Lenalidomide is an immunomodulatory drug (IMiD) with activity in lymphoid malignancies occurring primarily through immune modulation (eg, T-cell immune synapse enhancement and NK-cell/T-cell effector augmentation) and antiproliferative effects. Food and Drug Administration–approved for bortezomib-resistant, relapsed/refractory mantle-cell lymphoma, lenalidomide has demonstrated efficacy in several additional lymphoma subtypes. There are many ongoing clinical trials examining the use of lenalidomide alone or in combinatorial therapy. It will be important in these studies to delineate reliable, predictive biomarkers to optimally integrate lenalidomide into lymphoma treatment paradigms.

Lenalidomide is an immunomodulatory drug (IMiD) that has been studied extensively in non-Hodgkin lymphoma (NHL). There have been a multitude of preclinical and clinical studies that have helped define the role of lenalidomide in NHL.

The most prominent mechanisms of action of lenalidomide in lymphoid malignancies appear to be immunomodulatory, antiproliferative, and antiangiogenic (Figure 1).

Figure 1

Mechanisms of action of lenalidomide in lymphoid malignancies. Direct antitumor effects primarily mediated by lenalidomide binding CRL4CRBN, altering affinity for E3-ubiquitin ligase substrates. Lymphoid transcription factors IKZF1 (Ikaros) and IKZF3 (Aiolos) are preferentially ubiquitinated and degraded rapidly with lenalidomide, causing decreased NF-κB, decreased MYC and IRF4, increased p21WAF1, and suppression of cell cycle via degradation of cyclin-dependent kinases. There is also increased interferon production via decreased IRF4 (suppresses IFN response), which promotes cellular death. Other CRL4CRBN substrates may be affected, but are less defined. Immunomodulatory properties: Improvement in T-cell and NK-cell antitumor activity is seen with lenalidomide, including IL-2–driven costimulation of T cells (via increased degradation of IKZF1 and IKZF3 in T cells). Regulatory T cells are suppressed and there is a skewing toward Th1 population with lenalidomide. The NK- and T-cell effects of lenalidomide in lymphoma are synergistically enhanced with rituximab in preclinical studies. Anti-angiogenic properties: lenalidomide decreases angiogenesis in part via decreased microvessel density and it inhibits tumor growth and dissemination of disease through depletion of monocytes and macrophages associated with lymphangiogenesis. ADCC, antibody-dependent cell-directed cytotoxicity; Cdc42, cell division control protein 42; CLL, chronic lymphocytic leukemia; CRL4CRBN, Cullen 4 ring-E3 ubiquitin ligase–cereblon complex; FL, follicular lymphoma; His H3-Me, histone H3 methylation; ICAM-1, intercellular adhesion molecule 1; IFN, interferon; IKZF1, Ikaros; IKZF3, Aiolos; IRF4, interferon regulatory factor 4; LFA-1, lymphocyte function–associated antigen 1; MHC, major histocompatibility complex; TCR, T-cell receptor; WASp, Wiskott-Aldrich syndrome protein.

Figure 1

Mechanisms of action of lenalidomide in lymphoid malignancies. Direct antitumor effects primarily mediated by lenalidomide binding CRL4CRBN, altering affinity for E3-ubiquitin ligase substrates. Lymphoid transcription factors IKZF1 (Ikaros) and IKZF3 (Aiolos) are preferentially ubiquitinated and degraded rapidly with lenalidomide, causing decreased NF-κB, decreased MYC and IRF4, increased p21WAF1, and suppression of cell cycle via degradation of cyclin-dependent kinases. There is also increased interferon production via decreased IRF4 (suppresses IFN response), which promotes cellular death. Other CRL4CRBN substrates may be affected, but are less defined. Immunomodulatory properties: Improvement in T-cell and NK-cell antitumor activity is seen with lenalidomide, including IL-2–driven costimulation of T cells (via increased degradation of IKZF1 and IKZF3 in T cells). Regulatory T cells are suppressed and there is a skewing toward Th1 population with lenalidomide. The NK- and T-cell effects of lenalidomide in lymphoma are synergistically enhanced with rituximab in preclinical studies. Anti-angiogenic properties: lenalidomide decreases angiogenesis in part via decreased microvessel density and it inhibits tumor growth and dissemination of disease through depletion of monocytes and macrophages associated with lymphangiogenesis. ADCC, antibody-dependent cell-directed cytotoxicity; Cdc42, cell division control protein 42; CLL, chronic lymphocytic leukemia; CRL4CRBN, Cullen 4 ring-E3 ubiquitin ligase–cereblon complex; FL, follicular lymphoma; His H3-Me, histone H3 methylation; ICAM-1, intercellular adhesion molecule 1; IFN, interferon; IKZF1, Ikaros; IKZF3, Aiolos; IRF4, interferon regulatory factor 4; LFA-1, lymphocyte function–associated antigen 1; MHC, major histocompatibility complex; TCR, T-cell receptor; WASp, Wiskott-Aldrich syndrome protein.

