Immunotherapy has emerged as a viable clinical strategy to harness endogenous antitumor T-cell immunity. Lenalidomide is an oral immunomodulatory drug that repairs antitumor T-cell function and is showing efficacy in ongoing chronic lymphocytic leukemia (CLL) and lymphoma clinical trials. This article focuses on advances in our understanding of its mechanism of action in the tumor microenvironment and provides a clinical update in CLL. Challenges associated with this drug and its potential use in the targeted drug treatment era are discussed.

Chronic lymphocytic leukemia (CLL) is one of the most common B-cell malignancies in adults, characterized by an accumulation of monoclonal CD5+ mature B cells in lymphoid tissues and the peripheral blood. Clonal expansion and invasive migration typically causes the lymph nodes, spleen, and the bone marrow to become infiltrated with tumor. Current standard therapy combines chemotherapy with an anti-CD20 monoclonal antibody (mAb) (chemoimmunotherapy [CIT]). Although highly potent, CIT induces substantial toxicity and is not curative, with nearly all patients eventually relapsing. Recent advances using kinase inhibitors (eg, ibrutinib and idelalisib) that target B-cell receptor (BCR) signaling indicate an exciting shift toward a nonchemotherapy treatment era (reviewed by Jones and Byrd1 ). Present indications suggest these drugs are not producing many complete responses and should be taken continuously to avoid relapse. Unanswered questions include whether long-term persistent disease and prolonged therapy promote drug-resistant variants through clonal evolution and/or activation of compensatory oncogenic signaling in CLL. Ibrutinib resistance has already been detected in genetically high-risk patients,2  highlighting the necessity to identify combinatorial therapy using agents with distinct mechanism of action (MOA).

In addition to genome alterations, CLL exhibits another dimension of complexity: leukemic cells are nurtured and protected from anticancer therapies by a variety of resident and recruited ostensibly normal cells that constitute the tumor microenvironment (TME) in lymphoid organs. Nonmalignant components of the TME include: mesenchymal stromal/stem cells (MSCs), endothelial cells, tumor-associated macrophages (TAMs or “nurse-like cells” [NLCs]), dendritic cells (DCs) and T cells. A recent breakthrough in cancer therapeutics has been the use of immunotherapies (immune checkpoint blockade) that target mechanisms of T-cell evasion by tumors.3  In this review, we focus on the immunomodulatory drug (IMiD) lenalidomide which activates antitumor T-cell activity and is showing clinical activity in ongoing CLL clinical trials. Lenalidomide (Revlimid) is a derivative of thalidomide that is US Food and Drug Administration (FDA) approved for the treatment of multiple myeloma, myelodysplastic syndromes, and mantle cell lymphoma. Remarkably, in contrast to the BCR inhibitor drugs, IMiDs exemplify successful bedside-to-bench research, in that their clinical effectiveness was known before recent MOA data emerged that help explain their pleotropic effects in the TME.

Active crosstalk between leukemic cells and nonmalignant cells in the TME plays a critical role in activating tumor migration, survival, proliferation, and fostering immune privilege (reviewed by Burger and Gribben4 ).

Briefly, CLL cells migrate into their niches via prosurvival CXC chemokine ligand 12 (CXCL12)- and CXCL13-chemokine gradients released by MSCs, NLCs, follicular helper CD4+ T cells (TFH), and follicular DCs (FDCs). In the TME, MSCs and endothelial cells induce signal transducer and activator of transcription 3 (STAT3) and nuclear factor–κB (NF-κB)–mediated prosurvival signaling in CLL cells. Monocyte-derived NLCs supply pro-CLL survival signals and resemble proangiogenic and immunosuppressive TAMs.4  Notably, CLL-stroma crosstalk is bidirectional and stromal cells in turn also become activated by leukemic cells. CLL-secreted soluble factors also activate receptors on MSCs leading to AKT activation, proliferation, and secretion of proangiogenic vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF).5 

BCR activation in the TME has emerged as a central oncogenic pathway6  essential for CLL survival and proliferation. BCR engagement likely involves extrinsic autoantigens and/or microbial antigens, as well as autoreactive autonomous activation.6  Signals from the BCR and the tissue TME (cellular and molecular interactions) converge on several key intracellular signaling pathways including the phosphatidylinositol 3-kinase (PI3K)–AKT axis.4  FDCs and TFH cells are specialized reservoirs of intact antigen and cognate B-cell help, respectively, and likely contribute to BCR activation.

