Intrinsic factors such as genetic lesions, anti-apoptotic proteins, and aberrant signaling networks within leukemia cells have long been the main focus of chronic lymphocytic leukemia (CLL) research. However, over the past decade, it became increasingly clear that external signals from the leukemia microenvironment make pivotal contributions to disease progression in CLL and other B-cell malignancies. Consequently, increasing emphasis is now placed on exploring and targeting the CLL microenvironment. This review highlights critical cellular and molecular pathways of CLL-microenvironment cross-talk. In vitro and in vivo models for studying the CLL microenvironment are discussed, along with their use in searching for therapeutic targets and in drug testing. Clinically, CXCR4 antagonists and small-molecule antagonists of B cell receptor (BCR)-associated kinases (spleen tyrosine kinase [Syk], Bruton's tyrosine kinase [Btk], and PI3Kδ) are the most advanced drugs for targeting specific interactions between CLL cells and the miocroenvironment. Preclinical and first clinical evidence suggests that high-risk CLL patients can particularly benefit from these alternative agents. These findings indicate that interplay between leukemia-inherent and environmental factors, nature and nurture determines disease progression in CLL.

In the bone marrow (BM) and secondary lymphatic tissues, chronic lymphocytic leukemia (CLL) cells engage in complex yet incompletely defined cellular and molecular interactions with stromal cells and matrix, collectively referred to as the “microenvironment.”1  In vitro models and gene-expression profiles (GEPs) have defined critical pathways for cross-talk between CLL cells and the microenvironment (Figure 1). These interactions affect CLL-cell survival and proliferation and confer drug resistance that may be responsible for residual disease after conventional therapy. Key cellular players are mesenchymal stromal cells, monocyte-derived nurse-like cells (NLCs), and T cells. The BCR plays a central role in the maintenance and expansion of the CLL clone, involving critical downstream kinases such as the spleen tyrosine kinase (Syk), Bruton's tyrosine kinase (Btk), and PI3Kδ. Chemokine receptors such as CXCR4 and CXCR5, in concert with adhesion molecules (VLA-4 and others), regulate CLL-cell trafficking and tissue homing via tissue gradients of the respective ligands. TNF family members such as the CD40 ligand, B cell-activating factor of the tumor necrosis factor (TNF) family (BAFF), and a proliferation-inducing ligand (APRIL) activate CLL cells via corresponding receptors, promoting immune recognition and/or survival and expansion of the leukemia cells. Another layer of complexity is added by the fact that CLL cells are not simply “seeds” that grow in the supportive soil of the microenvironment. Upon BCR activation, CLL cells secrete cytokines such as CCL3, which shape the microenvironment by attracting accessory cells such as T cells and monocytes. Clinically, the most advanced approaches for molecular targeting of the microenvironment are CXCR4 antagonists and B-cell kinase inhibitors, which disrupt regulatory loops between CLL cells and the microenvironment and have shown encouraging results in first, ongoing clinical trials. These findings demonstrate that the CLL microenvironment has become a highly dynamic translational research field.

Figure 1.

Molecular interactions in the CLL microenvironment. Molecular interactions between CLL and stromal cells in the BM and/or lymphoid tissue microenvironments that are considered important for CLL-cell survival and proliferation, CLL-cell homing, and tissue retention. Contact between CLL cells and NLCs or BMSCs is established and maintained by chemokine receptors and adhesion molecules expressed on CLL cells. NLCs express the chemokines CXCL12 and CXCL13, whereas BMSCs predominantly express CXCL12. The chemokine receptors CXCR3 and CCR7 are additional chemokine receptors on CLL cells that are involved in lymphatic tissue homing. NLCs and BMSCs attract CLL cells via the G protein–coupled chemokine receptors CXCR4 and CXCR5, which are expressed at high levels on CLL cells. Integrins, particularly VLA-4 integrins (CD49d), expressed on the surface of CLL cells cooperate with chemokine receptors in establishing cell-cell adhesion through respective ligands on the stromal cells (VCAM-1 and fibronectin). NLCs also express the TNF family members BAFF and APRIL, providing survival signals to CLL cells via corresponding receptors (BCMA, TACI, and BAFF-R). CD38 expression allows CLL cells to interact with CD31, the ligand for CD38 that is expressed by stromal and NLCs. Ligation of CD38 activates ZAP-70 and downstream survival pathways. Self- and/or environmental Ags are considered key factors in the activation and expansion of the CLL clone by activation of the BCR and its downstream kinases. Stimulation of the BCR complex (BCR and CD79a,b) induces downstream signaling by recruitment and activation of Syk, Btk, and PI3Ks. Finally, BCR stimulation and coculture with NLCs also induces CLL cells to secrete chemokines (CCL3, CCL4, and CCL22) for the recruitment of immune cells (T cells and monocytes) for cognate interactions. CD40L+ (CD154+) T cells are preferentially found in CLL-proliferation centers,38  and can interact with CLL cells via CD40. (Modified with permission from Burger et al.1 )

Figure 1.