Close modal

Immunomodulatory

Elegant preclinical studies showed that T-cell immune synapse dysfunction in follicular lymphoma (FL) can be “repaired” with lenalidomide.1  Tumor-infiltrating T cells from FL patients had significant reduction in formation of the F-actin immune synapse vs healthy donor cells. Lenalidomide reversed these abnormalities by enhancing the immune synapse, an important finding also demonstrated in chronic lymphocytic leukemia (CLL).2  In addition, lenalidomide has been shown to reduce T regulatory cells, activate CD8 T cells, and skew T-helper (TH) subsets with TH1 > TH2 response.3 

In other NHL experiments, lenalidomide combined with rituximab resulted in antitumor effects via increased NK cell function, enhanced antibody-dependent cellular cytotoxicity, improved NK cell–mediated synapse formation, and CD20 capping.4-7  Zhu et al also reported that induction of apoptosis in Burkitt lymphoma (BL) and CLL cells occurred primarily through NK cell activation.8 

Antiproliferative effects

A significant discovery in the mechanism of lenalidomide was identification of the importance of cereblon (encoded by the CRBN gene). Ito et al first showed that CRBN was required for the teratogenic effects of thalidomide leading to downregulation of fibroblast growth factor.9  Subsequent research has shown that silencing of CRBN diminishes the effect of lenalidomide and results in resistant cancer cells.10  Gene expression studies with CRBN knockdown showed that many of the affected genes were targets of critical transcription factors (eg, MYC, SP1, and TP53). It has also been shown that myeloma cells are addicted to an interferon regulatory factor 4 (IRF4)-controlled network with MYC as a direct downstream target and an autoregulatory circuit involving MYC and IRF4.11 

Lenalidomide directly binds CRBN, reduces IRF4 and MYC expression, increases P21WAF-1 expression, and changes the immunomodulatory activity of T cells in myeloma models.12  Subsequent investigations with lenalidomide in diffuse large B-cell lymphoma (DLBCL) cells identified a direct tumoricidal effect with downregulation of IRF4 and SPIB transcription factors requiring CRBN expression.13,14  Moreover, this effect has been preferentially noted in nongerminal center (non-GC) DLBCLs, which are more dependent on IRF4 and NF-κB than GC-DLBCL because of oncogenic mutations in CARD11 and MYD88.13,15 

Cereblon has also been shown to be vital in lenalidomide-induced T-cell stimulation.16  Kronke et al showed that lenalidomide enhanced CRL4CRBN binding to Ikaros (IKZF1) and Aiolos (IKZF3) in myeloma and T-cell lines causing their ubiquitination and degradation (Figure 1).17  Ikaros and Aiolos are highly involved in the biology of T- and B-cells. Gandhi et al demonstrated in NHL cells that lenalidomide induced CRL4 interaction with and downregulated Ikaros and Aiolos, both transcriptional repressors of IL-2; the resultant IL-2 expression induced T-cell costimulation/activation. Lenalidomide also exerts direct antiproliferative activity in mantle-cell lymphoma (MCL)18  and CLL cells, the latter occurring in a cereblon- and p21-dependent, but p53-independent manner.19 

Antiangiogenic

Reddy et al showed that lenalidomide diminishes angiogenesis (decreased microvessel density) in NHL xenograft models.5  Related data in MCL mouse models revealed that lenalidomide inhibited tumor growth and dissemination by depleting monocytes and macrophages associated with lymphangiogenesis.20  Lenalidomide has also been shown to upregulate the tumor-suppressor gene SPARC, which has antiproliferative, antiadhesive, and antiangiogenic properties in 5q– syndrome21 ; similar preliminary findings were noted in NHL,22  which remains to be validated.

Phases 1, 2, and 3 clinical trials have defined the safety and efficacy of lenalidomide in multiple NHL subtypes including MCL, FL, DLBCL, and T-cell lymphoma (Table 1).23-46 

Mantle-cell lymphoma

Relapsed/refractory.