Intriguingly, subverted CD4+ T cells support CLL cells in the TME, whereas antileukemic CD8+ T cells are suppressed. Activated CD4+ T cells and TFH cells can provide CD40L costimulation and prosurvival T helper 2 (Th2) cytokines (interleukin-4 [IL-4], IL-6, IL-21) that trigger extracellular signal-regulated kinase (ERK), STAT-3/-6, and NF-κB signaling and leukemic proliferation. However, CLL cells also subvert T cells to avoid immune destruction by inducing defective immune synapsis.7  Immune synapses (formed at T-cell–APC interfaces during antigen recognition) have a master role in T-cell activation and polarized delivery of effector molecules. CLL cells express low levels of costimulatory molecules while coopting multiple immune checkpoint coinhibitory pathways including PD-L1 that deliver an inhibitory signal into PD1+ T cells to actively suppress synapses and secretion of IL-2 and lytic granules.8  Additional immune evasion mechanisms include leukemic-derived factors that suppress NK-cell lytic activity and induction of regulatory T cells (Tregs).4 

Seminal breakthrough studies have recently identified that IMiDs (thalidomide, lenalidomide, and pomalidomide) bind to the protein target cereblon (CRBN), altering an E3 ligase complex that modulates expression of critical transcription factors. In the absence of IMiDs, CRBN-directed E3 ubiquitin ligase activity is directed to CRBN itself and possibly other specific proteins that are tagged for proteasome-mediated degradation. IMiDs bind to CRBN and modulate its substrate recognition, preventing autoubiquitylation. Three independent studies have simultaneously revealed that IMiDs retarget CRBN-dependent ligase activity toward the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) and induce their proteasomal degradation.9-11  Their decreased abundance leads to loss of viability in myeloma B cells. Ikaros and Aiolos are highly expressed in CLL cells and there is evidence supporting their role in regulating survival signaling.12,13  Testing whether the ability of lenalidomide to downregulate these transcription factors in lymphocytes contributes to both direct and indirect anti-CLL activity will be of great interest. Lenalidomide is not directly cytotoxic to CLL cells in vitro, but does alter CLL-TME protumoral signals. Direct anti-CLL effects include inhibition of TME-induced proliferation in a CRBN/p21-dependent manner that is associated with reduced expression of IKZF1 and IKZF3 in common with myeloma (Figure 1i).14 

Figure 1

How lenalidomide targets the CLL TME. In contrast to conventional targeted drugs, lenalidomide seems to exert most of its anti-CLL activity by interfering with protumoral TME interactions rather than directly targeting prosurvival signaling in the tumor clone itself. Recent MOA data have shown that the IMiD lenalidomide binds to the protein CRBN, which modulates its substrate recognition and augments the ubiquitylation and degradation of critical transcription factors in lymphocytes. Ikaros (IKZF1) and Aiolos (IKZF3) are the downregulated targets in MM that cause direct malignant B-cell toxicity. Whether these transcription factors are targeted by lenalidomide in the diverse cellular components of the CLL TME remains to be investigated. This IMiD retargeted CRBN-dependent ubiquitin ligase activity simultaneously alters malignant B-cell and nonmalignant T-cell function. (i) Emerging data have shown that a retargeted CRBN complex directly inhibits TME-mediated proliferation signaling in CLL B cells that is associated with reduced expression of IKZF1 and IKZF3 in common with myeloma. (ii) Importantly, degradation of Ikaros and Aiolos in T cells stimulates IL-2 secretion that enhances immune function. Lenalidomide has been shown to effectively reverse tumor-induced immune suppression/privilege. Immunomodulatory MOA includes: promoting a Th2 to Th1 CD4+ T-cell switch, downregulating immunosuppressive signaling axes (eg, PD:L1:PD1) while enhancing costimulatory molecules that positively regulate antitumor CD8+ T-cell function and the induction of humoral immunity (residual normal B cells). CRBN-dependent activation of RHO GTPase activation signaling and cytoskeletal signaling repairs T-cell lytic immune synapse and motility function. (iii) Lenalidomide also modulates tumor-educated stromal cells (TAMs/NLCs and MSCs) in the TME that block essential survival signals for the expanding malignant B-cell clone. MM, multiple myeloma.

Figure 1

How lenalidomide targets the CLL TME. In contrast to conventional targeted drugs, lenalidomide seems to exert most of its anti-CLL activity by interfering with protumoral TME interactions rather than directly targeting prosurvival signaling in the tumor clone itself. Recent MOA data have shown that the IMiD lenalidomide binds to the protein CRBN, which modulates its substrate recognition and augments the ubiquitylation and degradation of critical transcription factors in lymphocytes. Ikaros (IKZF1) and Aiolos (IKZF3) are the downregulated targets in MM that cause direct malignant B-cell toxicity. Whether these transcription factors are targeted by lenalidomide in the diverse cellular components of the CLL TME remains to be investigated. This IMiD retargeted CRBN-dependent ubiquitin ligase activity simultaneously alters malignant B-cell and nonmalignant T-cell function. (i) Emerging data have shown that a retargeted CRBN complex directly inhibits TME-mediated proliferation signaling in CLL B cells that is associated with reduced expression of IKZF1 and IKZF3 in common with myeloma. (ii) Importantly, degradation of Ikaros and Aiolos in T cells stimulates IL-2 secretion that enhances immune function. Lenalidomide has been shown to effectively reverse tumor-induced immune suppression/privilege. Immunomodulatory MOA includes: promoting a Th2 to Th1 CD4+ T-cell switch, downregulating immunosuppressive signaling axes (eg, PD:L1:PD1) while enhancing costimulatory molecules that positively regulate antitumor CD8+ T-cell function and the induction of humoral immunity (residual normal B cells). CRBN-dependent activation of RHO GTPase activation signaling and cytoskeletal signaling repairs T-cell lytic immune synapse and motility function. (iii) Lenalidomide also modulates tumor-educated stromal cells (TAMs/NLCs and MSCs) in the TME that block essential survival signals for the expanding malignant B-cell clone. MM, multiple myeloma.

