7
Blood Reviews 24 Suppl. 1 (2010) S13–S19 Contents lists available at ScienceDirect Blood Reviews journal homepage: www.elsevier.com/locate/blre REVIEW Lenalidomide mode of action: linking bench and clinical findings Faith Davies a, *, Rachid Baz b a The Institute of Cancer Research and Royal Marsden Hospital, United Kingdom b Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, United States article info Keywords: Multiple myeloma Immunomodulatory agent Tumoricidal activity Immunomodulation Lenalidomide Dexamethasone Dual mechanism of action abstract New effective strategies are required that specifically address the challenges of multiple myeloma (MM) treatment, namely, disease recurrence, immunosuppression, and treatment-related toxicities. Recent preclinical and clinical findings suggest that the IMiDs ® immunomodulatory compound lenalidomide has a dual mechanism of action, involving both a direct tumoricidal activity and immunomodulation, which may result in rapid and sustained control of MM, respectively. The tumoricidal effect of lenalidomide occurs through several mechanisms, including disruption of stromal support, induction of tumor suppressor genes, and activation of caspases. The immunomodulatory effects of lenalidomide, including T-cell and natural killer (NK)-cell activation, and increased expression of death effector molecules, lead to enhanced immune cell function and may explain the beneficial effects of this agent in the maintenance setting. Lenalidomide appears to be effective regardless of prior thalidomide treatment, which may reflect mechanistic differences – lenalidomide has greater immunomodulatory properties than thalidomide, whereas thalidomide has greater antiangiogenic activity. Recent studies also suggest that the concomitant use of dexamethasone may influence lenalidomide’s direct and immunomodulatory effects. Lenalidomide in combination with dexamethasone synergistically inhibits proliferation and induces apoptosis; however, dexamethasone appears to antagonize the immune-enhancing effect of lenalidomide. A study has demonstrated that a regimen of lenalidomide in combination with an optimal dose and schedule of dexamethasone may increase survival by allowing synergistic antiproliferative effects, without affecting immunomodulatory activity. As preclinical and clinical research continue, additional insights into the dual mechanism of action of lenalidomide will help to further optimize the use of lenalidomide in MM. © 2010 Elsevier Ltd. All rights reserved. 1. Introduction Multiple myeloma (MM) is a chronic disease characterized by the presence of residual tumor and immune suppression. 1–3 Although newer chemotherapeutic agents have improved response rates and survival outcomes, the median duration of response has not exceeded 3 years and patients continue to experience disease relapse. 4 New effective strategies are required that specifically address the challenges of MM, namely, disease recurrence, immunosuppression, and treatment-related toxicities. Furthermore, long-term treatment may be needed to prolong the duration of response, reduce tumor proliferation rate, and delay or circumvent drug resistance. The IMiDs ® immunomodulatory compounds lenalidomide and pomalidomide are novel agents for the treatment of myeloma. Although they are analogs of thalidomide, they have demonstrated potent anticancer properties and better tolerability profiles than *Corresponding author. The Institute of Cancer Research and Royal Marsden Hospital, 15 Cotswold Road, Sutton, Surrey SM2 5NG, United Kingdom. Tel.: +44 207 352 8133 (ext 4743); fax: +44 0208 642 9634. E-mail address: [email protected] (F. Davies). thalidomide (Table 1). 5–11 Of the IMiDs ® immunomodulatory compounds, lenalidomide has been most extensively studied and is approved for use in combination with dexamethasone for the treatment of patients with MM who have received at least one prior therapy. 12 The mechanism of action of IMiDs ® immunomodulatory compounds has been the subject of much recent research. Such investigations have highlighted that lenalidomide has dual effects, involving both direct tumoricidal properties and immunomodulatory activity (Fig. 1). This article will focus on the dual actions of lenalidomide, describing preclinical findings and relating these to recent clinical results in patients with MM. 2. Tumoricidal effects of lenalidomide 2.1. Preclinical data Lenalidomide exerts its tumoricidal effects through several mech- anisms, including decreased production of cytokine and growth factors leading to disruption of stromal support, 9,13 induction of tumor suppressor genes leading to cell cycle arrest, and activation of caspases triggering tumor cell apoptosis (Fig. 2). 14–18 0268-960X /$ – see front matter © 2010 Elsevier Ltd. All rights reserved.

Lenalidomide mode of action: linking bench and clinical findings

Embed Size (px)

Citation preview

Blood Reviews 24 Suppl. 1 (2010) S13–S19

Contents lists available at ScienceDirect

Blood Reviews

journal homepage: www.elsevier .com/locate /b l re

REVIEW

Lenalidomide mode of action: linking bench and clinical findings

Faith Daviesa, *, Rachid Bazb

a The Institute of Cancer Research and Royal Marsden Hospital, United Kingdomb Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, United States

a r t i c l e i n f o

Keywords:

Multiple myeloma

Immunomodulatory agent

Tumoricidal activity

Immunomodulation

Lenalidomide

Dexamethasone

Dual mechanism of action

a b s t r a c t

New effective strategies are required that specifically address the challenges of multiple myeloma

(MM) treatment, namely, disease recurrence, immunosuppression, and treatment-related toxicities.

Recent preclinical and clinical findings suggest that the IMiDs® immunomodulatory compound

lenalidomide has a dual mechanism of action, involving both a direct tumoricidal activity and

immunomodulation, which may result in rapid and sustained control of MM, respectively. The

tumoricidal effect of lenalidomide occurs through several mechanisms, including disruption

of stromal support, induction of tumor suppressor genes, and activation of caspases. The

immunomodulatory effects of lenalidomide, including T-cell and natural killer (NK)-cell activation,

and increased expression of death effector molecules, lead to enhanced immune cell function and

may explain the beneficial effects of this agent in the maintenance setting. Lenalidomide appears to

be effective regardless of prior thalidomide treatment, which may reflect mechanistic differences –

lenalidomide has greater immunomodulatory properties than thalidomide, whereas thalidomide

has greater antiangiogenic activity. Recent studies also suggest that the concomitant use of

dexamethasone may influence lenalidomide’s direct and immunomodulatory effects. Lenalidomide

in combination with dexamethasone synergistically inhibits proliferation and induces apoptosis;

however, dexamethasone appears to antagonize the immune-enhancing effect of lenalidomide.

