 μg/kg per day subcutaneously until stem cell collection is complete).

Upfront use of plerixafor (AMD3100) in lenalidomide-treated patients

The panel discussed the question of routine use of plerixafor for mobilization in this patient group. It was felt that at this time, without a detailed cost benefit analysis, such a recommendation cannot be made and additional clinical trials specifically addressing its use in these patients will allow us to answer this question. Prospective trials should be conducted to study the use of plerixafor in patients failing to reach certain thresholds for peripheral blood CD34 counts.

The online version of this article contains a data supplement.

Contribution: K.C.A., P.G.R., R.N., A.P., E.A.S., L.B., S. Lonial, N.M., B.G.M.D., and S.V.R. attended Ad Board, collected data, and wrote the paper; S.K. attended Ad Board, wrote the paper (lead author), and revised the paper; and S.G., J.L.H., W.B., R.L.C., S. Lentzsch, J.S.M., H.L., J.B., P.T., P.S., O.S., D.V., M.C., and H.E. contributed comments and edited the paper

Conflict-of-interest disclosure: S.G. is on the Advisory Board for Celgene, Millennium, Novartis, and Genzyme; E.A.S. is on the Advisory Board for Genzyme; J.L.H. received Honoraria from Genzyme and Amgen, and is on the Advisory Board for Celgene and Janssen-Cilag; A.P. is on the Advisory Board for Ortho Biotech and Celgene; W.B. is on the Advisory Board for Celgene and Millennium, and received research funding from Genzyme, Millennium, Celgene, AstraZeneca, and Novartis; R.L.C. is on the Advisory Board for Millennium and Ortho Biotech; S.K. received clinical trial funding from Celgene, Millennium, and Genzyme; N.M. is on the Advisory Board for Celgene; R.N. received clinical trial funding from Celgene; J.S.M. is on the Advisory Board for Millennium, Janssen-Cilag, and Celgene; H.L. received clinical trial funding from Schering-Plough and Janssen-Cilag, and participated in the Speaker's Bureau for Amgen, Roche, and Janssen-Cilag; J.B. received honorarium for lectures, is on the Advisory Board for Celgene and Janssen-Cilag, and received a research grant from Celgene; S. Lonial is a consultant for Millennium, Celgene, Novartis, and BMS; H.E. is on the advisory Board for Celgene and Ortho Biotech; P.S. is on the Advisory Board for Ortho Biotech and Celgene; O.S. received clinical trial/research funding from Janssen-Cilag, Merck, and Novartis, and is on the Speaker's Bureau for Amgen, Celgene, Merck, Novartis, Ortho Biotech, Pharmion, and Roche; and P.G.R. and B.G.M.D. are on the Advisory Board for Celgene and Millennium. P.T., M.C., and S.V.R. declare no competing financial interests.

A complete list of IMWG participants can be found in the supplemental Appendix, available on the Blood website; see the Supplemental Materials link at the top of the online article.

Correspondence: Shaji Kumar, Associate Professor of Medicine, Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN 55906; e-mail: kumar.shaji@mayo.edu.