Close modal

Importantly, Ikaros and Aiolos are repressors of the IL-2 promoter, and their degradation in response to IMiDs explains enhanced costimulation and IL-2 secretion in T cells.11  Thus, the ability of IMiDs to target CRBN and the ubiquitin proteasome system modulates the expression of critical transcription factors that costimulate T cells, while degrading B-cell function simultaneously (Figure 1). It should be noted that the CRBN complex is likely to have distinct sets of targets in different healthy and cancer cell types. Moreover, proteins that are degraded (polyubiquitylated), compartmentalized (monoubiquitylated), or sequestered by the CRBN complex, or differentially released and stabilized by IMiDs, are likely to contribute to their biological activity.15 

IMiDs increase the DC cross-priming,16  expansion, and activation of CD8+ T cells while decreasing activated CD4+ T-cell–derived cytokines,17  promoting Th1 T-cell differentiation, and polarizing Th2 T cells to a Th1 phenotype (interferon-γ [IFN-γ], tumor necrosis factor-α [TNF-α]).1  This Th2 to Th1 switch is linked to increased expression of the transcription factor T-bet.1 

Lenalidomide repairs the tumor-induced T-cell immune synapse defect in CLL and lymphoma by increasing the assembly and activity of cytoskeletal signaling molecules including protein kinase C-θ (PKC-θ), WASp, and RHO GTPase CDC42 to the synapse, allowing effective T-cell receptor (TCR)/CD28 signal transduction and directional secretion of lytic granules.7,8  IMiD-mediated repair of NK-cell lytic synapse formation is also emerging.1,4  Lenalidomide also rescues T-cell motility in CLL by normalizing RAC1, RHOA, and CDC42 activity levels. Importantly, knockdown of CRBN blocks lenalidomide repair of T-cell function.18  Taken together, enhancement of T-cell–mediated responses by lenalidomide is linked to a retargeted CRBN complex and altered transcription and activation of critical RHO GTPases.8 

Lenalidomide also alters RHO GTPase activation signaling that degrades malignant B-cell function (migration capability) and TME interactions (blocks prosurvival signaling interactions with NLCs).19,20  Upregulation of costimulatory molecules such as CD80 and CD86 on CLL cells by lenalidomide contributes to the repair of T-cell synapses.7  Enhanced expression of CD40L on CLL cells by lenalidomide promotes immunoglobulin production by normal B cells21  that has been detected in long-term treatment responders. Thus, repair of the humoral defect may contribute to enhanced antitumor immunity. Lenalidomide downregulates the PD-L1:PD-1 immunosuppressive axis8  in CLL, lymphoma and myeloma, allowing T cells and NK cells to form lytic synapses with target tumor cells. Paradoxically, PD-1 positively regulates the suppressive activity of Tregs. Decreased PD-18  and transcription factor FOXP3 expression may explain how IMiDs reduce Tregs while activating CD8+ T cells17  (Figure 1ii).

CLL cells position themselves in close contact with endothelial cells in the TME.4  Lenalidomide inhibits bidirectional prosurvival crosstalk between endothelial cells and tumor cells including proangiogenic signals.22  Notably, plasma levels of proangiogenic VEGF and FGF were reduced in CLL patients who responded to therapy.22  IMiD inhibition of MSC-derived CXCL121  may interfere with the CXC chemokine receptor 4 (CXCR4)-CXCL12 migratory axis in the CLL-TME (Figure 1iii).

The initial dose of lenalidomide in CLL clinical trials was chosen based on experience in myeloma but this induced serious side effects including rapid tumor cell death (tumor lysis syndrome [TLS]) and acute inflammation (tumor flare reaction [TFR], that may be associated with immune-mediated clinical response). Therefore, a low initial dose with dose escalation has been applied in subsequent trials and has improved tolerability.