In a single-agent study (NHL-002)39  of relapsed/refractory aggressive NHL histologies, the overall response rate (ORR) of lenalidomide in MCL was 53%, with 14% complete remission (CR) (Table 1). Mature follow-up of NHL-002 showed a median duration of response (DOR) of 13.7 months.26  Eve et al treated 26 relapsed/refractory MCL cases with maintenance lenalidomide in responding patients to lenalidomide induction (Table 1).29  In NHL-003, 57 relapsed/refractory MCL patients had an ORR of 42%.47  Collectively, adverse effects (AEs) were primarily hematologic, with 5% of patients having thromboembolic events. Notably, the risk of thromboembolism in NHL does not differ from the risk in multiple myeloma. Longer-term follow-up of NHL-003 (median 20 months) showed a DOR of 16.3 months and progression-free survival (PFS) of 8.8 months in this heavily pretreated group of patients.47  Second malignancies were noted in 4 of 57 patients (2 shortly after starting lenalidomide). Continued follow-up of this and other lenalidomide studies are needed to examine the risk of second cancers and other late effects.

A prospective, single-arm, phase 2 International clinical trial (EMERGE, MCL-001) of 134 bortezomib-resistant, relapsed/refractory MCL patients showed meaningful clinical benefit across multiple prognostic groups to single-agent lenalidomide.30  Grade 3-4 AEs were similar to prior trials (43% neutropenia, 28% thrombocytopenia, 11% anemia). Grade 3 nonhematologic AEs included 7% fatigue, 6% diarrhea, and 5% dyspnea. These data led to Food and Drug Administration approval of lenalidomide in June 2013 for MCL patients whose disease relapsed or progressed after 2 prior therapies, one of which included bortezomib.

In an update to MCL-001, median time to CR was 4.1 months and median overall survival (OS) was 20.9 months.48  Dose reductions or interruptions as a result of AEs occurred in 40% and 58% of patients, respectively, and the average lenalidomide dose intensity was 20 mg per day. In addition, lower Ki67 levels (<30%) appeared to correlate with improved CR, DOR, and OS.

Untreated.

There are ongoing clinical trials incorporating lenalidomide into treatment paradigms for untreated MCL (supplemental Tables 1 and 2, available on the Blood Web site). Jerkeman et al evaluated lenalidomide combined concurrently with bendamustine and rituximab (BR) in newly-diagnosed older MCL patients.23  The phase 1 study identified an unexpected high rate of grade 3-4 allergic reactions and cutaneous toxicity, and there were 3 treatment-related deaths (Table 1). The study was amended to omit lenalidomide from cycle 1 (ie, starting cycle 2). Ruan et al presented data using lenalidomide combined with rituximab for first-line treatment of MCL.24  Treatment was well tolerated with grade 3-4 toxicities of 39% neutropenia, 13% thrombocytopenia, 7% anemia, 23% rash, and 7% tumor flare. Preliminary efficacy data were promising (Table 1).

Follicular lymphoma

Relapsed/refractory disease.

NHL-001 documented the single-agent efficacy of lenalidomide for relapsed/refractory indolent NHL (Table 1)32 ; ORR was modest, but responding patients had durable remissions. Tuscano et al reported results with lenalidomide and rituximab in relapsed/refractory indolent NHL half of whom were rituximab-refractory (Table 1).33  In addition, UPenn published results of a carefully planned and well-done phase 2 study in rituximab-refractory FL and MCL showing that lenalidomide had activity as a single-agent and that lenalidomide/rituximab combination therapy may overcome rituximab resistance.34  There is an ongoing phase 3 trial (the “AUGMENT” study) evaluating lenalidomide combined with rituximab vs single-agent lenalidomide in relapsed/refractory indolent NHL (supplemental Table 2).

Untreated patients.

Fowler et al evaluated lenalidomide and rituximab in 110 untreated FL patients.31  Responses were high, and at completion of therapy, most patients demonstrated molecular response. There are several ongoing randomized studies examining lenalidomide as part of frontline therapy for FL patients. This includes a randomized phase 2 study examining lenalidomide therapy after immunochemotherapy induction (the “BIONIC” trial) and a phase 3 study comparing the efficacy of rituximab plus lenalidomide vs rituximab plus chemotherapy in untreated FL (the “RELEVANCE” trial) (supplemental Figure 1).

DLBCL

Relapsed/refractory.

Lenalidomide has activity in relapsed/refractory DLBCL (Table 1). In NHL-002 and NHL-003, the ORRs in DLBCL were 19% and 28%, respectively.27,39  The differential efficacy of lenalidomide in DLBCL based on cell of origin was evaluated by Hernandez et al40 ; lenalidomide appeared to be more effective in non-GC DLBCL with an ORR of 53% vs 9% for the GC subtype.

Zinzani et al reported data on 23 elderly DLBCL patients treated with rituximab and lenalidomide (with lenalidomide maintenance),41  and Wang et al confirmed the clinical activity of lenalidomide/rituximab in relapsed/refractory de novo and transformed DLBCL (Table 1).42  Feldman et al studied the addition of lenalidomide to standard salvage chemotherapy for relapsed/refractory DLBCL before autologous stem cell transplantation followed by lenalidomide maintenance.38  Ongoing studies are assessing lenalidomide combined with other chemotherapy backbones (supplemental Table 2).