1
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
97
2
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
936
3
Bladé
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
3759
4
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
1883
5
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
9233
6
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
3136
7
Palumbo
A
Bringhen
S
Petrucci
MT
et al
Intermediate-dose melphalan improves survival of myeloma patients aged 50 to 70: results of a randomized controlled trial.
Blood
2004
104
3052
3057
8
Fermand
JP
Ravaud
P
Chevret
S
et al
Early versus late high dose therapy (HDT) and autologous peripheral blood stem cell transplantation in multiple myeloma (MM): results of a prospective randomized trial [abstract].
Blood
1996
88
suppl 1
685a
9
Rajkumar
SV
Blood
E
Vesole
DH
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
24
431
436
10
Rajkumar
SV
Hayman
S
Gertz
MA
et al
Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma.
J Clin Oncol
2002
20
4319
4323
11
Rajkumar
SV
Hayman
SR
Lacy
MQ
et al
Combination therapy with lenalidomide plus dexamethasone (Rev/Dex) for newly diagnosed myeloma.
Blood
2005
106
4050
4053
12
Rajkumar
SV
Jacobus
S
Callander
N
et al
Phase III trial of lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone in newly diagnosed multiple myeloma (E4A03): a trial coordinated by the Eastern Cooperative Oncology Group [abstract].
J Clin Oncol
2007
25
18S
LBA8025
(
ASCo Annual Meeting Abstracts
)
13
Richardson
P
Chanan-Khan
A
Schlossman
R
et al
A multicenter phase II trial of bortezomib in patients with previously untreated multiple myeloma: efficacy with manageable toxicity in patients with unexpectedly high rates of baseline peripheral neuropathy [abstract].
Blood
2005
106
2548
(
ASH Annual Meeting Abstracts
)
14
Richardson
P
Jagannath
S
Raje
N
et al
Lenalidomide, Bortezomib, and dexamethasone (Rev/Vel/Dex) as front-line therapy for patients with multiple myeloma (MM): preliminary results of a phase 1/2 study [abstract].
Blood
2007
110
187
(
ASH Annual Meeting Abstracts
)
15
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
24
431
436
16
Rajkumar
SV
Jacobus
S
Callander
N
et al
Randomized trial of lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone in newly diagnosed myeloma (E4A03), a trial coordinated by the Eastern Cooperative Oncology Group: analysis of response, survival, and outcome with primary therapy and with stem cell transplantation [abstract].
J Clin Oncol
2008
26
8504
(
Meeting Abstracts
)
17
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 [abstract].
J Clin Oncol
2008
26
8505
(
Meeting Abstracts
)
18
Cavo
M
Zamagni
E
Tosi
P
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
35
39
19
Zonder
JA
Crowley
J
Hussein
MA
et al
Superiority of lenalidomide (Len) plus high-dose dexamethasone (HD) compared to HD alone as treatment of newly-diagnosed multiple myeloma (NDMM): results of the randomized, double-blinded, placebo-controlled SWOG Trial S0232 [abstract].
Blood
2007
110
77
(
ASH Annual Meeting Abstracts
)
20
Sonneveld
P
van der Holt
B
Schmidt-Wolf
IGH
et al
First analysis of HOVON-65/GMMG-HD4 randomized phase iii trial comparing bortezomib, adriamycine, dexamethasone (PAD) vs VAD as induction treatment prior to high dose melphalan (HDM) in patients with newly diagnosed multiple myeloma (MM) [abstract].
Blood
2008
112
653
(
ASH Annual Meeting Abstracts
)
21
Jagannath
S
Durie
BG
Wolf
J
et al
Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma.
Br J Haematol
2005
129
776
783
22
Attal
M
Harousseau
JL
Leyvraz
S
et al
Maintenance therapy with thalidomide improves survival in patients with multiple myeloma.
Blood
2006
108
3289
3294
23
Abdelkefi
A
Ladeb
S
Torjman
L
et al
Single autologous stem-cell transplantation followed by maintenance therapy with thalidomide is superior to double autologous transplantation in multiple myeloma: results of a multicenter randomized clinical trial.
Blood
2008
111
1805
1810
24
Attal
M
Harousseau
JL
Facon
T
et al
Single versus double autologous stem-cell transplantation for multiple myeloma [see comment].
N Engl J Med
2003
349
2495
2502
25
Knudsen
LM
Rasmussen
T
Jensen
L
Johnsen
HE
Reduced bone marrow stem cell pool and progenitor mobilisation in multiple myeloma after melphalan treatment.
Med Oncol
1999
16
245
254
26
de la Rubia
J
Blade
J
Lahuerta
JJ
et al