Published results discussed here are summarized and referenced in Table 1 (ongoing studies: supplemental Table 1, see supplemental Data available at the Blood Web site).23-41  When used as monotherapy, overall response rates (ORRs) have been up to 72% (mostly partial responses) for first-line therapy and 25% to 30% for fludarabine-refractory CLL. Lenalidomide has shown clinical activity in patients with high-risk features such as del(17p) (31%-38% ORR). Lenalidomide was shown to antagonize NK-cell killing of rituximab-treated CLL cells if both agents were used simultaneously.42  However, this has not translated to the clinic (likely due to drug scheduling) as results suggest improved efficacy and lower occurrence of TLS and TFR when combining rituximab or ofatumumab with lenalidomide.

Table 1

Published trials with lenalidomide in CLL

ReferenceStatus*PhaseNo. of patientsAge, median (range)No. of prior therapies, median (range)del17p, %SchemeLenalidomideResponse, %||PFS, moToxicity, %#
Dose, mg/d§SchemeORRCRPR, incl nPRGrade 3-4 neutropeniaTLSTFR
Upfront                 
 Badoux et al24 ** Completed 60 71 (66-85)  10 len mono 5→25 Continuous 65 15 50 NR 83 52 
 Chen et al26 †† Completed 25 60 (33-78)  32‡‡ len mono 2.5→25 21 of 28 d 56 56 NR 72 88 
 Chen et al27 †† Completed 25 60 (33-78)  32‡‡ len mono 2.5→25 21 of 28 d 72 12 60 NR 76 88 
 James et al28  Completed 40 56 (45-64)  10 len+R 2.5→10 21 of 28 d 95 20 75 19 53 UN 83 
   29 70 (65-80)     79 10 69 20 66 UN 66 
 Brown et al29  Terminated 59 (37-66)  None len+FR 2.5→25 21 of 28 d 56 11 45 UN Study halted because of grade 3-4 hematologic toxicity and TFS 
 Flinn et al30  Open 1/2 51 62 (44-82)  12a len+FR 2.5→5 21 of 28 d 43 11 41 NR 47 UN UN 
 Egle et al31 b Completed 1/2 45 66 (43-79)  UNc len+FR 2.5→25 21 of 28 d 87 49 38 NR 88 UN 
 Egle et al32 b Completed 1/2 40 67 (43-79)  UNc len+FR + len+R maint. 2.5→25 21 of 28 d 96 67 29 46 72 UN 
 Strati et al25  Open 25 68 (44-79)  12 len+R + len maint. 10 Continuous 85 10 75 NR 36 UN 29 
Relapsed or refractory                 
 Chanan-Khan et al33  Completed 45 64 (42-75) 3 (1-10) 13 len mono 25 21 of 28 d 47 38 NR 70 58 
 Ferrajoli et al34  Completed 44 64 (49-86) 5 (1-15) 18 len mono 10→25 Continuous 32 25 NR 41d 12e 
 Wendtner et al35  Completed 52 65 (37-80) 4 (1-14) 17 len mono 2.5→20 Continuous 11.5 11.5 65 44 
 Buhler et al36  Open 104 64 (32-81) >1 25e len mono 5→25 Continuous 44 UN UN 11 UN UN UN 
 Ferrajoli et al37  Open 36 62 (34-82) 2 (1-8) 26 len+O 10 Continuous 65 15 50 NR 44 UN 24 
 Costa et al38  Completed 17 65 (51-80) 2 (1-4) 25 len+O 10 21 of 28 d 43 UN UN NR 79 UN 57 
 Sylvan et al39  Completed 12 71 (65-79) 4 (1-6) 42 len+A 2.5 or 5 Continuous 42 42 75 33 
 Badoux et al40  Open 59 62 (42-82) 2 (1-9) 25 len+R 10 Continuous 66 12 54 17.4 73 1.7 27 
Maintenance                 
 Shanafelt et al41  Open 34 65 (44-78) After PCR 2.3 len maint. 5→10 Continuous Improved quality of response in 8 patients (24%) NR 49 N/A N/A 
ReferenceStatus*PhaseNo. of patientsAge, median (range)No. of prior therapies, median (range)del17p, %SchemeLenalidomideResponse, %||PFS, moToxicity, %#
Dose, mg/d§SchemeORRCRPR, incl nPRGrade 3-4 neutropeniaTLSTFR
Upfront                 
 Badoux et al24 ** Completed 60 71 (66-85)  10 len mono 5→25 Continuous 65 15 50 NR 83 52 
 Chen et al26 †† Completed 25 60 (33-78)  32‡‡ len mono 2.5→25 21 of 28 d 56 56 NR 72 88 
 Chen et al27 †† Completed 25 60 (33-78)  32‡‡ len mono 2.5→25 21 of 28 d 72 12 60 NR 76 88 
 James et al28  Completed 40 56 (45-64)  10 len+R 2.5→10 21 of 28 d 95 20 75 19 53 UN 83 
   29 70 (65-80)     79 10 69 20 66 UN 66 
 Brown et al29  Terminated 59 (37-66)  None len+FR 2.5→25 21 of 28 d 56 11 45 UN Study halted because of grade 3-4 hematologic toxicity and TFS 
 Flinn et al30  Open 1/2 51 62 (44-82)  12a len+FR 2.5→5 21 of 28 d 43 11 41 NR 47 UN UN 
 Egle et al31 b Completed 1/2 45 66 (43-79)  UNc len+FR 2.5→25 21 of 28 d 87 49 38 NR 88 UN 
 Egle et al32 b Completed 1/2 40 67 (43-79)  UNc len+FR + len+R maint. 2.5→25 21 of 28 d 96 67 29 46 72 UN 
 Strati et al25  Open 25 68 (44-79)  12 len+R + len maint. 10 Continuous 85 10 75 NR 36 UN 29 
Relapsed or refractory                 
 Chanan-Khan et al33  Completed 45 64 (42-75) 3 (1-10) 13 len mono 25 21 of 28 d 47 38 NR 70 58 
 Ferrajoli et al34  Completed 44 64 (49-86) 5 (1-15) 18 len mono 10→25 Continuous 32 25 NR 41d 12e 
 Wendtner et al35  Completed 52 65 (37-80) 4 (1-14) 17 len mono 2.5→20 Continuous 11.5 11.5 65 44 
 Buhler et al36  Open 104 64 (32-81) >1 25e len mono 5→25 Continuous 44 UN UN 11 UN UN UN 
 Ferrajoli et al37  Open 36 62 (34-82) 2 (1-8) 26 len+O 10 Continuous 65 15 50 NR 44 UN 24 
 Costa et al38  Completed 17 65 (51-80) 2 (1-4) 25 len+O 10 21 of 28 d 43 UN UN NR 79 UN 57 
 Sylvan et al39  Completed 12 71 (65-79) 4 (1-6) 42 len+A 2.5 or 5 Continuous 42 42 75 33 
 Badoux et al40  Open 59 62 (42-82) 2 (1-9) 25 len+R 10 Continuous 66 12 54 17.4 73 1.7 27 
Maintenance                 
 Shanafelt et al41  Open 34 65 (44-78) After PCR 2.3 len maint. 5→10 Continuous Improved quality of response in 8 patients (24%) NR 49 N/A N/A 
*