A study has demonstrated that a regimen of lenalidomide in combination with an optimal dose

and schedule of dexamethasone may increase survival by allowing synergistic antiproliferative

effects, without affecting immunomodulatory activity. As preclinical and clinical research continue,

additional insights into the dual mechanism of action of lenalidomide will help to further optimize

the use of lenalidomide in MM.

© 2010 Elsevier Ltd. All rights reserved.

1. Introduction

Multiple myeloma (MM) is a chronic disease characterized by the

presence of residual tumor and immune suppression.1–3 Although

newer chemotherapeutic agents have improved response rates

and survival outcomes, the median duration of response has not

exceeded 3 years and patients continue to experience disease

relapse.4 New effective strategies are required that specifically

address the challenges of MM, namely, disease recurrence,

immunosuppression, and treatment-related toxicities. Furthermore,

long-term treatment may be needed to prolong the duration of

response, reduce tumor proliferation rate, and delay or circumvent

drug resistance.

The IMiDs® immunomodulatory compounds lenalidomide and

pomalidomide are novel agents for the treatment of myeloma.

Although they are analogs of thalidomide, they have demonstrated

potent anticancer properties and better tolerability profiles than

*Corresponding author. The Institute of Cancer Research and Royal

Marsden Hospital, 15 Cotswold Road, Sutton, Surrey SM2 5NG, United

Kingdom. Tel.: +442073528133 (ext 4743); fax: +4402086429634.

E-mail address: [email protected] (F. Davies).

thalidomide (Table 1).5–11 Of the IMiDs® immunomodulatory

compounds, lenalidomide has been most extensively studied

and is approved for use in combination with dexamethasone

for the treatment of patients with MM who have received

at least one prior therapy.12 The mechanism of action of

IMiDs® immunomodulatory compounds has been the subject of

much recent research. Such investigations have highlighted that

lenalidomide has dual effects, involving both direct tumoricidal

properties and immunomodulatory activity (Fig. 1). This article will

focus on the dual actions of lenalidomide, describing preclinical

findings and relating these to recent clinical results in patients with

MM.

2. Tumoricidal effects of lenalidomide

2.1. Preclinical data

Lenalidomide exerts its tumoricidal effects through several mech-

anisms, including decreased production of cytokine and growth

factors leading to disruption of stromal support,9,13 induction of

tumor suppressor genes leading to cell cycle arrest, and activation

of caspases triggering tumor cell apoptosis (Fig. 2).14–18

0268-960X /$ – see front matter © 2010 Elsevier Ltd. All rights reserved.

S14 F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19

Table 1

Characteristics of thalidomide and the immunomodulatory compounds lenalidomide and pomalidomide.7–11

Characteristic Thalidomide Immunomodulatory compounds

Lenalidomide Pomalidomide

Structure N

N

O

O

O

O N

NH

O O

O

NH2

N

NH

O

O O

O

NH2

Plasma Cmax, mM7,8 5.4 2.2a 0.19

Tumoricidal properties

Inhibition of DNA synthesis in MM.1S cell line, IC50, mM9

>100 0.1–1 0.01–0.1

Immunomodulation

Interleukin-2 enhancement, EC50, mM10

>100 0.15 0.010

Antiangiogenesis

Inhibition of sprout formation from human umbilical artery ring explants, IC50, mM11

~0.1 ~1.0 0.1–1.0

a Cmax reported in ng/mL.

Reducesmultiple myeloma

cell burden

Tum

or

burd

en

Time

Improves immunefunction andtumor killing

Lenalidomide

NH2

O

O

ONH

IM MU

NO

MO

DU

LA

TO

RY

TU

MO

RIC

ID

AL

N

Fig. 1. Lenalidomide has a dual mechanism of action involving a direct tumoricidal

activity that leads to multiple myeloma cell death, and an immunomodulatory

effect that keeps the tumor in remission and improves immune function. These

dual effects make lenalidomide an optimal foundation therapy for the necessary

long-term treatment of multiple myeloma. © Celgene Corporation.

The bone marrow microenvironment comprises a complex array

of physical and soluble factors that interact to promote MM cell

growth and survival, and may contribute to minimal residual

disease and the development of acquired drug resistance.19–21

Physical constituents of the bone marrow include extracellular

matrix glycoproteins and bone marrow stromal cells (BMSCs). The

extracellular matrix glycoprotein fibronectin has been shown to

control MM cell growth and survival, and plays a key role in cell-

adhesion-mediated drug resistance of MM cells.19,20 Fibronectin

adhesion activates a4b1 and a5b1 integrins, and signaling via

these integrin complexes confers resistance to several pro-apoptotic

effectors, including chemotherapeutic drugs and death receptor

activation.19,20 Adhesion of MM cells to BMSCs also induces secretion

of interleukin (IL)-6.21–23 IL-6 is known to mediate both MM cell

proliferation and the inhibition of Fas-induced apoptosis.22,24 In

a recent study using four MM cell lines, concomitant exposure

to IL-6 and fibronectin adhesion increased signal transducers

and activators of transcription (STAT) 3 phosphorylation, nuclear

translocation, and DNA binding – all of which are known to

lead to the transactivation of genes involved in proliferation,

differentiation, and survival.20 IL-6 also enhances the production

and secretion of vascular endothelial growth factor (VEGF) by MM

cells and vice versa.21–23 VEGF plays a role in tumor cell migration,

growth, and survival, and enhances microvascular density, which

correlates with disease progression and poor prognosis.13,21,22

Disruption of stromal support in myeloma is, therefore, an im-

portant treatment strategy. IMiDs® immunomodulatory compounds

have been shown to decrease the upregulation of IL-6 and VEGF

secretion induced by MM and BMSC interactions;9,23 this may

lead to disruption of stromal support and have subsequent effects

on MM cell migration, growth, and survival. A preclinical study

suggests that elotuzumab, a humanized antibody against CS1 (the

cell surface glycoprotein), can overcome cell-adhesion-mediated

drug resistance by altering extracellular-signal-regulated kinase

(ERK) 1/2 signaling of MM cells and osteoclasts.25 Furthermore,

in a recent phase I/II study, elotuzumab (5, 10, or 20mg/kg) in

combination with lenalidomide 25mg (days 1–21 of a 28-day cycle)

plus dexamethasone 40mg/week had an objective response rate

(defined as partial response [PR] and very good partial response

[VGPR]) of 82% and an overall response rate of 95% in lenalidomide-

naive patients,26 suggesting a combined beneficial effect on adverse

stromal interactions.