Molecular interactions in the CLL microenvironment. Molecular interactions between CLL and stromal cells in the BM and/or lymphoid tissue microenvironments that are considered important for CLL-cell survival and proliferation, CLL-cell homing, and tissue retention. Contact between CLL cells and NLCs or BMSCs is established and maintained by chemokine receptors and adhesion molecules expressed on CLL cells. NLCs express the chemokines CXCL12 and CXCL13, whereas BMSCs predominantly express CXCL12. The chemokine receptors CXCR3 and CCR7 are additional chemokine receptors on CLL cells that are involved in lymphatic tissue homing. NLCs and BMSCs attract CLL cells via the G protein–coupled chemokine receptors CXCR4 and CXCR5, which are expressed at high levels on CLL cells. Integrins, particularly VLA-4 integrins (CD49d), expressed on the surface of CLL cells cooperate with chemokine receptors in establishing cell-cell adhesion through respective ligands on the stromal cells (VCAM-1 and fibronectin). NLCs also express the TNF family members BAFF and APRIL, providing survival signals to CLL cells via corresponding receptors (BCMA, TACI, and BAFF-R). CD38 expression allows CLL cells to interact with CD31, the ligand for CD38 that is expressed by stromal and NLCs. Ligation of CD38 activates ZAP-70 and downstream survival pathways. Self- and/or environmental Ags are considered key factors in the activation and expansion of the CLL clone by activation of the BCR and its downstream kinases. Stimulation of the BCR complex (BCR and CD79a,b) induces downstream signaling by recruitment and activation of Syk, Btk, and PI3Ks. Finally, BCR stimulation and coculture with NLCs also induces CLL cells to secrete chemokines (CCL3, CCL4, and CCL22) for the recruitment of immune cells (T cells and monocytes) for cognate interactions. CD40L+ (CD154+) T cells are preferentially found in CLL-proliferation centers,38  and can interact with CLL cells via CD40. (Modified with permission from Burger et al.1 )

Close modal

BMSCs and other MSCs

BM stromal cells (BMSCs) were the first stromal cells characterized to support CLL cells.2–4  Normal hematopoiesis depends on BMSCs, which provide attachment sites and growth factors to hematopoietic precursors. In CLL, BMSCs are thought to function in a similar fashion, creating niches within the BM in which CLL cells lodge and are nourished and protected from cytotoxic agents. CLL cells have a high affinity for BMSCs: coculture with BMSCs results in the adhesion and rapid, spontaneous migration of a fraction of CLL cells beneath and underneath the BMSCs (pseudoemperipolesis), inducing a cobblestone-like appearance4  that depends upon CXCR4 and VLA-4 expression by leukemia cells.4  Consequently, the protective effects of BMSCs are largely dependent on close proximity between CLL and stromal cells.3,5  Murine and human BMSCa are very similar in their capacity to protect CLL cells,5  and a recent murine in vivo model of CLL6  also demonstrated that the murine BM microenvironment was adequate for supporting CLL-cell growth, suggesting that key cellular and molecular pathways in the BM are evolutionarily conserved between mice and humans.

BMSCs are of mesenchymal origin and are similar to actin (αSMA+)–positive mesenchymal stromal cells (MSCs) in other tissues such as the secondary lymphatic tissues. Dense infiltration with αSMA+ stromal cells in lymphatic tissues from CLL patients7  suggests that interactions between CLL cells and this type of stromal cells plays an important role even outside of the BM. Follicular dendritic cells (FDCs), another cell type present in lymphatic tissues, can also support CLL cells,8  although the comparability of the FDC cell line used in this study to FDCs in vivo is questionable. Interestingly, the cross-talk between CLL and MSCs is bidirectional, causing activation of both CLL cells and MSCs.9  CLL cells have a high affinity for BMSCs and the CXCR4-CXCL12 axis is critical for this interaction.

NLCs differentiate in vitro from monocytes into large, round, adherent cells that attract CLL cells and protect them from undergoing spontaneous or drug-induced cell death in a contact-dependent fashion.10,11  Because these cells share features in common with thymic nurse cells that nurture developing thymocytes, we designated these cells “nurse-like cells,” or NLCs.10  NLCs can be found in the spleen and secondary lymphoid tissue of patients with CLL,11,12  and thus represent a model for the microenvironment in secondary lymphatic tissues. This is supported by the notion that GEPs of CLL cells after NLC coculture13  are strikingly similar to GEPs of CLL cells isolated from secondary lymphatic tissues,14  which show signs of BCR and NF-κB activation and up-regulation of BCR target genes such as CCL3 and CCL4.13,14  The diverse molecular mechanisms of CLL-NLC cross-talk have been extensively studied,10–13,15–20  and have revealed important pathways and therapeutic targets of CLL-microenvironment cross-talk. For example, NLCs express CXCL12,21  CXCL13,12  CD31, plexin B1,16  BAFF, APRIL,15  and vimentin.10,18  These findings, along with the GEP data, suggest that NLCs represent a highly relevant in vitro model for studying the CLL lymphatic tissue microenvironment. High levels of CD68 make NLCs comparable to CD68+ lymphoma-associated macrophages in follicular lymphoma and other B-cell lymphomas, and suggest that NLCs could be a model system for studying the lymphatic tissue microenvironment in other B-cell malignancies.