Effect of chemotherapy with alkylating agents on the yield of CD34+ cells in patients with multiple myeloma: results of the Spanish Myeloma Group (GEM) Study.
Haematologica
2006
91
621
627
27
Boccadoro
M
Palumbo
A
Bringhen
S
et al
Oral melphalan at diagnosis hampers adequate collection of peripheral blood progenitor cells in multiple myeloma.
Haematologica
2002
87
846
850
28
Jansen
J
Thompson
J
Dugan
M
et al
Impaired PBPC collection in patients with myeloma after high-dose melphalan.
Cytotherapy
2004
6
498
504
29
Bensinger
W
Appelbaum
F
Rowley
S
et al
Factors that influence collection and engraftment of autologous peripheral-blood stem cells.
J Clin Oncol
1995
13
2547
2555
30
Desikan
KR
Tricot
G
Munshi
NC
et al
Preceding chemotherapy, tumour load and age influence engraftment in multiple myeloma patients mobilized with granulocyte colony-stimulating factor alone.
Br J Haematol
2001
112
242
247
31
Morris
CL
Siegel
E
Barlogie
B
et al
Mobilization of CD34+ cells in elderly patients (>/= 70 years) with multiple myeloma: influence of age, prior therapy, platelet count and mobilization regimen.
Br J Haematol
2003
120
413
423
32
Alegre
A
Tomas
JF
Martinez-Chamorro
C
et al
Comparison of peripheral blood progenitor cell mobilization in patients with multiple myeloma: high-dose cyclophosphamide plus GM-CSF vs G-CSF alone.
Bone Marrow Transplant
1997
20
211
217
33
Kumar
S
Dispenzieri
A
Lacy
MQ
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
21
2035
2042
34
Arora
M
Burns
LJ
Barker
JN
et al
Randomized comparison of granulocyte colony-stimulating factor versus granulocyte-macrophage colony-stimulating factor plus intensive chemotherapy for peripheral blood stem cell mobilization and autologous transplantation in multiple myeloma.
Biol Blood Marrow Transplant
2004
10
395
404
35
Hiwase
DK
Bollard
G
Hiwase
S
Bailey
M
Muirhead
J
Schwarer
AP
Intermediate-dose CY and G-CSF more efficiently mobilize adequate numbers of PBSC for tandem autologous PBSC transplantation compared with low-dose CY in patients with multiple myeloma.
Cytotherapy
2007
9
539
547
36
Lévesque
JP
Hendy
J
Takamatsu
Y
Simmons
PJ
Bendall
LJ
Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide.
J Clin Invest
2003
111
187
196
37
Donzella
GA
Schols
D
Lin
SW
et al
AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor.
Nat Med
1998
4
72
77
38
Liles
WC
Broxmeyer
HE
Rodger
E
et al
Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist.
Blood
2003
102
2728
2730
39
Devine
SM
Flomenberg
N
Vesole
DH
et al
Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin's lymphoma.
J Clin Oncol
2004
22
1095
1102
40
DiPersio
J
Stadtmauer
EA
Nademanee
AP
et al
A phase III, multicenter, randomized, double-blind, placebo-controlled, comparative trial of AMD3100 (Plerixafor)+G-CSF vs. G-CSF+placebo for mobilization in multiple myeloma (MM) patients for autologous hematopoietic stem cell (aHSC) transplantation [abstract].
Blood
2007
110
445
(
ASH Annual Meeting Abstracts
)
41
Rajkumar
SV
Rosinol
L
Hussein
M
et al
Multicenter, randomized, double-blind, placebo-controlled study of thalidomide plus dexamethasone compared with dexamethasone as initial therapy for newly diagnosed multiple myeloma.
J Clin Oncol
2008
26
2171
2177
42
Lokhorst
HM
Schmidt-Wolf
I
Sonneveld
P
et al
Thalidomide in induction treatment increases the very good partial response rate before and after high-dose therapy in previously untreated multiple myeloma.
Haematologica
2008
93
124
127
43
Lacy
M
Gertz
M
Dispenzieri
A
et al
Long-term results of response to therapy, time to progression, and survival with lenalidomide plus dexamethasone in newly diagnosed myeloma.
Mayo Clin Proc
2007
82
1179
1184
44
Kumar
S
Hayman
SR
Buadi
FK
et al
Phase II trial of lenalidomide, cyclophosphamide, and dexamethasone (CRd) for newly diagnosed myeloma. [abstract].
Blood
2007
110
190
(
ASH Annual Meeting Abstracts
)
45
Richardson
PG
Lonial
S
Jakubowiak
A
et al
Safety and efficacy of lenalidomide (Len), bortezomib (Bz), and dexamethasone (Dex) in patients (pts) with newly diagnosed multiple myeloma (MM): a phase I/II study [abstract].
J Clin Oncol
2008
26
8520
(
Meeting Abstracts
)
46
Niesvizky
R
Jayabalan
DS
Christos
PJ
et al
BiRD (Biaxin [clarithromycin]/Revlimid [lenalidomide]/dexamethasone) combination therapy results in high complete- and overall-response rates in treatment-naive symptomatic multiple myeloma.
Blood
2008
111
1101
1109
47
Rosiñol
L
Oriol
A
Mateos
MV
et al