In addition to altering the bone marrow microenvironment,

IMiDs® immunomodulatory compounds have a direct effect on the

proliferation of MM cells via upregulation of tumor suppressor

genes.9,14,18 In a study by Gandhi et al., lenalidomide-mediated

growth inhibition was accompanied by induction of the cyclin-

dependent kinase (CDK) inhibitors p15, p16, p21, and p27, and

the early-response transcription factors Egr1, Egr2, and Egr3, in

cells derived from patients with MM.14 Lenalidomide also induced

p15, p21, Egr1, Egr2, and Egr3 expression in several MM cell

lines. Moreover, lenalidomide in combination with dexamethasone

synergistically upregulated these tumor suppressor genes, resulting

in inhibition of retinoblastoma protein phosphorylation, G0/G1 cell-

cycle arrest, and apoptosis.

Verhelle et al. investigated the effects of lenalidomide and

pomalidomide on proliferation of two myeloma cell lines (LP-1

and U266), the Namalwa lymphoma cell line, and normal

CD34+ progenitor cells.18 In cancer cell lines, both IMiDs®

immunomodulatory compounds inhibited proliferation, which was

associated with upregulation of p21 and inhibition of CDK activity,

leading to retinoblastoma hypophosphorylation and G0/G1 cell-

cycle arrest. In contrast, treatment of normal CD34+ cells with

these compounds led to p21 upregulation without CDK inhibition,

and there was significant expansion of normal CD34+ progenitor

cells. These findings may help explain the differential selective

effects of lenalidomide on malignant cells (tumoricidal effect) and

their normal counterparts (absence of suppression of bone marrow

progenitor cells). Such effects have been demonstrated in patients

with myelodysplastic syndrome where lenalidomide was shown

F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19 S15

G0/G1arrest

ChromatinHistone

M

G2

S

G1LENALIDOMIDE

IL-6

VEGF

Myeloma cell

Stromal

Cell

Integrin

Tumor suppressor

gene activation

Reduced

IL-6 and VEGF

expressionReduced MM cell migration,

growth, and survival

Inhibition of tumor cell proliferation

Cell-cycle arrest

Caspase activation

Tumor cell

apoptosis

+ DEXAMETHASONE

T

NK

NKTB

IgA

LENALIDOMIDE

IMMUNOMODULATORY

MM cells

Activated immune cells

release cytokines

and proliferate

Expansion of immunecells and improved function

T

NK

NKTNKT

TT

TNK

NKNK

NK

NKT

IFN-у

IL-2

Enhancement of humoral and

vaccine responses

Activated immune cells

target and trigger

MM cell apoptosis

B

TUMORICIDALA

NH2

N

o

o

oNH

NH2

N

o

o

oNH

NH2

N

o

o

oNH

Fig. 2. Overview of the mechanism of action of lenalidomide in MM, which leads to tumor cell death and increased immune response. IFN: interferon; Ig: immunoglobulin;

IL: interleukin; MM: multiple myeloma; NK: natural killer; VEGF: vascular endothelial growth factor. © Celgene Corporation.

to not only reduce the number of malignant cells, but also to

restore bone marrow function leading to improved hemopoiesis and

transfusion independence.27

A recent study demonstrated that IMiDs® immunomodulatory

compounds increase p21 expression by a novel epigenetic

mechanism.28 Lenalidomide and pomalidomide were found to

regulate p21 expression at the transcriptional level via Egr1

and Egr2, and other GC-rich DNA transcription factors, Sp1

and Sp3. Through activation of lysine-specific demethylase 1,

the immunomodulatory compounds increased p21 expression by

reducing histone methylation and increasing histone acetylation at

the p21 promoter, thus increasing transcription factor access to the

DNA. In addition to inducing cell-cycle arrest, these compounds

have been shown to activate caspases, directly triggering tumor

cell death.14,15 In the study by Gandhi et al., lenalidomide activated

caspases 3, 8, and 9 to varying degrees in different MM cell

lines. Furthermore, lenalidomide plus dexamethasone produced

synergistic activation of caspases and induction of apoptosis. In

another study, lenalidomide-induced apoptosis was attenuated by a

caspase-8 inhibitor in MM.1S cells,15 providing further evidence for

the role of immunomodulatory compounds in caspase activation.

2.2. Clinical data

The tumoricidal actions of lenalidomide are thought to lead to

the initial high-quality clinical responses observed in patients with

MM.29–37 Table 2 summarizes data from recent studies of different

regimens for the treatment of newly diagnosed or relapsed/

refractory MM, and highlights the short time to response and high

response rates of patients treated with lenalidomide-containing

S16 F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19

Table 2

Response data for regimens investigated for the treatment of newly diagnosed or relapsed/refractory MM.

Regimen Newly diagnosed patients with MM

BiRD31 RD32 Rd32 MPR30 MP30 MPV38

Relapsed/refractory patients with MM

RD39 RAD35 V36 VPLD40

Median time to first response, months 1.8a − − 1.9 2.8 1.4 1.9 1 1.4 1.4

Median time to best response, months 7a − − − − 4.2 − − − −

CR+VGPR/nCR, % 74 50 40 33 12 41 32.3 60 13 27

a Mean values.

BiRD: clarithromycin, lenalidomide, and dexamethasone; CR: complete response; MM: multiple myeloma; MP: melphalan and prednisone; MPR: melphalan,

prednisone, and lenalidomide; MPV: melphalan, prednisone, and bortezomib; nCR: near CR; −: not reported; RAD: lenalidomide, adriamycin, and dexamethasone;

Rd: lenalidomide and low-dose dexamethasone; RD: lenalidomide and high-dose dexamethasone; V: bortezomib; VGPR: very good partial response;

VPLD: bortezomib and pegylated liposomal doxorubicin.

regimens.30–32,35,36,38–40 In 72 patients with newly diagnosed MM

receiving lenalidomide in combination with clarithromycin and

dexamethasone (BiRD) as first-line therapy for MM, the response

rate (VGPR plus complete response [CR] plus stringent CR) was

74%.31 Moreover, mean time to first response, defined as greater

than 50% decrease of M-protein, was 53.9 days (range 21–816)

and mean time to maximum response was 209 days (range 27–

850). Furthermore, a recent study by Rajkumar et al. compared

lenalidomide in combination with either low-dose dexamethasone

(160mg per 28-day cycle) or high-dose dexamethasone (480mg

per 28-day cycle) in 445 patients with newly diagnosed MM.