T lymphocytes

The importance of T cells remains one of the most controversial areas in CLL research. In untreated CLL patients, the overall number of circulating T cells, oligoclonal in both the CD4 and the CD8 compartment, is increased. It is unresolved whether these T cells are expanded because of interactions with the CLL clone, with microbial antigens that are more prevalent in CLL patients, or for other reasons. To make matters more complex, T cells can either suppress or stimulate the expansion of the CLL clone. Another layer of complexity is added by functional impairments of T cells from CLL patients, which fail to form appropriate immunological synapses with CLL cells,22  a dysfunction that can be reversed with the immunomodulatory drug lenalidomide.22,23  In tissue areas of CLL-cell proliferation, called proliferation centers or pseudofollicles, activated CD4+ T cells colocalize with proliferating CD38+ CLL cells,24  suggesting that T-cell subpopulations promote the expansion of the CLL clone. This is supported by recent in vivo evidence in a CLL-transfer model into immunodeficient mice demonstrating that activated CD4+ T cells support CLL-cell proliferation.6 

The CXCR4-CXCL12 axis

The CXCR4 chemokine receptor (CD184) is expressed at high levels on the surface of peripheral blood CLL cells4  and mediates leukemia cell chemotaxis, migration across vascular endothelium, actin polymerization, and migration beneath and underneath BMSCs in response to CXCL12 gradients (pseudoemperipolesis).4  CXCL12 also displays a direct prosurvival effect in CLL.10,15  CXCR4 surface expression is regulated by its ligand CXCL12 (previously called stromal cell-derived factor-1/SDF-1) via receptor endocytosis (Table 1).4  This characteristic can be used to distinguish tissue (lymphatic and BM derived) from blood CLL cells, which express low or high CXCR4 levels, respectively.4,14  Proliferating Ki-67+ CLL cells from BM and lymphatic tissue display significantly lower levels of CXCR4 and CXCR5 than nonproliferating CLL cells.25  In vivo deuterium (2H) labeling of CLL cells revealed an enrichment of CLL cells expressing lower CXCR4 surface levels in the CD38+/CD5bright fraction, along with increased 2H incorporation.26  These in vivo data indicate that CLL subclones with lower blood CXCR4 surface levels are a fraction of cells that has recently exited the tissues into the blood. BCR signaling results in the down-modulation of CXCR4,17,27  along with enhanced chemotaxis toward CXCL12 and CXCL13.17  This may explain why ZAP-70+ CLL cells display increased chemotaxis and survival in response to CXCL12 compared with ZAP-70 CLL cells,28  given that ZAP-70 expression is associated with a higher responsiveness to BCR stimulation.29  CD38+ CLL cells also display higher levels of chemotaxis,30  and CD38 activation enhances chemotaxis toward CXCL12, whereas a blocking anti-CD38 mAb inhibits chemotaxis.31  CXCR4 signaling in CLL cells is pertussis toxin–sensitive and induces calcium mobilization, activation of PI3Ks,4  p44/42 MAPKs,10  and serine phosphorylation of STAT3.32  CXCR4 signaling can be inhibited by isoform-selective PI3K inhibitors,33  Syk inhibitors,17  and Btk inhibitors,34  leading to impaired migration of normal B cells and CLL cells.

Table 1.

Chemokine receptors (top) and inducible chemokines (bottom) involved in cross-talk between CLL cells and the microenvironment

Chemokine receptors (top) and inducible chemokines (bottom) involved in cross-talk between CLL cells and the microenvironment
Chemokine receptors (top) and inducible chemokines (bottom) involved in cross-talk between CLL cells and the microenvironment

CXCR4 can be specifically blocked by CXCR4 antagonists,35  which inhibit CLL-cell activation by CXCL12 and reverse, at least in part, stomal cell–mediated drug resistance.32  These data are the basis for an ongoing clinical trial in relapsed CLL patients, in which patients are treated with a combination of rituximab and plerixafor, a small-molecule CXCR4 antagonist. This is a proof-of-principle trial to determine whether leukemia cells can be mobilized from the tissues using a CXCR4 antagonist and then be targeted outside of the protective tissues. Preliminary data from this trial indicate a plerixafor dose–dependent mobilization of CLL cells from the tissues to the blood.36 

CXCR5 (CD185) is the receptor for the chemokine CXCL13, which regulates lymphocyte homing and positioning within lymph follicles (Table 1). CXCR5 gene-deleted mice display defective formation of primary follicles and germinal centers in the spleen and Payer patches and lack inguinal lymph nodes. CXCL13 gradients induce recruitment of circulating naive B cell to follicles,37  and are involved in the microanatomic positioning of B cells within the germinal center.37  CLL cells express high levels of CXCR5,12  and stimulation with CXCL13 induces activation via Gi proteins, PI3Ks, and p44/42 MAPK, resulting in actin polymerization, CXCR5 endocytosis, and chemotaxis.12  CXCL13 mRNA and protein are expressed by NLC in vitro and in vivo.12  These data suggest that CXCR5 plays a role in CLL-cell positioning and cognate interactions between CLL and CXCL13-secreting NLCs in lymphoid tissues.