Untreated.

The addition of lenalidomide to rituximab, cyclophosphamide, doxorubicin, oncovin, and prednisone (R2-CHOP) was evaluated in older, untreated DLBCL patients (Table 1).36  Efficacy appeared to be high, although ∼70% of patients had grade 3-4 hematologic AEs. Nowakowski et al added modified lenalidomide to R-CHOP, which was well tolerated and it appeared to overcome the negative prognostic impact of the non-GC phenotype in DLCBL.37  A large, randomized phase 2 trial randomizing untreated DLBCL patients to R2-CHOP vs R-CHOP (supplemental Figure 1) and a similarly designed industry-led study (only ABC-type DLBCL; the ROBUST study) are underway.

The role of lenalidomide in the postchemotherapy setting is also being investigated as maintenance therapy alone or in combination with rituximab. Early results from a randomized phase 2 trial of lenalidomide ± rituximab in intermediate-high/high-risk DLBCL have been presented (Table 1).35 

T-cell lymphoma

Relapsed/refractory.

The EXPECT phase 2 trial studied patients with relapsed/refractory peripheral T-cell lymphoma (PTCL) treated with single-agent lenalidomide (Table 1).44  The majority of patients (85%) had angioimmunoblastic (AITL) and PTCL–not otherwise specified histologies. Efficacy was documented, although serious AEs were seen in 54% of patients, with 6 deaths unrelated to progression. In additional single-agent studies, Tournishey et al reported results in untreated and relapsed/refractory PTCL,46  whereas Querfeld et al showed clinical activity in relapsed/refractory mycosis fungoides/Sézary syndrome (Table 1).45  In the latter study, tumor flare was seen in many patients, which prompted an amendment for lower initial lenalidomide dosing.

Untreated.

There are limited data examining lenalidomide in the front-line setting for PTCL. A trial investigating the efficacy of lenalidomide combined with CHOP in AITL is ongoing, and another study is evaluating romidepsin and lenalidomide in untreated PTCL (supplemental Table 2).

Biomarkers

Clinical studies have examined potential biomarkers of lenalidomide efficacy,28,29,34  and pivotal studies are ongoing in distinct NHL subsets (eg, non-GC DLBCL). In relapsed/refractory MCL, Zaja et al showed that microvessel density, macrophage, and NK cell counts were altered with lenalidomide,28  and Ki67 is being studied as a biomarker.48  Despite breakthrough studies that identified CRBN as a critical lenalidomide target,9-12,49  there are challenges in its use as a biomarker (eg, messenger RNA/protein correlation). The CRBN-associated transcription factors, Ikaros/Aiolos, may serve as more functional biomarkers16  as well as downstream substrates (eg, IRF4 and MYC). Other potential lenalidomide-related biomarkers include components of PI3K signaling (eg, GSK3β)50  and the T-cell immune synapse with granzyme B expression.51,52 

Novel combinations

In DLBCL cells, azacytidine and lenalidomide increased CRBN expression and enhanced cytotoxicity,53  whereas ibrutinib and lenalidomide synergistically suppressed IRF4.13  In vitro FL and MCL studies confirmed synergistic activity with bortezomib and lenalidomide.54  In T-cell lymphoma models, romidepsin combined with lenalidomide resulted in increased oxidative stress and alteration of PI3K and MAP kinase/ERK-signaling pathways.55  Conversely, preclinical studies in CLL suggest that the PI3Kδ inhibitor, idelalisib, antagonizes the immune-modulating properties of lenalidomide.

It will be important to translate these findings in clinical studies to confirm efficacy as well as safety. The latter was highlighted in two phase 1 studies combining lenalidomide with the PI3Kδ inhibitor, idelalisbub (and rituximab), where prohibitive toxicities suggestive of severe cytokine-release syndrome (eg, rash, fevers, hypotension) were seen.56  Other clinical studies combining lenalidomide with novel/targeted agents have shown good tolerability and encouraging efficacy,57  whereas additional novel combinatorial trials are underway (eg, obinutuzumab, romidepsin, everolimus, ibruitinib) (supplemental Table 2).