This study demonstrated a CR-plus-VGPR rate of 40% and 50% in

lenalidomide plus low-dose dexamethasone and lenalidomide plus

high-dose dexamethasone, respectively.32 As part of a phase III

study by Palumbo et al., the combination of melphalan and

prednisone (MP) (n =154) versus MP plus lenalidomide (MPR)

(n =153) was investigated in the initial treatment of elderly patients

with newly diagnosed MM.30 In an interim analysis, the CR-plus-

VGPR rate was significantly higher in the MPR arm versus the

MP arm (33% versus 12%, respectively; p < 0.001). Similarly, median

time to first response was significantly shorter in the MPR arm

compared with the MP arm (1.9 versus 2.8 months; p < 0.001).30

This study also investigated the effect of continuous lenalidomide

treatment on efficacy outcomes, which will be discussed in a

separate section.

In the relapsed/refractory MM setting, two large, multicenter, ran-

domized, double-blind, placebo-controlled phase III trials (MM-009

and MM-010) compared the efficacy of lenalidomide 25mg (days

1–21 of each 28-day cycle) plus dexamethasone 40mg (days 1–4,

9–12, and 17–20 of the first four cycles, then days 1–4 only

of subsequent cycles) versus placebo plus dexamethasone 40mg

continued until disease progression.33,41 In a recent pooled analysis

of these studies (n = 704), patients treated with lenalidomide plus

dexamethasone had a significantly improved overall response rate

(ORR; 60.6% versus 21.9%; p < 0.001) and CR rate (15.0% versus

2.0%; p < 0.001) compared with patients who received placebo

plus dexamethasone (Fig. 3).39 The time to first response was

similar in patients treated with lenalidomide plus dexamethasone

and those treated with placebo plus dexamethasone (median

1.9 versus 2.0 months, respectively). However, lenalidomide

treatment was associated with significant improvements in time

to progression (median 13.4 versus 4.6 months; p < 0.001) and

duration of response (median 15.8 versus 7.0 months; p < 0.001).

At a median follow-up of 48 months for surviving patients, a

significant benefit in overall survival (OS; median 38.0 versus

31.6 months; p =0.045) was observed despite 47.6% of placebo-

plus-dexamethasone patients crossing over to lenalidomide-based

therapy after disease progression or study unblinding.

Also in the relapsed/refractory MM setting, the speed of

response to lenalidomide was evaluated using a similar regimen

of lenalidomide plus dexamethasone in an open-label phase IIIb

Fig. 3. Pooled analysis of responses from two phase III trials of lenalidomide

plus dexamethasone versus placebo plus dexamethasone in patients with

relapsed/refractory multiple myeloma.39 CR: complete response; ORR: overall

response rate; PR: partial response; VGPR: very good PR.

study in 150 patients with MM who had received at least one

previous therapy.34 Preliminary results indicate that a 50% reduction

in M-protein or serum free light chain levels was reached by half

of the patients within 28 days, and by 72% of the patients within

32 days. Furthermore, a PR or better was observed in 74% of

patients.

3. Immunodulatory effects of lenalidomide

3.1. Preclinical data

In addition to the rapid tumoricidal effects, lenalidomide provides

sustained disease control by improving immune function, resulting

in continued tumor killing (Fig. 2).42–50 One mechanism by

which lenalidomide exerts immunomodulatory effects is via

enhanced antigen-specific CD8+ T-cell cytolysis.43 The effects

of lenalidomide and thalidomide on CD8+ T-cell activity in

response to viruses were investigated in an in vitro dendritic cell/

CD8+ T-cell co-culture system.43 Thalidomide, and to a greater

extent lenalidomide, enhanced the lysis of influenza virus matrix

peptide-pulsed dendritic-cell targets, with lenalidomide also

stimulating a detectable cytomegalovirus-specific lytic response.

The enhancement of antiviral response was thought to be due

to IL-2-stimulated expansion of antigen-specific memory-effector

CD8+ T cells.

Another important mechanism by which lenalidomide induces

immunomodulation is by increasing NK-cell expression of death

effector molecules.44 In a recent study, NK cells were cultured

in the presence of immobilized immunoglobulin G, IL-2, and

increasing concentrations of lenalidomide.44 Lenalidomide was

found to stimulate NK-cell activity by enhancing Fc-g receptor

signaling, which, in turn, led to increased production of granzyme

B and increased Fas-ligand expression on NK cells. Lenalidomide

has also been shown to have beneficial effects on NKT cells.45

Lenalidomide enhanced antigen-specific expansion of NKT cells in

response to the NKT ligand a-galactosylceramide in both healthy

F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19 S17

donors and in patients with MM. Moreover, NKT cells activated

by lenalidomide were found to have greater ability to secrete

interferon (IFN)-g. These findings are important given that the

antitumor activity of NKT cells is associated with their ability to

secrete IFN-g.

3.2. Clinical data

A recent study assessed the effect of lenalidomide combination

therapy on NK/NKT cell numbers and receptor expression.51 Ten

patients with newly diagnosed or relapsed/refractory MM received

lenalidomide 25mg (days 1–21), dexamethasone 40mg (days

1, 8, 15, and 22) and cyclophosphamide (500mg on day 1 in

cycle 1, and subsequently on days 1 and 8 of each cycle).

Despite the concomitant administration of immunosuppressive

drugs, lenalidomide treatment increased the number of NK and

NKT cells by 73% and 45% versus baseline, respectively, very early

in the course of treatment (at the end of cycle 1). In addition, there

was increased expression of the activating receptors NKG2D, NKp30,

and NKp44 on NK cells, and NKG2D on NKT cells.