CCL3, CCL4, and CCL22

Activated CLL cells secrete CCL3, CCL4,13  and CCL22,38  which are chemoattractants for T lymphocytes and monocytes (Table 1). CCL3 and CCL4 normally are secreted by B cells after activation via the BCR and CD40 ligand.39  CLL cells secrete CCL3 and CCL4 in response to BCR stimulation and in coculture with NLCs,13  which are sensitive to inhibition of BCR signaling, using, for example, a Syk or Btk inhibitor.13,17  CLL patients display elevated plasma levels of CCL3 and CCL4,13  and plasma levels of CCL3 were strongly associated with established prognostic markers and time to treatment.40  A multivariable analysis revealed that CCL3, advanced clinical stage, poor-risk cytogenetics, and CD38 expression were independent prognostic markers in a cohort of 351 CLL patients.40  Conceivably, CLL cell–derived CCL3 may induce trafficking of T cells to activated, CD38+/Ki-67+ CLL cells for cognate T-CLL cell interactions that foster CLL-cell proliferation.13,24,41  Conceptually, by attracting T cells and other immune cells, CLL-cell–derived chemokines foster the coevolution of CLL cells and their supportive microenvironment, thereby actively creating a favorable microenvironment.

Adhesion molecules

Integrins are heterodimeric glycoproteins consisting of various α and β subunits, the function of which is to mediate cell-cell and cell-matrix adhesion. β1 integrins are very late activation antigens (VLAs) that have the same β1 subunit but various α chains (α1-α6). The α4β1 integrin VLA-4 (CD49d) is a receptor for fibronectin and vascular cell adhesion molecule-1 (VCAM-1/CD106). VLA-4 plays a particularly important role in interactions between normal and malignant hematopoietic cells and the BM microenvironment. VLA-4 integrins cooperate with CXCR4 in CLL-cell adhesion to BMSCs.4  Moreover, VLA-4 expression on CLL cells has prognostic impact,42,43  indicating the relevance of these interactions in vivo. These studies indicate that VLA-4 integrins play a key role in the adhesion of CLL and other leukemia cells to stromal cells and the extracellular matrix. Other important adhesion molecules in CLL include L-selectin (CD62L) and CD44 (Table 2).

Table 2.

Expression and function of CLL adhesion molecules

Expression and function of CLL adhesion molecules
Expression and function of CLL adhesion molecules

CD40-CD154 interactions

Within proliferation centers, a significant number of T cells display CD40 ligand (CD154)38,44  that can bind to CD40 on CLL cells, rescuing them from apoptosis.45  Conversely, CD40 cross-linking also induces up-regulation of CD80 and CD54 and turns nonimmunogenic CLL cells into effective T-cell stimulators.46  CLL cells engineered to express CD154 by adenoviral gene transfer can cross-link CD40 on bystander CLL cells and induce the same sequence of activation and immune recognition. CLL patients infused with CLL cells transduced in vitro with an adenovirus encoding CD40L (Ad-CD154)47  can make antibodies to ROR1,48  an oncofetal antigen restricted to CLL cells, indicating that this approach can overcome immune tolerance. These data suggest that CD40 activation of CLL cells can result in different outcomes that are not mutually contradictory: activation of prosurvival and proliferation pathways if triggered by CD154+ T-cells in the context of proliferation centers, or immune recognition and induction of a specific immune response if triggered in the context of Ad-CD154–transduced CLL cells.

BAFF and APRIL are related TNF family ligands that bind to members of the TNF receptor family such as B-cell maturation antigen (BCMA) and transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI). BAFF also binds another TNF receptor, the BAFF receptor (BAFF-R). Binding to heparin sulfate proteoglycans allows for tissue retention of APRIL. Via BCMA, TACI, and BAFF-R expressed on the CLL cells, NLC-derived BAFF and APRIL induce activation of the canonical NF-κB pathway and protect CLL cells from apoptosis.15,49  Male v-Myc myelocytomatosis viral oncogene homolog (c-Myc) transgenic mice develop a CD5 B-cell leukemia resembling CLL after cross-breeding with Baff-transgenic mice,50  with Baff-induced enhancement of c-Myc, suggesting an important relationship between BAFF and c-MYC in CLL. Aging April-transgenic mice develop an expansion of CD5+ B1 B cells with organ infiltration, which can be reversed with anti-April mAbs.51 

Currently, we assume that in an appropriate microenvironment, the BCRs of CLL cells become engaged by microbial or auto-antigens,52–54  which, along with other costimulatory signals, promote the expansion of the CLL clone. Several lines of evidence suggest that antigen stimulation and BCR-derived signals play a critical role in the pathogenesis and prognosis of CLL.44,52  First, the prognosis of CLL patients is correlated with the number of somatic mutations in the variable regions of the BCR. Second, CLL cells express a restricted set of BCRs with Ig heavy-chain variable gene sequences that are identical or stereotyped in subsets of patients, suggesting that these BCRs bind similar antigens that are relevant to the pathogenesis of CLL. Third, Ig-unmutated and/or ZAP-70+ patients preferentially respond to BCR stimulation29  and display GEPs suggesting activation downstream of the BCR.55  BCR signaling is a complex process. The BCR is composed of an antigen-specific membrane Ig paired with Ig-α/Ig-β heterodimers (CD79α/CD79β). Engagement of BCRs by antigen induces phosphorylation of immunoreceptor tyrosine-based activation motifs in the cytoplasmic tails of Ig-α and Ig-β, with subsequent recruitment of Syk to BCR microclusters, followed by downstream activation of Btk and PI3Ks (Table 3). Upon phosphorylation, Syk, Btk, and PI3Ks propagate BCR-derived signals by activating downstream signaling pathways, including calcium mobilization and activation of AKT kinase, ERK1/2, and myeloid cell leukemia-1 (MCL-1). The notion that ZAP-70+29  and unmutated CLL cells20,56  are more responsive to BCR stimulation and other microenvironmental signals28  suggests that patients with high-risk disease features may be particularly well suited for alternative treatments that target the microenvironment. Indeed, early clinical data from ongoing trials with the Btk inhibitor PCI-3276557  and the PI3K inhibitor CAL-10158  suggest that high-risk CLL patients respond well to these agents that disrupt CLL-microenvironment interactions.