In addition, translational studies of new cereblon-binding agents (eg, CC-122) are eagerly anticipated.58  CC-122 is a first-in-class, pleiotropic pathway modifier that binds cereblon and induces Aiolos and Ikaros degradation in DLBCL and T cells.59  In DLBCL, it results in depression of interferon-stimulated genes/proteins, ultimately resulting in apoptosis.60  Furthermore, CC-122 appears to have distinct activity from lenalidomide in part as it is active in both GC and non-GC DLBCL.58,60 

Owing to its unique immunomodulatory and antiproliferative activity, as well as its relative ease of use, lenalidomide has garnered clinical consideration in multiple NHL subtypes. Durable responses with manageable side effects have moved lenalidomide into the front-line setting of randomized clinical trials for various NHL subtypes. In addition, combinatorial therapy with novel/targeted therapeutic agents is of particular interest. Finally, continued understanding of the biological mechanisms and the associated validation of predictive biomarkers will be critical in optimally integrating lenalidomide into NHL treatment paradigms.

The online version of this article contains a data supplement.

Contribution: A.K. and A.M.E. designed research, performed research, analyzed data, and wrote the paper; M.C. designed research, performed research, and wrote the paper; and J.S. performed research and wrote the paper.

Conflict-of-interest disclosure: A.M.E. is on the advisory board and speakers bureau (with honorarium) for Celgene. The remaining authors declare no competing financial interests.

Correspondence: Andrew M. Evens, Tufts Medical Center, Division of Hematology/Oncology, 800 Washington St, Boston, MA 02111; e-mail: aevens@tuftsmedicalcenter.org.

1
Ramsay
 
AG
Clear
 
AJ
Kelly
 
G
, et al. 
Follicular lymphoma cells induce T-cell immunologic synapse dysfunction that can be repaired with lenalidomide: implications for the tumor microenvironment and immunotherapy.
Blood
2009
, vol. 
114
 
21
(pg. 
4713
-
4720
)
2
Ramsay
 
AG
Johnson
 
AJ
Lee
 
AM
, et al. 
Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug.
J Clin Invest
2008
, vol. 
118
 
7
(pg. 
2427
-
2437
)
3
Kater
 
AP
Tonino
 
SH
Egle
 
A
Ramsay
 
AG
How does lenalidomide target the chronic lymphocytic leukemia microenvironment?
Blood
2014
, vol. 
124
 
14
(pg. 
2184
-
2189
)
4
Hernandez-Ilizaliturri
 
FJ
Reddy
 
N
Holkova
 
B
Ottman
 
E
Czuczman
 
MS
Immunomodulatory drug CC-5013 or CC-4047 and rituximab enhance antitumor activity in a severe combined immunodeficient mouse lymphoma model.
Clin Cancer Res
2005
, vol. 
11
 
16
(pg. 
5984
-
5992
)
5
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
, vol. 
140
 
1
(pg. 
36
-
45
)
6
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
, vol. 
14
 
14
(pg. 
4650
-
4657
)
7
Zhang
 
L
Qian
 
Z
Cai
 
Z
, et al. 
Synergistic antitumor effects of lenalidomide and rituximab on mantle cell lymphoma in vitro and in vivo.
Am J Hematol
2009
, vol. 
84
 
9
(pg. 
553
-
559
)
8
Zhu
 
D
Corral
 
LG
Fleming
 
YW
Stein
 
B
Immunomodulatory drugs Revlimid (lenalidomide) and CC-4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation.
Cancer Immunol Immunother
2008
, vol. 
57
 
12
(pg. 
1849
-
1859
)
9
Ito
 
T
Ando
 
H
Suzuki
 
T
, et al. 
Identification of a primary target of thalidomide teratogenicity.
Science
2010
, vol. 
327
 
5971
(pg. 
1345
-
1350
)
10
Zhu
 
YX
Braggio
 
E
Shi
 
C-X
, et al. 
Cereblon expression is required for the antimyeloma activity of lenalidomide and pomalidomide.
Blood
2011
, vol. 
118
 
18
(pg. 
4771
-
4779
)
11
Shaffer
 
AL
Emre
 
NC
Lamy
 
L
, et al. 
IRF4 addiction in multiple myeloma.
Nature
2008
, vol. 
454
 
7201
(pg. 
226
-
231
)
12
Lopez-Girona
 
A
Mendy
 
D
Ito
 
T
, et al. 
Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2326
-
2335
)
13
Yang
 
Y
Shaffer
 
AL
Emre
 
NC
, et al. 
Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma.
Cancer Cell
2012
, vol. 
21
 
6
(pg. 
723
-
737
)
14
Zhang
 
LH
Kosek
 
J
Wang
 
M
Heise
 
C
Schafer
 
PH
Chopra
 
R
Lenalidomide efficacy in activated B-cell-like subtype diffuse large B-cell lymphoma is dependent upon IRF4 and cereblon expression.
Br J Haematol
2013
, vol. 
160
 
4
(pg. 
487
-
502
)
15
Bea
 
S
Zettl
 
A
Wright
 
G
, et al. 
Lymphoma/Leukemia Molecular Profiling Project
Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve gene-expression-based survival prediction.
Blood
2005
, vol. 
106
 