As registered in clinicaltrials.gov; June 2014.

UN, unknown/not reported.

A, alemtuzumab; C, cyclophosphamide; F, fludarabine; len, lenalidomide; maint, maintenance; mono, monotherapy; MPS, methylprednisolone; O, ofatumumab; P, pentostatin; R, rituximab.

§

In most studies, dose escalation of lenalidomide was applied; doses before and after the arrow represent the minimum and maximum dose of lenalidomide per protocol. Of note, the highest dose level was not reached in all studies.

||

CR, complete remission; nPR, nodular partial remission; ORR, overall response rate; PR, partial remission.

NR, median PFS not reached; PFS, progression-free survival; UN, unknown/not reported.

#

N/A, not applicable; TFR, tumor flare reaction; TLS, tumor lysis syndrome.

**

An update on long-term outcome of these patients was published by Strati et al.25 

††

Two reports presenting results of 1 trial at different time points (at median follow-up of 20.7 and 47 mo, respectively).

‡‡

Including patients with del11q.

a

Including patients with del11q.

b

Two reports presenting results of 1 trial at different time points.

c

At least 1 molecular high risk feature (by CD38 expression, fluorescence in situ hybridization analysis, mutation status analysis) was present in 64% of patients.

d

Toxicity in percentage of cycles (as opposed to percentage of patients).

e

A TP53 mutation was found in 36/96 (38%) of the patients in this study; of these 20 patients did not have del17p.

Experience with lenalidomide in the consolidation/maintenance setting has been limited. Results from a phase 2 trial of CIT followed by lenalidomide showed that 24% of patients improved their quality of response with consolidation and some converted to minimal residual disease–negative status.41  These results are encouraging for an active placebo-controlled phase 3 trial of lenalidomide maintenance in minimal residual disease–positive patients post-CIT.

The randomized ORIGIN trial that compared the safety and efficacy of front-line lenalidomide vs chlorambucil in elderly patients was halted by the FDA for safety concerns following an imbalance in the number of deaths in the lenalidomide treatment arm. This experience is in contrast to a trial reporting long-lasting efficacy with lenalidomide monotherapy25  and highlights the safety and toxicity challenges associated with trial management. Clinical experience suggests an individual component to how CLL patients tolerate lenalidomide, and identifying predictive pretreatment factors requires future research. Grade 3/4 neutropenia has been reported in 70% to 83% of treated CLL patients (Table 1). The cause of lenalidomide-induced neutropenia may be related to the downregulation of transcription factor PU.1,43  that can be alleviated with granulocyte colony-stimulating factor (G-CSF) treatment.