Phase II PETHEMA trial of alternating bortezomib and dexamethasone as induction regimen before autologous stem-cell transplantation in younger patients with multiple myeloma: efficacy and clinical implications of tumor response kinetics.
J Clin Oncol
2007
25
4452
4458
48
Kumar
S
Hayman
S
Buadi
F
et al
Phase II trial of lenalidomide (RevlimidTM) with cyclophosphamide and dexamethasone (RCd) for newly diagnosed myeloma [abstract].
Blood
2008
112
91
(
ASH Annual Meeting Abstracts
)
49
Reeder
CB
Reece
DE
Kukreti
V
et al
Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial.
Leukemia
2009
23
1337
1341
50
Kumar
S
Flinn
IW
Noga
SJ
et al
Safety and efficacy of novel combination therapy with bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in newly diagnosed multiple myeloma: initial results from the phase I/II multi-center EVOLUTION Study [abstract].
Blood
2008
112
93
(
ASH Annual Meeting Abstracts
)
51
Richardson
P
Lonial
S
Jakubowiak
A
et al
Lenalidomide, bortezomib, and dexamethasone in patients with newly diagnosed multiple myeloma: encouraging efficacy in high risk groups with updated results of a phase i/ii study [abstract].
Blood
2008
112
92
(
ASH Annual Meeting Abstracts
)
52
Cavo
M
Tacchetti
P
Patriarca
F
et al
Superior complete response rate and progression-free survival after autologous transplantation with up-front velcade-thalidomide- dexamethasone compared with thalidomide-dexamethasone in newly diagnosed multiple myeloma [abstract].
Blood
2008
112
158
(
ASH Annual Meeting Abstracts
)
53
Rosinol
L
Cibeira
MT
Martinez
J
et al
Thalidomide/dexamethasone (TD) vs. bortezomib (Velcade(R))/thalidomide/dexamethasone (VTD) vs. VBMCP/VBAD/Velcade(R) as induction regimens prior autologous stem cell transplantation (ASCT) in younger patients with multiple myeloma (MM): first results of a prospective phase III PETHEMA/Gem Trial [abstract].
Blood
2008
112
654
(
ASH Annual Meeting Abstracts
)
54
Breitkreutz
I
Lokhorst
HM
Raab
MS
et al
Thalidomide in newly diagnosed multiple myeloma: influence of thalidomide treatment on peripheral blood stem cell collection yield.
Leukemia
2007
21
1294
1299
55
Mazumder
A
Kaufman
J
Niesvizky
R
Lonial
S
Vesole
D
Jagannath
S
Effect of lenalidomide therapy on mobilization of peripheral blood stem cells in previously untreated multiple myeloma patients.
Leukemia
2008
22
1280
1281
author reply: 1281–1282
56
Paripati
H
Stewart
AK
Cabou
S
et al
Compromised stem cell mobilization following induction therapy with lenalidomide in myeloma.
Leukemia
2008
22
1282
1284
57
Kumar
S
Lacy
M
Dispenzieri
A
et al
Stem cell mobilization following initial therapy with lenalidomide and dexamethasone in patients with newly diagnosed multiple myeloma [abstract].
Blood
2008
112
3467
(
ASH Annual Meeting Abstracts
)
58
Mark
T
Stern
J
Furst
JR
et al
Stem cell mobilization with cyclophosphamide overcomes the suppressive effect of lenalidomide therapy on stem cell collection in multiple myeloma.
Biol Blood Marrow Transplant
2008
14
795
798
59
Cook
RJ
Vogl
D
Mangan
PA
et al
Lenalidomide and stem cell collection in patients with multiple myeloma [abstract].
J Clin Oncol
2008
26
8547
(
Meeting Abstracts
)
60
Popat
U
Saliba
R
Thandi
R
et al
Impairment of filgrastim-induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma.
Biol Blood Marrow Transplant
2009
15
718
723
61
Jagannath
S
Durie
BG
Wolf
J
et al
Bortezomib therapy alone and in combination with dexamethasone for previously untreated symptomatic multiple myeloma.
Br J Haematol
2005
129
776
783
62
Goldschmidt
H
Lokhorst
HM
Bertsch
U
et al
Successful harvesting of peripheral hematopoietic stem cells after induction treatment with bortezomib, adriamycin, dexamethasone (PAD) in patients with newly diagnosed multiple myeloma (MM) [abstract].
Blood
2008
112
3470
(
ASH Annual Meeting Abstracts
)
63
Bensinger
W
Jagannath
S
Vescio
R
et al
A phase II study of bortezomib (Velcade (R)), cyclophosphamide (cytoxan(R)), thalidomide (thalomid(R)) and dexamethasone as first-line therapy for multiple myeloma [abstract].
Blood
2008
112
94
(
ASH Annual Meeting Abstracts
)
64
Koh
KR
Janz
M
Mapara
MY
et al
Immunomodulatory derivative of thalidomide (IMiD CC-4047) induces a shift in lineage commitment by suppressing erythropoiesis and promoting myelopoiesis.
Blood
2005
105
3833
3840
65
Anderson
G
Gries
M
Kurihara
N
et al
Thalidomide derivative CC-4047 inhibits osteoclast formation by down-regulation of PU.1.
Blood
2006
107
3098
3105
Sign in via your Institution