9
(pg. 
3183
-
3190
)
16
Gandhi
 
AK
Kang
 
J
Havens
 
CG
, et al. 
Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.).
Br J Haematol
2014
, vol. 
164
 
6
(pg. 
811
-
821
)
17
Krönke
 
J
Udeshi
 
ND
Narla
 
A
, et al. 
Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells.
Science
2014
, vol. 
343
 
6168
(pg. 
301
-
305
)
18
Qian
 
Z
Zhang
 
L
Cai
 
Z
, et al. 
Lenalidomide synergizes with dexamethasone to induce growth arrest and apoptosis of mantle cell lymphoma cells in vitro and in vivo.
Leuk Res
2011
, vol. 
35
 
3
(pg. 
380
-
386
)
19
Fecteau
 
JF
Corral
 
LG
Ghia
 
EM
, et al. 
Lenalidomide inhibits the proliferation of CLL cells via a cereblon/p21(WAF1/Cip1)-dependent mechanism independent of functional p53.
Blood
2014
, vol. 
124
 
10
(pg. 
1637
-
1644
)
20
Song
 
K
Herzog
 
BH
Sheng
 
M
, et al. 
Lenalidomide inhibits lymphangiogenesis in preclinical models of mantle cell lymphoma.
Cancer Res
2013
, vol. 
73
 
24
(pg. 
7254
-
7264
)
21
Pellagatti
 
A
Jädersten
 
M
Forsblom
 
AM
, et al. 
Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients.
Proc Natl Acad Sci USA
2007
, vol. 
104
 
27
(pg. 
11406
-
11411
)
22
Zhang
 
L-H
Schafer
 
PH
Muller
 
G
Stirling
 
D
Bartlett
 
B
Direct inhibitory effects of lenalidomide on the proliferation and VEGF production of non-Hodgkin lymphoma cells are associated with increased SPARC expression.
Blood
2008
(pg. 
905
-
905
)
23
Jerkeman
 
M A-LA
Kolstad
 
A
Lenalidomide, bendamustine, and rituximab as first-line therapy for patients> 65 years with mantle cell lymphoma: results from the phase I portion of the Nordic lymphoma group MCL4 (LENA-BERIT) trial [abstract].
Blood
2013
, vol. 
122
 
21
pg. 
Abstract 4377
 
24
Ruan
 
J
Martin
 
P
Shah
 
BD
, et al. 
Sustained remission with the combination biologic doublet of lenalidomide plus rituximab as initial treatment for mantle cell lymphoma: a multi-center phase II study report [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 625
 
25
Wang
 
M
Fayad
 
L
Wagner-Bartak
 
N
, et al. 
Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial.
Lancet Oncol
2012
, vol. 
13
 
7
(pg. 
716
-
723
)
26
Habermann
 
TM
Lossos
 
IS
Justice
 
G
, et al. 
Lenalidomide oral monotherapy produces a high response rate in patients with relapsed or refractory mantle cell lymphoma.
Br J Haematol
2009
, vol. 
145
 
3
(pg. 
344
-
349
)
27
Witzig
 
TE
Vose
 
JM
Zinzani
 
PL
, et al. 
An international phase II trial of single-agent lenalidomide for relapsed or refractory aggressive B-cell non-Hodgkin’s lymphoma.
Ann Oncol
2011
, vol. 
22
 
7
(pg. 
1622
-
1627
)
28
Zaja
 
F
De Luca
 
S
Vitolo
 
U
, et al. 
Salvage treatment with lenalidomide and dexamethasone in relapsed/refractory mantle cell lymphoma: clinical results and effects on microenvironment and neo-angiogenic biomarkers.
Haematologica
2012
, vol. 
97
 
3
(pg. 
416
-
422
)
29
Eve
 
HE
Carey
 
S
Richardson
 
SJ
, et al. 
Single-agent lenalidomide in relapsed/refractory mantle cell lymphoma: results from a UK phase II study suggest activity and possible gender differences.
Br J Haematol
2012
, vol. 
159
 
2
(pg. 
154
-
163
)
30
Goy
 
A
Sinha
 
R
Williams
 
ME
, et al. 
Single-agent lenalidomide in patients with mantle-cell lymphoma who relapsed or progressed after or were refractory to bortezomib: phase II MCL-001 (EMERGE) study.
J Clin Oncol
2013
, vol. 
31
 
29
(pg. 
3688
-
3695
)
31
Fowler
 
NH
Davis
 
RE
Rawal
 
S
, et al. 
Safety and activity of lenalidomide and rituximab in untreated indolent lymphoma: an open-label, phase 2 trial.
Lancet Oncol
2014
, vol. 
15
 