Table 3.

Expression and function of BCR-associated kinases

Expression and function of BCR-associated kinases
Expression and function of BCR-associated kinases

In vivo evidence

In vivo studies of the CLL microenvironment have been decelerated by the lack of suitable mouse models and limited access to primary CLL tissue samples. However, this has recently changed after publication of a landmark paper.14  Herishanu et al explored GEPs of CLL cells isolated from different tissues (blood, BM, and lymphatic tissues)14  which revealed BCR and NF-κB activation in CLL cells in the lymphatic tissues,14  indicating that the lymphatic tissues are a key site of CLL activation and proliferation. Activation of these pathways was particularly prominent in high-risk CLL patients, suggesting more effective BCR signaling in this subtype in vivo. Recent studies using mouse models are also starting to explore the question of how different elements of the microenvironment contribute to CLL pathogenesis. For example, a recent murine adoptive-transfer model demonstrated a central role of T cells in the progression of CLL in this setting.6 

The CXCR4-CXCL12 axis

CXCR4 antagonists, such as plerixafor (Mozobil, AMD3100) and T140 analogs, disrupt CLL-cell adhesion to BMSCs32  and mobilize CLL cells from protective tissue microenvironments to the blood, making them more accessible to conventional drugs. Targeting the CXCR4-CXCL12 axis is currently being explored in a first clinical trial in CLL patients.36  CXCR4 antagonists were initially developed for the treatment of HIV, in which CXCR4 functions as a coreceptor for virus entry into T cells. In these first trials, CXCR4 antagonists were observed to induce leukocytosis and thus were developed for mobilization of hematopoietic progenitors in the context of autologous stem cell mobilization in myeloma and lymphoma patients. This is the currently approved indication for plerixafor. The ongoing CLL trial combines plerixafor with rituximab, and preliminary data indicate a plerixafor dose–dependent CLL-cell mobilization to the blood and show the safety of this drug combination.36  Future studies in CLL could combine a CXCR4 antagonist with established cytotoxic agents or antibodies or in the setting of minimal residual disease, in which these agents could help to mobilize and eliminate residual CLL cells from tissue sanctuaries.35 

Orally bioavailable inhibitors of kinases downstream of the BCR are currently tested in first clinical trials in CLL patients, and are generating excitement because of promising early response data and benign side effect profiles, particularly the lack of myelosuppression.57–59  These new targeted agents are the Syk inhibitor fostamatinib disodium,59  the Btk inhibitor PCI-32765,60  and the PI3Kδ inhibitor CAL-10161  (Table 3). Characteristically, these kinase inhibitors induce rapid lymph node shrinkage, along with a transient lymphocytosis during the first weeks of treatment, which is presumably due to mobilization of CLL cells from the tissues into the blood. Inhibition of signaling through CXCR4, and potentially other chemokine receptors and adhesion molecules,17,33,34  has been proposed as the basis for this remarkable phenomenon. Future research on these agents will need to address the question of whether CLL-cell mobilization is a prerequisite for the efficacy of these agents. Mobilized into the blood, CLL cells may simply die from neglect (ie, a lack of tissue-derived survival and growth signals). However, in vitro data suggest that these agents also effectively block signaling downstream of the BCR and other surface receptors,17,62  implying dual effects on CLL cells: deprivation from exogenous survival signals and blockade of prosurvival signaling.

Targeting of BCR signaling interferes not only with pathways related to survival, but also with tissue homing and retention of CLL cells. Specifically, BCR signaling within the lymphatic tissues can enhance responsiveness of CLL cells to chemokines, which then can result in the retention of such activated CLL cells. We previously reported that BCR engagement on CLL cells induces an adhesive phenotype with increased expression of adhesion and costimulatory molecules, along with increased chemotaxis toward the chemokines CXCL12 and CXCL13 and increased migration beneath BM stromal cells.17  These responses were more noticeable in ZAP-70+ cases and were sensitive to inhibition by R406, a small-molecule Syk inhibitor.17  A question remaining is what is the mode of BCR activation and the nature of antigen(s) that stimulate CLL BCRs. Indirect evidence based on cytokine (CCL3 and CCL4) secretion and GEPs of CLL cells cultured with NLCs,13  which are remarkably similar to GEPs of activated CLL cells isolated from lymph nodes, suggests that such BCRs can react with antigen on stromal cells (ie, NLCs).14  Direct binding of CLL BCRs to vimentin on the surface of stromal cells (BMSCs and NLCs) and vimentin-induced activation support this concept.18  Nonmuscle myosin heavy chain IIA (MYHIIA) is an alternative antigen derived from apoptotic cells that is recognized by CLL-cell mAbs.53 