Lenalidomide-induced TFR can be effectively managed with anti-inflammatory drugs (eg, dexamethasone). Notably, the combination of lenalidomide with fludarabine and rituximab results in a dramatic reduction in TFR.

TLS has been reported in 0% to 4.5% of patients from phase 2 studies. Slow-dose escalation, close monitoring, and prophylaxis (eg, allopurinol) can effectively prevent TLS.

The immune synapse bioassay in combination with the lytic biomarker Granzyme-B8  has demonstrated utility as a knowledge-based T-cell–monitoring assay in the phase 2 trial of CIT followed by lenalidomide consolidation.41  CRBN’s central role as a target of IMiD immunomodulation supports its use as a biomarker. However, its multiple splice variants and potential lack of correlation between messenger RNA and protein highlights the challenges when assessing biomarkers. Furthermore, CRBN does not appear to be a good predictive marker in CLL as it exhibits uniform expression regardless of clinical response. In contrast, expression of GSK-3 has shown potential as a response biomarker.44,45  Future research on the modulation of CRBN-Ikaros/Aiolos signaling pathways by IMiDs in CLL may reveal effective biomarkers. Proof-of-principle results indicate that Aiolos could act as a biomarker for T-cell activation as in vivo lenalidomide treatment resulted in downregulation of this transcription factor.11 

Preclinical studies suggest that the PI3Kδ inhibitor idelalisib may antagonize the immune-modulating properties of lenalidomide including repair of the humoral defect.45  Whether this translates to the clinic remains to be seen. Combining immune checkpoint blockade (eg, anti-PD-1 mAb) with lenalidomide may enhance antitumor T-cell immunity in CLL and lymphoma as both agents block immunosuppressive signaling.8  However, such immunostimulatory trials will need careful monitoring for potential autoimmune reactions. Ibrutinib, via ITK (interleukin-2-inducible T-cell kinase)-mediated inhibition of proleukemic Th2 CD4+ T cells,46  may also have complementary immunomodulatory potential with IMiDs, enhancing Th1 CD4+ and CD8+ T-cell immunity. In contrast to lenalidomide, the Bcl-2–specific inhibitor ABT-199/GDC-0199 directly induces tumor lysis. Thus, combining lenalidomide/IMiDs with ibrutinib or other new agents with distinct MOA has strong preclinical rationale and future studies will be of great interest.

The online version of this article contains a data supplement.

Contribution: A.G.R., A.P.K., S.H.T., and A.E. wrote the paper and approved the final version.

Conflict-of-interest disclosure: A.P.K. received research funding from Celgene. A.E. received research funding from, and provided consultancy for, Celgene. The remaining authors declare no competing financial interests.

Correspondence: Alan G. Ramsay, Department of Haemato-Oncology, Division of Cancer Studies, The Rayne Institute, King’s College London, 123 Coldharbour Lane, London, SE5 9NU, United Kingdom; e-mail: alan.ramsay@kcl.ac.uk.

1
Jones
 
JA
Byrd
 
JC
How will B-cell-receptor-targeted therapies change future CLL therapy?
Blood
2014
, vol. 
123
 
10
(pg. 
1455
-
1460
)
2
Burger
 
JA
Landau
 
D
Hoellenriegel
 
J
, et al. 
Clonal evolution in patients with chronic lymphocytic leukemia (CLL) developing resistance to BTK inhibition [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 866
3
Pardoll
 
DM
Immunology beats cancer: a blueprint for successful translation.
Nat Immunol
2012
, vol. 
13
 
12
(pg. 
1129
-
1132
)
4
Burger
 
JA
Gribben
 
JG
The microenvironment in chronic lymphocytic leukemia (CLL) and other B cell malignancies: insight into disease biology and new targeted therapies.
Semin Cancer Biol
2014
, vol. 
24
 (pg. 
71
-
81
)
5
Ding
 
W
Knox
 
TR
Tschumper
 
RC
, et al. 
Platelet-derived growth factor (PDGF)-PDGF receptor interaction activates bone marrow-derived mesenchymal stromal cells derived from chronic lymphocytic leukemia: implications for an angiogenic switch.
Blood
2010
, vol. 
116
 
16
(pg. 
2984
-
2993
)
6
Burger
 
JA
Chiorazzi
 
N
B cell receptor signaling in chronic lymphocytic leukemia.
Trends Immunol
2013
, vol. 
34
 
12
(pg. 
592
-
601
)
7
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
)
8
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
)
9
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
)
10
Lu
 
G
Middleton
 
RE
Sun
 
H
, et al. 
The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins.
Science
2014
, vol. 
343
 
6168
(pg. 
305
-
309
)
11
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
)
12
Billot
 
K
Soeur
 
J
Chereau
 
F
, et al. 
Deregulation of Aiolos expression in chronic lymphocytic leukemia is associated with epigenetic modifications.
Blood
2011
, vol. 
117
 