12
(pg. 
1311
-
1318
)
32
Witzig
 
TE
Wiernik
 
PH
Moore
 
T
, et al. 
Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin’s Lymphoma.
J Clin Oncol
2009
, vol. 
27
 
32
(pg. 
5404
-
5409
)
33
Tuscano
 
JM
Dutia
 
M
Chee
 
K
, et al. 
Lenalidomide plus rituximab can produce durable clinical responses in patients with relapsed or refractory, indolent non-Hodgkin lymphoma.
Br J Haematol
2014
, vol. 
165
 
3
(pg. 
375
-
381
)
34
Chong
 
EA
Ahmadi
 
T
Aqui
 
N
, et al. 
Combination of lenalidomide and rituximab overcomes rituximab-resistance in patients with indolent B-cell and mantle cell lymphomas.
Clin Cancer Res
2015
35
Reddy
 
NM
Morgan
 
DS
Park
 
SI
, et al. 
Phase II Randomized Study Of Lenalidomide Or Lenalidomide and Rituximab As Maintenance Therapy Following Standard Chemotherapy For Patients With Intermediate-High/High Risk Diffuse Large B-Cell Lymphoma (DLBCL) [abstract].
Blood
2013
, vol. 
122
 
21
pg. 
Abstract 3061
 
36
Vitolo
 
U
Chiappella
 
A
Franceschetti
 
S
, et al. 
Fondazione Italiana Linfomi
Lenalidomide plus R-CHOP21 in elderly patients with untreated diffuse large B-cell lymphoma: results of the REAL07 open-label, multicentre, phase 2 trial.
Lancet Oncol
2014
, vol. 
15
 
7
(pg. 
730
-
737
)
37
Nowakowski
 
GS
LaPlant
 
B
Macon
 
WR
, et al. 
Lenalidomide combined with R-CHOP overcomes negative prognostic impact of non-germinal center B-cell phenotype in newly diagnosed diffuse large B-Cell lymphoma: a phase II study.
J Clin Oncol
2015
, vol. 
33
 
3
(pg. 
251
-
257
)
38
Feldman
 
T
Mato
 
AR
Chow
 
KF
, et al. 
Addition of lenalidomide to rituximab, ifosfamide, carboplatin, etoposide (RICER) in first-relapse/primary refractory diffuse large B-cell lymphoma.
Br J Haematol
2014
, vol. 
166
 
1
(pg. 
77
-
83
)
39
Wiernik
 
PH
Lossos
 
IS
Tuscano
 
JM
, et al. 
Lenalidomide monotherapy in relapsed or refractory aggressive non-Hodgkin’s lymphoma.
J Clin Oncol
2008
, vol. 
26
 
30
(pg. 
4952
-
4957
)
40
Hernandez-Ilizaliturri
 
FJ
Deeb
 
G
Zinzani
 
PL
, et al. 
Higher response to lenalidomide in relapsed/refractory diffuse large B-cell lymphoma in nongerminal center B-cell-like than in germinal center B-cell-like phenotype.
Cancer
2011
, vol. 
117
 
22
(pg. 
5058
-
5066
)
41
Zinzani
 
PL
Pellegrini
 
C
Derenzini
 
E
Argnani
 
L
Pileri
 
S
Long-term efficacy of the combination of lenalidomide and rituximab in elderly relapsed/refractory diffuse large B-cell lymphoma patients.
Hematol Oncol
2013
, vol. 
31
 
4
(pg. 
223
-
224
)
42
Wang
 
M
Fowler
 
N
Wagner-Bartak
 
N
, et al. 
Oral lenalidomide with rituximab in relapsed or refractory diffuse large cell, follicular and transformed lymphoma: a phase II clinical trial.
Leukemia
2013
, vol. 
27
 
9
(pg. 
1902
-
1909
)
43
Zinzani
 
PL
Pellegrini
 
C
Broccoli
 
A
, et al. 
Lenalidomide monotherapy for relapsed/refractory peripheral T-cell lymphoma not otherwise specified.
Leuk Lymphoma
2011
, vol. 
52
 
8
(pg. 
1585
-
1588
)
44
Morschhauser
 
F
Fitoussi
 
O
Haioun
 
C
, et al. 
A phase 2, multicentre, single-arm, open-label study to evaluate the safety and efficacy of single-agent lenalidomide (Revlimid) in subjects with relapsed or refractory peripheral T-cell non-Hodgkin lymphoma: the EXPECT trial.
Eur J Cancer
2013
, vol. 
49
 
13
(pg. 
2869
-
2876
)
45
Querfeld
 
C
Rosen
 
ST
Guitart
 
J
, et al. 
Results of an open-label multicenter phase 2 trial of lenalidomide monotherapy in refractory mycosis fungoides and Sézary syndrome.
Blood
2014
, vol. 
123
 