The immunomodulatory effects of lenalidomide have been

demonstrated in other clinical studies.46,47,52,53 In a study by Lioznov

et al., the effects of lenalidomide salvage therapy (15–25mg/day

on days 1–21 of each 28-day cycle) were assessed on T and NK

cells after allogeneic stem cell transplantation (SCT).46 Significant

increases in the levels of activated T cells (p=0.005), regulatory

T cells (p =0.0036), and activated NK cells (p =0.0008) were

observed in patients after starting lenalidomide therapy (n =8).

Maximum levels of activated T and NK cells were achieved

approximately 3 months after starting lenalidomide therapy,

and maximum levels of regulatory T cells were reached after

approximately 6 months. Moreover, in a phase I study of heavily

treated patients with metastatic malignant melanoma and other

advanced cancers, evidence of T-cell activation was indicated

by significantly increased serum levels of soluble IL-2 receptor,

granulocyte-macrophage colony-stimulating factor, IL-12, tumor

necrosis factor-a, and IL-8 in 9 patients who received lenalidomide

5–50mg for 28 days.47

There is also evidence that lenalidomide improves the humoral

immune system. The use of lenalidomide before pneumococcal

vaccination in patients with advanced/relapsed MM was found

to increase antigen-specific antibodies and T-cell responses

compared with patients who were vaccinated prior to receiving

lenalidomide.54 The immunomodulatory effects of lenalidomide

observed in preclinical and clinical studies may be the main

contributors to the beneficial effects of this agent in the

maintenance setting.30,55–58 In a retrospective pooled analysis of

trials MM-009 and MM-010, over half of patients with an initial

documented PR subsequently achieved CR/VGPR, and patients who

achieved a PR by cycle 6 still had a substantial probability (38%)

of achieving a CR/VGPR with continued treatment.55 Continued

treatment was not associated with increased toxicity, although the

tolerability of the regimen may be further improved by reducing

the dose of dexamethasone.59

Findings from studies in MM patients after autologous SCT also

suggest that lenalidomide may be a highly effective maintenance

treatment.57,58 In one recent study, 614 patients were randomized

to receive consolidation therapy with lenalidomide (25mg/day for

21 days of each 28-day cycle for 2 months) followed by maintenance

with either placebo or lenalidomide (10–15mg/day) until relapse.

Maintenance with lenalidomide significantly improved 3-year

progression-free survival (PFS) rates versus consolidation therapy

alone (68% versus 35%; hazard ratio [HR] = 0.46; p < 0.000001).

This benefit was observed irrespective of achieving a CR after

SCT, indicating a true maintenance effect.57 In a phase III study

from the Cancer and Leukemia Group B (CALGB), patients with

newly diagnosed MM received (at day 100–110 after autologous

SCT) either lenalidomide (starting dose 10mg/day, escalated to

15mg/day after 3 months) or placebo maintenance therapy until

disease progression.58 During an interim analysis based on 28% PFS

at a median follow-up of 12 months, progression was noted in

13.8% of patients (29 of 210) receiving maintenance lenalidomide

treatment compared with 27.9% (58 of 208) for patients who

received placebo (p < 0.0001), representing a 58% risk reduction for

disease progression. The estimated median time to progression was

25.5 months in the placebo arm, but it had not been reached in the

lenalidomide arm.

Furthermore, in an interim analysis of the study of elderly

patients with newly diagnosed MM by Palumbo et al., PFS was

13.0 months in patients who received MP alone, 13.2 months

in patients who received MPR followed by placebo, and not

reached in patients who received MPR followed by maintenance

lenalidomide therapy (MPR-R) (p < 0.001 for MPR-R versus MP).30

A landmark analysis of PFS in patients remaining on therapy

following cycle 9 demonstrated that maintenance treatment with

lenalidomide versus placebo resulted in a 75% reduced risk of

disease progression (HR=0.245; 95% confidence interval [CI] 0.126–

0.476; p < 0.001). Taken together, these findings indicate that

continuous lenalidomide therapy may provide sustained disease

control. To date no benefit in OS has been observed in the

maintenance setting.

4. Differences in the mode of action of thalidomide and

IMiDs® immunomodulatory compounds

Table 1 provides an overview of pharmacokinetic and pharmacody-

namic characteristics of thalidomide and the IMiDs® immunomod-

ulatory compounds. Of note, lenalidomide and pomalidomide

have greater tumoricidal effects than thalidomide. In an in vitro

study in MM.1S cells, DNA synthesis was inhibited by 50%

with pomalidomide 0.01–0.1mM and lenalidomide 0.1–1.0mM;

however, only 15% inhibition was noted with thalidomide 100mM.9

Lenalidomide also has a more potent effect on the immune system,

specifically by increased stimulation of T-cell proliferation and

cytokine production.60 Indeed, lenalidomide has been found to be

100–1000 times more potent in stimulating T-cell proliferation

and IFN-g and IL-2 production than thalidomide.6 In contrast, both

thalidomide and pomalidomide were found to be more potent at

inhibiting sprout formation than lenalidomide when antiangiogenic

properties were assessed in a human umbilical explant model

(Table 1).11

Lenalidomide has a more favorable nonhematologic-toxicity

profile than thalidomide, exhibiting reduced somnolence, constipa-

tion, and peripheral neuropathy.60 Myelosuppression occurs more

frequently with lenalidomide than with thalidomide,60 but may

be overcome through the use of granulocyte colony-stimulating

factor or other agents.61–64 As lenalidomide is an analog of

thalidomide, there could be a question of possible resistance to

lenalidomide in patients who had relapsed after, or who were

refractory to, treatment with thalidomide.60 However, evidence now

indicates that clinical benefit from lenalidomide can be achieved

in thalidomide-treated and -refractory patients.60,65 In a post hoc

pooled analysis of trials MM-009 and MM-010, lenalidomide

plus dexamethasone was more effective than dexamethasone

alone, irrespective of prior thalidomide exposure.60 Furthermore,

lenalidomide plus dexamethasone showed considerable activity

in patients who had relapsed while on thalidomide (ORR 41.9%;

CR 6.5%) or had never previously responded to thalidomide

(ORR 50.0%; CR 5.0%). This apparent lack of resistance observed

between lenalidomide and thalidomide may reflect their different

mechanisms of action.

S18 F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19

5. Influence of dexamethasone on the dual effects of

lenalidomide

Recent evidence suggests that coadministration of dexamethasone

may alter the dual effects of lenalidomide.9,14,16,17 As mentioned,

lenalidomide in combination with dexamethasone synergistically

inhibits myeloma cell growth, activates caspases, and induces

apoptosis,14 enhancing the tumoricidal effects of lenalidomide.