During the coming years, increasing emphasis will be placed on targeting the microenvironment in CLL. Clinically, CXCR4 and the BCR-associated kinases Syk, Btk, and PI3Kδ represent the most advanced therapeutic targets in the complex cross-talk between CLL cells and their microenvironment. Despite the current enthusiasm for these new targeted approaches, which is justified based on the clinical activity and the lack of major side effects such as myelosuppression,57–59  the precise mechanism of action of these agents and the potential benefit of combinations with conventional agents remain to be explored. Additional in vivo and in vitro studies will accelerate the development of these new concepts and will help to define the best drug combinations. Moreover, numerous other pathways of CLL-microenvironment interactions, such as VLA-4 or BAFF and APRIL, represent alternative therapeutic targets that are likely to be explored in the near future.

This work was supported by a CLL Global Research Foundation grant and by a Cancer Prevention and Research Institute of Texas (CPRIT) grant.

Conflict-of-interest disclosure: The author has received research funding from Noxxon, Calistoga, and Pharmacyclics and has consulted for Noxxon, Pharmacyclics, Genzyme, and Celgene. Off-label drug use: Plerixafor for leukemia cell mobilization.

Jan A. Burger, MD, PhD, Department of Leukemia, Unit 428, The University of Texas M. D. Anderson Cancer Center, PO Box 301402, Houston, TX 77230; Phone: (713) 563-1487 or (713) 792-1865; Fax: (713) 794-4297; e-mail: jaburger@mdanderson.org.

1
Burger
 
JA
Ghia
 
P
Rosenwald
 
A
Caligaris-Cappio
 
F
The microenvironment in mature B-cell malignancies: a target for new treatment strategies
Blood
2009
, vol. 
114
 
16
(pg. 
3367
-
3375
)
2
Panayiotidis
 
P
Jones
 
D
Ganeshaguru
 
K
Foroni
 
L
Hoffbrand
 
AV
Human bone marrow stromal cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro
Br J Haematol
1996
, vol. 
92
 
1
(pg. 
97
-
103
)
3
Lagneaux
 
L
Delforge
 
A
Bron
 
D
De Bruyn
 
C
Stryckmans
 
P
Chronic lymphocytic leukemic B cells but not normal B cells are rescued from apoptosis by contact with normal bone marrow stromal cells
Blood
1998
, vol. 
91
 
7
(pg. 
2387
-
2396
)
4
Burger
 
JA
Burger
 
M
Kipps
 
TJ
Chronic Lymphocytic Leukemia B Cells Express Functional CXCR4 Chemokine Receptors That Mediate Spontaneous Migration Beneath Bone Marrow Stromal Cells
Blood
1999
, vol. 
94
 
11
(pg. 
3658
-
3667
)
5
Kurtova
 
AV
Balakrishnan
 
K
Chen
 
R
, et al. 
Diverse marrow stromal cells protect CLL cells from spontaneous and drug-induced apoptosis: development of a reliable and reproducible system to assess stromal cell adhesion-mediated drug resistance
Blood
2009
, vol. 
114
 
20
(pg. 
4441
-
4450
)
6
Bagnara
 
D
Kaufman
 
MS
Calissano
 
C
, et al. 
A novel adoptive transfer model of chronic lymphocytic leukemia suggests a key role for T lymphocytes in the disease
Blood
2011
, vol. 
117
 
20
(pg. 
5463
-
5472
)
7
Ruan
 
J
Hyjek
 
E
Kermani
 
P
, et al. 
Magnitude of stromal hemangiogenesis correlates with histologic subtype of non-Hodgkin's lymphoma
Clin Cancer Res
2006
, vol. 
12
 
19
(pg. 
5622
-
5631
)
8
Pedersen
 
IM
Kitada
 
S
Leoni
 
LM
, et al. 
Protection of CLL B cells by a follicular dendritic cell line is dependent on induction of Mcl-1
Blood
2002
, vol. 
100
 
5
(pg. 
1795
-
1801
)
9
Ding
 
W
Nowakowski
 
GS
Knox
 
TR
, et al. 
Bi-directional activation between mesenchymal stem cells and CLL B-cells: implication for CLL disease progression
Br J Haematol
2009
, vol. 
147
 
4
(pg. 
471
-
483
)
10
Burger
 
JA
Tsukada
 
N
Burger
 
M
Zvaifler
 
NJ
Dell'Aquila
 
M
Kipps
 
TJ
Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1
Blood
2000
, vol. 
96
 
8
(pg. 
2655
-
2663
)
11
Tsukada
 
N
Burger
 
JA
Zvaifler
 
NJ
Kipps
 
TJ
Distinctive features of “nurselike” cells that differentiate in the context of chronic lymphocytic leukemia
Blood
2002
, vol. 
99
 
3
(pg. 
1030
-
1037
)
12
Bürkle
 
A
Niedermeier
 
M
Schmitt-Graff
 
A
Wierda
 
WG
Keating
 
MJ
Burger
 
JA
Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia
Blood
2007
, vol. 
110
 
9
(pg. 
3316
-
3325
)
13
Burger
 
JA
Quiroga
 
MP
Hartmann
 
E
, et al. 
High-level expression of the T-cell chemokines CCL3 and CCL4 by chronic lymphocytic leukemia B cells in nurselike cell cocultures and after BCR stimulation
Blood
2009
, vol. 
113
 