6
(pg. 
1917
-
1927
)
13
Orozco
 
CA
Acevedo
 
A
Cortina
 
L
, et al. 
The combined expression patterns of Ikaros isoforms characterize different hematological tumor subtypes.
PLoS ONE
2013
, vol. 
8
 
12
pg. 
e82411
 
14
Fecteau
 
J-F
Corral
 
LG
Futalan
 
D
, et al. 
Lenalidomide inhibits the proliferation of chronic lymphocytic leukemia cells via a cereblon/p21WAF1/Cip1-dependent mechanism [abstract].
Blood
 
2013;122(21). Abstract 4139
15
Fischer
 
ES
Böhm
 
K
Lydeard
 
JR
, et al. 
Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide.
Nature
2014
, vol. 
512
 
7512
(pg. 
49
-
53
)
16
Henry
 
JY
Labarthe
 
MC
Meyer
 
B
Dasgupta
 
P
Dalgleish
 
AG
Galustian
 
C
Enhanced cross-priming of naive CD8+ T cells by dendritic cells treated by the IMiDs immunomodulatory compounds lenalidomide and pomalidomide.
Immunology
2013
, vol. 
139
 
3
(pg. 
377
-
385
)
17
Lee
 
BN
Gao
 
H
Cohen
 
EN
, et al. 
Treatment with lenalidomide modulates T-cell immunophenotype and cytokine production in patients with chronic lymphocytic leukemia.
Cancer
2011
, vol. 
117
 
17
(pg. 
3999
-
4008
)
18
Ramsay
 
AG
Evans
 
R
Kiaii
 
S
Svensson
 
L
Hogg
 
N
Gribben
 
JG
Chronic lymphocytic leukemia cells induce defective LFA-1-directed T-cell motility by altering Rho GTPase signaling that is reversible with lenalidomide.
Blood
2013
, vol. 
121
 
14
(pg. 
2704
-
2714
)
19
Troeger
 
A
Johnson
 
AJ
Wood
 
J
, et al. 
RhoH is critical for cell-microenvironment interactions in chronic lymphocytic leukemia in mice and humans.
Blood
2012
, vol. 
119
 
20
(pg. 
4708
-
4718
)
20
Schulz
 
A
Dürr
 
C
Zenz
 
T
, et al. 
Lenalidomide reduces survival of chronic lymphocytic leukemia cells in primary cocultures by altering the myeloid microenvironment.
Blood
2013
, vol. 
121
 
13
(pg. 
2503
-
2511
)
21
Lapalombella
 
R
Andritsos
 
L
Liu
 
Q
, et al. 
Lenalidomide treatment promotes CD154 expression on CLL cells and enhances production of antibodies by normal B cells through a PI3-kinase-dependent pathway.
Blood
2010
, vol. 
115
 
13
(pg. 
2619
-
2629
)
22
Maffei
 
R
Fiorcari
 
S
Bulgarelli
 
J
, et al. 
Endothelium-mediated survival of leukemic cells and angiogenesis-related factors are affected by lenalidomide treatment in chronic lymphocytic leukemia.
Exp Hematol
2014
 
42(2):126-136
23
Gonzalez-Rodriguez
 
AP
Payer
 
AR
Acebes-Huerta
 
A
, et al. 
Lenalidomide and chronic lymphocytic leukemia.
Biomed Res Int
2013
 
2013:932010
24
Badoux
 
XC
Keating
 
MJ
Wen
 
S
, et al. 
Lenalidomide as initial therapy of elderly patients with chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
13
(pg. 
3489
-
3498
)
25
Strati
 
P
Keating
 
MJ
Wierda
 
WG
, et al. 
Lenalidomide induces long-lasting responses in elderly patients with chronic lymphocytic leukemia.
Blood
2013
, vol. 
122
 
5
(pg. 
734
-
737
)
26
Chen
 
CI
Bergsagel
 
PL
Paul
 
H
, et al. 
Single-agent lenalidomide in the treatment of previously untreated chronic lymphocytic leukemia.
J Clin Oncol
2011
, vol. 
29
 
9
(pg. 
1175
-
1181
)
27
Chen
 
C
Paul
 
H
Wang
 
T
, et al. 
Long-term follow-up of a phase 2 study of single agent lenalidomide in previously untreated, symptomatic chronic lymphocytic leukemia (CLL) [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 718
28
James
 
DF
Werner
 
L
Brown
 
JR
, et al. 
Lenalidomide and rituximab for the initial treatment of patients with chronic lymphocytic leukemia: a multicenter clinical-translational study from the chronic lymphocytic leukemia research consortium.
J Clin Oncol
2014
, vol. 
32
 