However, dexamethasone appears to antagonize the immune-

enhancing effect of lenalidomide by inhibiting the T- and NK-cell

costimulatory effects of lenalidomide.14 Specifically, dexamethasone

inhibited lenalidomide-enhanced IL-2 production in primary

human T cells, and inhibited lenalidomide-mediated IFN-g and

granzyme B production in primary human NK cells in a dose-

dependent manner in vitro.14

These findings may help to explain the results of the recent

phase III trial in patients with newly diagnosed MM by

Rajkumar et al., where low-dose dexamethasone plus lenalidomide

demonstrated a benefit in early OS compared with patients who

received lenalidomide plus high-dose dexamethasone.32 One-year

OS was 96% (95%CI 94–99) in the low-dose group compared with

87% (95%CI 82–92) in the high-dose group, and 2-year OS was 87%

(95%CI 81–93) and 75% (95%CI 68–93), respectively. Taken together,

results from the preclinical studies and clinical trial suggest that a

regimen of lenalidomide plus low-dose dexamethasone may result

in the best clinical activity by providing synergistic antiproliferative

effects, but without affecting the immunomodulatory effects of

lenalidomide.14 An alternative explanation for the prolonged OS

with the lower dose of dexamethasone could be related to the

tolerability profile. A lower dose would be associated with a better

safety profile, which may allow longer treatment exposure.

Alternatively, patients may benefit from receiving lenalidomide

alone or independent of corticosteroids. A recent retrospective

study evaluated the activity of lenalidomide monotherapy in newly

diagnosed MM.66 In 17 patients treated with lenalidomide without

corticosteroids, the ORR was 47% at a median follow-up of 7 months

(range 1–26). The median time to first response was 50 days (range

28–98) and median time to best response was 69 days (range 30–

591). Based on these positive findings, a phase II trial has been

initiated to investigate the role of a response-adapted approach

using single-agent lenalidomide as primary therapy for older

patients with newly diagnosed standard-risk MM (ClinicalTrials.gov

identifier: NCT00772915).

6. Conclusions

Current available data suggest MM requires continuous therapy

to reduce tumor burden and prolong survival. Lenalidomide

has the attributes of an agent that targets both tumor growth

and concomitant immunosuppression, while being well tolerated

and convenient for long-term use. Lenalidomide has potent

tumoricidal and immunomodulatory effects on myeloma cells and

their microenvironment, which are thought to result in both

rapid and sustained control of MM in the clinic. Lenalidomide,

in combination with dexamethasone, is active and generally

well tolerated in the relapsed/refractory MM setting and there

is evidence that continuous treatment with lenalidomide may

improve clinical response. Furthermore, lenalidomide appears to

be effective regardless of prior thalidomide treatment, which may

reflect the different mechanisms of the two agents – lenalidomide

has greater immunostimulatory properties and tolerability than

thalidomide, whereas thalidomide has more potent antiangiogenic

activity. Combined findings from preclinical and clinical studies

also provide insights into how the use of concomitant lenalidomide

and dexamethasone can be further optimized. The use of low-dose

dexamethasone may provide synergistic antiproliferative effects

without antagonizing lenalidomide-mediated immune effects,

which may explain the early survival benefit observed compared

with high-dose dexamethasone. As preclinical and clinical research

continues, additional insights into the dual mechanism of action of

lenalidomide will be obtained, which may help to further maximize

the use of lenalidomide in the treatment of MM.

7. Conflict of interest

Dr Davies has served on the Speakers Bureau for Celgene

Corporation and Ortho Biotech and has participated in Advisory

Boards for Celgene, Ortho Biotech, and Novartis. Dr Baz has

participated in Advisory Boards for, and has received research

funding from, Celgene Corporation.

Acknowledgements

The authors received editorial support from Excerpta Medica in the

preparation of this manuscript, funded by Celgene Corporation. The

authors are fully responsible for content and editorial decisions for

this manuscript.

References

1. Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma progenitors,

stem cell properties, and drug resistance. Cancer Res 2008;68:190–7.

2. Cook G, Campbell JD. Immune regulation in multiple myeloma: the host-tumour

conflict. Blood Rev 1999;13:151–62.

3. Schutt P, Brandhorst D, Stellberg W, et al. Immune parameters in multiple

myeloma patients: influence of treatment and correlation with opportunistic

infections. Leuk Lymphoma 2006;47:1570–82.

4. Attal M, Harousseau JL, Leyvraz S, et al. Maintenance therapy with

thalidomide improves survival in patients with multiple myeloma. Blood

2006;108:3289–94.

5. Chanan-Khan AA, Cheson BD. Lenalidomide for the treatment of B-cell

malignancies. J Clin Oncol 2008;26:1544–52.

6. Kotla V, Goel S, Nischal S, et al. Mechanism of action of lenalidomide in

hematological malignancies. J Hematol Oncol 2009;2:36.

7. Chung F, Lu J, Palmer BD, et al. Thalidomide pharmacokinetics and metabolite

formation in mice, rabbits, and multiple myeloma patients. Clin Cancer Res

2004;10:5949–56.

8. Chen N, Lau H, Kong L, et al. Pharmacokinetics of lenalidomide in subjects with

various degrees of renal impairment and in subjects on hemodialysis. J Clin

Pharmacol 2007;47:1466–75.

9. Hideshima T, Chauhan D, Shima Y, et al. Thalidomide and its analogs overcome

drug resistance of human multiple myeloma cells to conventional therapy. Blood

2000;96:2943–50.

10. Corral LG, Haslett PA, Muller GW, et al. Differential cytokine modulation and

T cell activation by two distinct classes of thalidomide analogues that are potent

inhibitors of TNF-alpha. J Immunol 1999;163:380–6.

11. Lu L, Payvandi F, Wu L, et al. The anti-cancer drug lenalidomide inhibits

angiogenesis and metastasis via multiple inhibitory effects on endothelial cell

function in normoxic and hypoxic conditions. Microvasc Res 2009;77:78–86.

12. Revlimid® (lenalidomide). Prescribing information. Celgene Corporation,

Summit, NJ. http://www.revlimid.com/pdf/REVLIMID_PI.pdf.