13
(pg. 
3050
-
3058
)
14
Herishanu
 
Y
Perez-Galan
 
P
Liu
 
D
, et al. 
The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia
Blood
2011
, vol. 
117
 
2
(pg. 
563
-
574
)
15
Nishio
 
M
Endo
 
T
Tsukada
 
N
, et al. 
Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF-1alpha
Blood
2005
, vol. 
106
 
3
(pg. 
1012
-
1020
)
16
Deaglio
 
S
Vaisitti
 
T
Bergui
 
L
, et al. 
CD38 and CD100 lead a network of surface receptors relaying positive signals for B-CLL growth and survival
Blood
2005
, vol. 
105
 
8
(pg. 
3042
-
3050
)
17
Quiroga
 
MP
Balakrishnan
 
K
Kurtova
 
AV
, et al. 
B-cell antigen receptor signaling enhances chronic lymphocytic leukemia cell migration and survival: specific targeting with a novel spleen tyrosine kinase inhibitor, R406
Blood
2009
, vol. 
114
 
5
(pg. 
1029
-
1037
)
18
Binder
 
M
Lechenne
 
B
Ummanni
 
R
, et al. 
Stereotypical chronic lymphocytic leukemia B-cell receptors recognize survival promoting antigens on stromal cells
PLoS One
2010
, vol. 
5
 
12
pg. 
e15992
 
19
Ferretti
 
E
Bertolotto
 
M
Deaglio
 
S
, et al. 
A novel role of the CX(3)CR1/CX(3)CL1 system in the cross-talk between chronic lymphocytic leukemia cells and tumor microenvironment
Leukemia
2011
, vol. 
25
 
8
(pg. 
1268
-
1277
)
20
Coscia
 
M
Pantaleoni
 
F
Riganti
 
C
, et al. 
IGHV unmutated CLL B cells are more prone to spontaneous apoptosis and subject to environmental prosurvival signals than mutated CLL B cells
Leukemia
2011
, vol. 
25
 
5
(pg. 
828
-
837
)
21
Burger
 
JA
Tsukada
 
N
Burger
 
M
Zvaifler
 
NJ
Dell'Aquila
 
M
Kipps
 
TJ
Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1
Blood
2000
, vol. 
96
 
8
(pg. 
2655
-
2663
)
22
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
)
23
Gorgun
 
G
Ramsay
 
AG
Holderried
 
TA
, et al. 
E(mu)-TCL1 mice represent a model for immunotherapeutic reversal of chronic lymphocytic leukemia-induced T-cell dysfunction
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
15
(pg. 
6250
-
6255
)
24
Patten
 
PE
Buggins
 
AG
Richards
 
J
, et al. 
CD38 expression in chronic lymphocytic leukemia is regulated by the tumor microenvironment
Blood
2008
, vol. 
111
 
10
(pg. 
5173
-
5181
)
25
Bennett
 
F
Rawstron
 
A
Plummer
 
M
, et al. 
B-cell chronic lymphocytic leukaemia cells show specific changes in membrane protein expression during different stages of cell cycle
Br J Haematol
2007
, vol. 
139
 
4
(pg. 
600
-
604
)
26
Calissano
 
C
Damle
 
RN
Hayes
 
G
, et al. 
In vivo intraclonal and interclonal kinetic heterogeneity in B-cell chronic lymphocytic leukemia
Blood
2009
, vol. 
114
 
23
(pg. 
4832
-
4842
)
27
Vlad
 
A
Deglesne
 
PA
Letestu
 
R
, et al. 
Down-regulation of CXCR4 and CD62L in chronic lymphocytic leukemia cells is triggered by B-cell receptor ligation and associated with progressive disease
Cancer Res
2009
, vol. 
69
 
16
(pg. 
6387
-
6395
)
28
Richardson
 
SJ
Matthews
 
C
Catherwood
 
MA
, et al. 
ZAP-70 expression is associated with enhanced ability to respond to migratory and survival signals in B-cell chronic lymphocytic leukemia (B-CLL)
Blood
2006
, vol. 
107
 
9
(pg. 
3584
-
3592
)
29
Chen
 
L
Widhopf
 
G
Huynh
 
L
, et al. 
Expression of ZAP-70 is associated with increased B-cell receptor signaling in chronic lymphocytic leukemia
Blood
2002
, vol. 
100
 
13
(pg. 
4609
-
4614
)
30
Deaglio
 
S
Vaisitti
 
T
Aydin
 
S
, et al. 
CD38 and ZAP-70 are functionally linked and mark CLL cells with high migratory potential
Blood
2007
, vol. 
110
 
12
(pg. 
4012
-
4021
)
31
Vaisitti
 
T
Aydin
 
S
Rossi
 
D
, et al. 
CD38 increases CXCL12-mediated signals and homing of chronic lymphocytic leukemia cells
Leukemia
2010
, vol. 
24
 
5
(pg. 
958
-
969
)
32
Burger
 
M
Hartmann
 
T
Krome
 
M
, et al. 
Small peptide inhibitors of the CXCR4 chemokine receptor (CD184) antagonize the activation, migration, and antiapoptotic responses of CXCL12 in chronic lymphocytic leukemia B cells
Blood
2005
, vol. 
106
 