8
(pg. 
1159
-
1166
)
46
Toumishey
 
E
Prasad
 
A
Dueck
 
G
, et al. 
Final report of a phase 2 clinical trial of lenalidomide monotherapy for patients with T-cell lymphoma.
Cancer
2015
, vol. 
121
 
5
(pg. 
716
-
723
)
47
Zinzani
 
PL
Vose
 
JM
Czuczman
 
MS
, et al. 
Long-term follow-up of lenalidomide in relapsed/refractory mantle cell lymphoma: subset analysis of the NHL-003 study.
Ann Oncol
2013
, vol. 
24
 
11
(pg. 
2892
-
2897
)
48
Goy
 
A
Williams
 
ME
Besisik
 
SK
Updated efficacy and safety, and exploratory Ki- 67 results for the MCL-001 study of lenalidomide in mantle cell lymphoma patients who relapsed or were refractory to bortezomib [abstract].
Blood
2013
, vol. 
122
 
21
pg. 
Abstract 3057
 
49
Escoubet-Lozach
 
L
Lin
 
I-L
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
, vol. 
69
 
18
(pg. 
7347
-
7356
)
50
Herman
 
SE
Lapalombella
 
R
Gordon
 
AL
, et al. 
The role of phosphatidylinositol 3-kinase-δ in the immunomodulatory effects of lenalidomide in chronic lymphocytic leukemia.
Blood
2011
, vol. 
117
 
16
(pg. 
4323
-
4327
)
51
Ramsay
 
AG
Clear
 
AJ
Fatah
 
R
Gribben
 
JG
Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer.
Blood
2012
, vol. 
120
 
7
(pg. 
1412
-
1421
)
52
Shanafelt
 
TD
Ramsay
 
AG
Zent
 
CS
, et al. 
Long-term repair of T-cell synapse activity in a phase II trial of chemoimmunotherapy followed by lenalidomide consolidation in previously untreated chronic lymphocytic leukemia (CLL).
Blood
2013
, vol. 
121
 
20
(pg. 
4137
-
4141
)
53
Locatelli
 
SLPR
Careddu
 
G
, et al. 
Upregulation of Cereblon Expression by the DNA methyltransferase inhibitor azacytidine strongly enhances lenalidomide cytoxicity in Germinal center B-cell-like (GCB) and Activated B-cell-like (ABC) Diffuse Large B-cell Lymphoma (DLBCL) [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 2253
 
54
Cosenza
 
M
Civallero
 
M
Pozzi
 
S
Marcheselli
 
L
Bari
 
A
Sacchi
 
S
The combination of bortezomib with enzastaurin or lenalidomide enhances cytotoxicity in follicular and mantle cell lymphoma cell lines [published online ahead of print November 13, 2014].
Hematol Oncol
 
doi:10.1002/hon.2179
55
Cosenza
 
MCM
Fiorcari
 
S
, et al. 
Romidepsin synergizes with lenalidomide in T cell lymphoma cell lines by increasing reactive oxygen species and modulating PI3K/AKT and MAPK/ERK signaling [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 1778
 
56
Smith
 
SMPB
MS2*, Jung SH, et al. Unexpected and Serious Toxicity Observed with Combined Idelalisib, Lenalidomide and Rituximab in Relapsed/Refractory B Cell Lymphomas: Alliance A051201 and A051202 [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 3091
 
57
Smith
 
SMCK
Kline
 
JP
, et al. 
Phase I trial of temsirolimus and lenalidomide in pts with rel/ref lymphomas [abstract].
J Clin Oncol
2012
, vol. 
30
 
suppl
pg. 
Abstract 8075
 
58
Ribrag
 
V
Damien
 
D
Gharibo
 
M
CC-122 degrades the lymphoid transcription factor Aiolos (IKZF3) by modulating cereblon and shows clinical activity in a phase Ib study of subjects with relapsed or refractory non-Hodgkin’s lymphoma and multiple myeloma [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 3500
 
59
Blocksidge
 
J
Glenn
 
M
Gandhi
 
AK
CC-122 has robust anti-proliferative activity in a primary chronic lymphocytic leukemia (CLL) co-culture model and is superior to lenalidomide.
Blood
2014
, vol. 
124
 pg. 
Abstract 4682
 
60
Hagner
 
P
Wang
 
M
Couto
 
S
, et al. 
CC-122 has potent anti-lymphoma activity through destruction of the Aiolos and Ikaros transcription factors and induction of interferon response pathways [abstract].
Blood
2014
, vol. 
124
 
21
pg. 
Abstract 3035
 

Author notes

A.K. and M.C. contributed equally to this paper.

Sign in via your Institution