13. Hideshima T, Chauhan D, Podar K, Schlossman RL, Richardson P, Anderson KC.

Novel therapies targeting the myeloma cell and its bone marrow microenviron-

ment. Semin Oncol 2001;28:607–12.

14. Gandhi AK, Kang J, Capone L, et al. Dexamethasone synergizes with lenalidomide

to inhibit multiple myeloma tumor growth, but reduces lenalidomide-induced

immunomodulation of T and NK cell function. Curr Cancer Drug Targets 2010;

10:155–67.

15. Mitsiades N, Mitsiades CS, Poulaki V, et al. Apoptotic signaling induced by

immunomodulatory thalidomide analogs in human multiple myeloma cells:

therapeutic implications. Blood 2002;99:4525–30.

16. Schafer PH, Gandhi AK, Zhang LH, et al. Opposing effects of dexamethasone

on lenalidomide activity in multiple myeloma: additive/synergistic effects on

anti-proliferative activity on myeloma cells and antagonistic effects on immune

function [abstract]. Blood 2008;112: Abstract 2761.

17. Schafer PH, Gandhi AK, Zhang LH, et al. Opposing effects of dexamethasone

on lenalidomide activity in multiple myeloma [abstract]. Clin Lymph Myeloma

Suppl 2009;9(Suppl 1): Abstract B444.

18. Verhelle D, Corral LG, Wong K, et al. Lenalidomide and CC-4047 inhibit the

proliferation of malignant B cells while expanding normal CD34+ progenitor

cells. Cancer Res 2007;67:746–55.

19. Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion

mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis

in human myeloma cell lines. Blood 1999;93:1658–67.

F. Davies, R. Baz / Blood Reviews 24 (2010) S13–S19 S19

20. Shain KH, Yarde DN, Meads MB, et al. Beta1 integrin adhesion enhances IL-6-

mediated STAT3 signaling in myeloma cells: implications for microenvironment

influence on tumor survival and proliferation. Cancer Res 2009;69:1009–15.

21. Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma.

Lancet 2009;374:324–39.

22. Dankbar B, Padro T, Leo R, et al. Vascular endothelial growth factor and

interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma.

Blood 2000;95:2630–6.

23. Gupta D, Treon SP, Shima Y, et al. Adherence of multiple myeloma cells to bone

marrow stromal cells upregulates vascular endothelial growth factor secretion:

therapeutic applications. Leukemia 2001;15:1950–61.

24. Chauhan D, Kharbanda S, Ogata A, et al. Interleukin-6 inhibits Fas-induced

apoptosis and stress-activated protein kinase activation in multiple myeloma

cells. Blood 1997;89:227–34.

25. Tai YT, Fulciniti M, Hideshima T, et al. Targeting MEK induces myeloma-cell

cytotoxicity and inhibits osteoclastogenesis. Blood 2007;110:1656–63.

26. Lonial S, Vij R, Harousseau J, et al. Elotuzumab in combination with lenalidomide

and low-dose dexamethasone in relapsed or refractory multiple myeloma:

a phase I/II study [abstract]. J Clin Oncol 2010;28(15s): Abstract 8020.

27. List A, Dewald G, Bennett J, et al. Lenalidomide in the myelodysplastic syndrome

with chromosome 5q deletion. N Engl J Med 2006;355:1456–65.

28. Escoubet-Lozach L, Lin IL, Jensen-Pergakes K, et al. Pomalidomide and

lenalidomide induce p21 WAF-1 expression in both lymphoma and multiple

myeloma through a LSD1-mediated epigenetic mechanism. Cancer Res

2009;69:7347–56.

29. Palumbo A, Falco P, Corradini P, et al. Melphalan, prednisone, and lenalidomide

treatment for newly diagnosed myeloma: a report from the GIMEMA-Italian

Multiple Myeloma Network. J Clin Oncol 2007;25:4459–65.

30. Palumbo A, Dimopoulos MA, Delforge M, et al. A phase 3 study to determine the

efficacy and safety of lenalidomide combined with melphalan and prednisone in

patients ≥65 years with newly diagnosed multiple myeloma (NDMM) [abstract].

Haematologica 2010;95(Suppl 2): Abstract 0566.

31. 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;111:1101–9.

32. 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.

33. Dimopoulos M, Spencer A, Attal M, et al. Lenalidomide plus dexamethasone for

relapsed or refractory multiple myeloma. N Engl J Med 2007;357:2123–32.

34. Weisel KC, Hanel M, Niederwieser D, et al. Speed of response with lenalidomide

and dexamethasone in patients with relapsed or refractory multiple myeloma:

first results of the MM-019 German compassionate use protocol [abstract].

Haematologica 2009;94(Suppl 2): Abstract 0397.

35. Knop S, Gerecke C, Liebisch P, et al. Lenalidomide, adriamycin, and

dexamethasone (RAD) in patients with relapsed and refractory multiple

myeloma: a report from the German Myeloma Study Group DSMM (Deutsche

Studiengruppe Multiples Myelom). Blood 2009;113:4137–43.

36. Richardson PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose

dexamethasone for relapsed multiple myeloma. N Engl J Med 2005;352:

2487–98.

37. Morgan GJ, Schey SA, Wu P, et al. Lenalidomide (Revlimid), in combination

with cyclophosphamide and dexamethasone (RCD), is an effective and tolerated

regimen for myeloma patients. Br J Haematol 2007;137:268–9.

38. San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and

prednisone for initial treatment of multiple myeloma. N Engl J Med 2008;359:

906–17.

39. Dimopoulos MA, Chen C, Spencer A, et al. Long-term follow-up on overall

survival from the MM-009 and MM-010 phase III trials of lenalidomide plus

dexamethasone in patients with relapsed or refractory multiple myeloma.

Leukemia 2009;23:2147–52.

40. Orlowski RZ, Nagler A, Sonneveld P, et al. Randomized phase III study of

pegylated liposomal doxorubicin plus bortezomib compared with bortezomib

alone in relapsed or refractory multiple myeloma: combination therapy

improves time to progression. J Clin Oncol 2007;25:3892–901.

41. Weber DM, Chen C, Niesvizky R, et al. Lenalidomide plus dexamethasone for

relapsed multiple myeloma in North America. N Engl J Med 2007;357:2133–42.

42. Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory

derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood

2001;98:210–6.

43. Haslett PA, Hanekom WA, Muller G, Kaplan G. Thalidomide and a thalidomide

analogue drug costimulate virus-specific CD8+ T cells in vitro. J Infect Dis 2003;

187:946–55.

44. Wu L, Adams M, Carter T, et al. Lenalidomide enhances natural killer cell

and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-

treated CD20+ tumor cells. Clin Cancer Res 2008;14:4650–7.

45. Chang DH, Liu N, Klimek V, et al. Enhancement of ligand-dependent activation

of human natural killer T cells by lenalidomide: therapeutic implications. Blood

2006;108:618–21.

46. Lioznov M, El-Cheikh J Jr, Hoffmann F, et al. Lenalidomide as salvage therapy

after allo-SCT for multiple myeloma is effective and leads to an increase of

activated NK (NKp44(+)) and T (HLA-DR(+)) cells. Bone Marrow Transplant 2010;

45:349–53.

47. Bartlett JB, Michael A, Clarke IA, et al. Phase I study to determine the safety,

tolerability and immunostimulatory activity of thalidomide analogue CC-5013

in patients with metastatic malignant melanoma and other advanced cancers.

Br J Cancer 2004;90:955–61.

48. Reddy N, Hernandez-Ilizaliturri FJ, Deeb G, et al. Immunomodulatory drugs

stimulate natural killer-cell function, alter cytokine production by dendritic cells,

and inhibit angiogenesis enhancing the anti-tumour activity of rituximab in vivo.

Br J Haematol 2008;140:36–45.

49. Tai YT, Li XF, Catley L, et al. Immunomodulatory drug lenalidomide (CC-5013,

IMiD3) augments anti-CD40 SGN-40-induced cytotoxicity in human multiple

myeloma: clinical implications. Cancer Res 2005;65:11712–20.

50. Chiarenza A, Parrinello N, La Cava P, et al. Lenalidomide is able to restore

immune system in multiple myeloma patients [abstract]. Blood 2009;114:

Abstract 4909.

51. van Heteren P, Renz O, Hahn-Ast C, et al. Activation of innate immunity

in patients receiving combination therapy with lenalidomide (Revlimid),

cyclophosphamide and low-dose dexamethasone (RCD) for newly diagnosed

or relapsed/refractory myeloma [abstract]. Haematologica 2010;95(Suppl 2):

Abstract 0352.

52. Noonan KA, Ferguson A, Huff CA, et al. The immunomodulatory role of

lenalidomide on Prevnar® responses in patients with relapsed multiple

meyloma: a comprehensive analysis of the immune response [abstract]. Blood

2008;112: Abstract 2772.

53. Baz R, Dimopoulos M, Richardson P, Yu Z, Hussein M, Chanan-Khan A.

Lenalidomide-based therapy leads to improvement of humoral immune system

in relapsed or refractory multiple myeloma patients who respond to the therapy

[abstract]. Haematologica 2009;94(Suppl 2): Abstract 0395.

54. Noonan KA, Ferguson A, Emerling A, et al. Lenalidomide augments immune

responses to Prevnar vaccination in relapsed myeloma patients: implications

for cancer and infectious vaccines [abstract]. Blood 2009;114: Abstract 1864.

55. Harousseau JL, Dimopoulos MA, Wang M, et al. Better quality of response

to lenalidomide plus dexamethasone is associated with improved clinical

outcomes in patients with relapsed or refractory multiple myeloma.

Haematologica 2010;95:1738–44.

56. San Miguel JF, Dimopoulos MA, Stadtmauer EA, et al. Longer duration

of treatment and maintenance of best response with lenalidomide and

dexamethasone prolongs overall survival in patients with relapsed or refractory

multiple myeloma [abstract]. Blood 2008;112: Abstract 3702.

57. Attal M, Cristini C, Marit G, et al. Lenalidomide maintenance after transplanta-

tion for myeloma [abstract]. J Clin Oncol 2010;28(15s): Abstract 8018.

58. McCarthy PL, Owzar K, Anderson KC, et al. Phase III intergroup study of

lenalidomide versus placebo maintenance therapy following single autologous

stem cell transplant (ASCT) for multiple myeloma (MM): CALGB 100104

[abstract]. J Clin Oncol 2010;28(15s): Abstract 8017.

59. San Miguel JF, Dimopoulos M, Weber D, et al. Dexamethasone dose

adjustments seem to result in better efficacy and improved tolerability

in patients with relapsed/refractory multiple myeloma who are treated

with lenalidomide/dexamethasone (MM009/010 sub-analysis) [abstract]. Blood

2007;110: Abstract 2712.

60. Wang M, Dimopoulos MA, Chen C, et al. Lenalidomide plus dexamethasone is

more effective than dexamethasone alone in patients with relapsed or refractory

multiple myeloma regardless of prior thalidomide exposure. Blood 2008;112:

4445–51.

61. 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;14:795–8.

62. Palumbo A, Ben Yehuda D, Cavallo F, et al. Adequate mobilization of CD34 in

newly diagnosed multiple myeloma patients receiving lenalidomide induction

therapy [abstract]. Haematologica 2009;94(Suppl 2): Abstract 0707.

63. Talamo G, Claxton DF, Ehmann WC, Mierski J, Rybka W, Ibrahin S. Effect of

mobilization chemotherapy with cyclophosphamide and etoposide on stem cell

collection by lenalidomide [abstract]. J Clin Oncol 2009;27(15s): Abstract 7096.

64. Tarantolo SR, McSweeney PA, Micallef IN, Klein LM, Calandra G, Huebner D.

Plerixafor can predictably mobilize hematopoietic stem cells in patients with

multiple myeloma previously treated with lenalidomide [abstract]. Biol Blood

Marrow Transplant 2009;15(Suppl 1): Abstract 91.

65. Guglielmelli T, Bringhen S, Berruti A, et al. Sequential therapy with IMiD’s

in relapsed-refractory multiple myeloma patients [abstract]. Blood 2009;114:

Abstract 2888.

66. Baz R, Patel M, Finley-Oliver E, et al. Single agent lenalidomide in newly

diagnosed multiple myeloma: a retrospective analysis. Leuk Lymphoma

2010;51:1015–9.