19
(pg. 
2067
-
2073
)
29
Brown
 
JR
Abramson
 
J
Hochberg
 
E
, et al. 
A phase I study of lenalidomide in combination with fludarabine and rituximab in previously untreated CLL/SLL.
Leukemia
2010
, vol. 
24
 
11
(pg. 
1972
-
1975
)
30
Flinn
 
IW
Cooper
 
RS
Thompson
 
DS
, et al. 
Fludarabine, Rituximab, and Lenalidomide in Previously Untreated Patients with Chronic Lymphocytic Leukemia (CLL): A Phase I/II Trial of the Sarah Cannon Research Institute [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 715
31
Egle
 
A
Steurer
 
M
Gassner
 
F
, et al. 
A combination of fludarabine/rituximab with escalating doses of lenalidomide in previously untreated chronic lymphocytic leukemia (CLL): The REVLIRIT CLL5 AGMT phase I/II study, Clinical and exploratory analyses of induction results [abstract].
Blood
2011
, vol. 
118
 
21
 
Abstract 292
32
Egle
 
A
Steurer
 
M
Gassner
 
F
, et al. 
Lenalidomide/rituximab maintenance after induction with fludarabine/rituximab in combination with escalating doses of lenalidomide in previously untreated chronic lymphocytic leukemia (CLL): The Revlirit CLL5 AGMT phase I/II study, final results [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 4146
33
Chanan-Khan
 
A
Miller
 
KC
Musial
 
L
, et al. 
Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a phase II study.
J Clin Oncol
2006
, vol. 
24
 
34
(pg. 
5343
-
5349
)
34
Ferrajoli
 
A
Lee
 
BN
Schlette
 
EJ
, et al. 
Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia.
Blood
2008
, vol. 
111
 
11
(pg. 
5291
-
5297
)
35
Wendtner
 
CM
Hillmen
 
P
Mahadevan
 
D
, et al. 
Final results of a multicenter phase 1 study of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia.
Leuk Lymphoma
2012
, vol. 
53
 
3
(pg. 
417
-
423
)
36
Buhler
 
A
Wendtner
 
C
Hallek
 
M
, et al. 
TP53 mutation or deletion and efficacy with single-agent lenalidomide in relapsed or refractory chronic lymphocytic leukemia (CLL) (CC-5013-CLL-009 Study) [abstract].
Blood
2013
, vol. 
122
 
21
 
Abstract 4146
37
Ferrajoli
 
A
O’Brien
 
S
Wierda
 
WG
, et al. 
Combination therapy with ofatumumab and lenalidomide in patients with relapsed chronic lymphocytic leukemia (CLL): results of a phase II trial [abstract].
Blood
2011
, vol. 
118
 
21
 
Abstract 1788
38
Costa
 
LJ
Fanning
 
S
Stephenson
 
J
, et al. 
Phase 2 trial of intracycle sequential ofatumumab and lenalidomide for the treatment of relapsed and refractory chronic lymphocytic leukemia [abstract].
Blood
2012
, vol. 
120
 
21
 
Abstract 3933
39
Sylvan
 
SE
Rossmann
 
E
Mozaffari
 
F
, et al. 
Phase I study of lenalidomide and alemtuzumab in refractory chronic lymphocytic leukaemia: maintaining immune functions during therapy-induced immunosuppression.
Br J Haematol
2012
, vol. 
159
 
5
(pg. 
608
-
612
)
40
Badoux
 
XC
Keating
 
MJ
Wen
 
S
, et al. 
Phase II study of lenalidomide and rituximab as salvage therapy for patients with relapsed or refractory chronic lymphocytic leukemia.
J Clin Oncol
2013
, vol. 
31
 
5
(pg. 
584
-
591
)
41
Shanafelt
 
TD
Ramsay
 
AG
Zent
 
CS
, et al. 
Longterm 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
)
42
Lapalombella
 
R
Yu
 
B
Triantafillou
 
G
, et al. 
Lenalidomide down-regulates the CD20 antigen and antagonizes direct and antibody-dependent cellular cytotoxicity of rituximab on primary chronic lymphocytic leukemia cells.
Blood
2008
, vol. 
112
 
13
(pg. 
5180
-
5189
)
43
Pal
 
R
Monaghan
 
SA
Hassett
 
AC
, et al. 
Immunomodulatory derivatives induce PU.1 down-regulation, myeloid maturation arrest, and neutropenia.
Blood
2010
, vol. 
115
 
3
(pg. 
605
-
614
)
44
Trudel
 
S
Scanga
 
S
Mastronardi
 
FG
, et al. 
GSK-3 spatial regulation has an essential role in lenalidomide activity and represents a biomarker of clinical response in CLL.
Blood
2012
 
120(21). Abstract 1793
45
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
)
46
Dubovsky
 
JA
Beckwith
 
KA
Natarajan
 
G
, et al. 
Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes.
Blood
2013
, vol. 
122
 
15
(pg. 
2539
-
2549
)
Sign in via your Institution