5
(pg. 
1824
-
1830
)
33
Niedermeier
 
M
Hennessy
 
BT
Knight
 
ZA
, et al. 
Isoform-selective phosphoinositide 3′-kinase inhibitors inhibit CXCR4 signaling and overcome stromal cell-mediated drug resistance in chronic lymphocytic leukemia: a novel therapeutic approach
Blood
2009
, vol. 
113
 
22
(pg. 
5549
-
5557
)
34
de Gorter
 
DJ
Beuling
 
EA
Kersseboom
 
R
, et al. 
Bruton's tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing
Immunity
2007
, vol. 
26
 
1
(pg. 
93
-
104
)
35
Burger
 
JA
Peled
 
A
CXCR4 antagonists: targeting the microenvironment in leukemia and other cancers
Leukemia
2009
, vol. 
23
 
1
(pg. 
43
-
52
)
36
Andritsos
 
LA
Byrd
 
JC
Hewes
 
B
Kipps
 
TJ
Johns
 
D
Burger
 
JA
Preliminary results from a phase I/II dose escalation study to determine the maximum tolerated dose of plerixafor in combination with rituximab in patients with relapsed chronic lymphocytic leukemia [Abstract]
Haematologica
2010
, vol. 
95
 
suppl 2
 
0772
37
Mueller
 
SN
Germain
 
RN
Stromal cell contributions to the homeostasis and functionality of the immune system
Nat Rev Immunol
2009
, vol. 
9
 
9
(pg. 
618
-
629
)
38
Ghia
 
P
Strola
 
G
Granziero
 
L
, et al. 
Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22
Eur J Immunol
2002
, vol. 
32
 
5
(pg. 
1403
-
1413
)
39
Krzysiek
 
R
Lefevre
 
EA
Zou
 
W
, et al. 
Antigen receptor engagement selectively induces macrophage inflammatory protein-1 alpha (MIP-1 alpha) and MIP-1 beta chemokine production in human B cells
J Immunol
1999
, vol. 
162
 
8
(pg. 
4455
-
4463
)
40
Sivina
 
M
Hartmann
 
E
Kipps
 
TJ
, et al. 
CCL3 (MIP-1alpha) plasma levels and the risk for disease progression in chronic lymphocytic leukemia
Blood
2011
, vol. 
117
 
5
(pg. 
1662
-
1669
)
41
Zucchetto
 
A
Tripodo
 
C
Benedetti
 
D
, et al. 
Monocytes/macrophages but not T lymphocytes are the major targets of the CCL3/CCL4 chemokines produced by CD38(+)CD49d(+) chronic lymphocytic leukaemia cells
Br J Haematol
2010
, vol. 
150
 
1
(pg. 
111
-
113
)
42
Shanafelt
 
TD
Geyer
 
SM
Bone
 
ND
, et al. 
CD49d expression is an independent predictor of overall survival in patients with chronic lymphocytic leukaemia: a prognostic parameter with therapeutic potential
Br J Haematol
2008
, vol. 
140
 
5
(pg. 
537
-
546
)
43
Majid
 
A
Lin
 
TT
Best
 
G
, et al. 
CD49d is an independent prognostic marker that is associated with CXCR4 expression in CLL
Leuk Res
2011
, vol. 
35
 
6
(pg. 
750
-
756
)
44
Stevenson
 
FK
Caligaris-Cappio
 
F
Chronic lymphocytic leukemia: revelations from the B-cell receptor
Blood
2004
, vol. 
103
 
12
(pg. 
4389
-
4395
)
45
Kitada
 
S
Zapata
 
JM
Andreeff
 
M
Reed
 
JC
Bryostatin and CD40-ligand enhance apoptosis resistance and induce expression of cell survival genes in B-cell chronic lymphocytic leukaemia
Br J Haematol
1999
, vol. 
106
 
4
(pg. 
995
-
1004
)
46
Ranheim
 
EA
Kipps
 
TJ
Activated T cells induce expression of B7/BB1 on normal or leukemic B cells through a CD40-dependent signal
J Exp Med
1993
, vol. 
177
 
4
(pg. 
925
-
935
)
47
Wierda
 
WG
Cantwell
 
MJ
Woods
 
SJ
Rassenti
 
LZ
Prussak
 
CE
Kipps
 
TJ
CD40-ligand (CD154) gene therapy for chronic lymphocytic leukemia
Blood
2000
, vol. 
96
 
9
(pg. 
2917
-
2924
)
48
Fukuda
 
T
Chen
 
L
Endo
 
T
, et al. 
Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
8
(pg. 
3047
-
3052
)
49
Endo
 
T
Nishio
 
M
Enzler
 
T
, et al. 
BAFF and APRIL support chronic lymphocytic leukemia B-cell survival through activation of the canonical NF-kappaB pathway
Blood
2007
, vol. 
109
 
2
(pg. 
703
-
710
)
50
Zhang
 
W
Kater
 
AP
Widhopf
 
GF
, et al. 
B-cell activating factor and v-Myc myelocytomatosis viral oncogene homolog (c-Myc) influence progression of chronic lymphocytic leukemia
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
44
(pg. 
18956
-
18960
)
51
Guadagnoli
 
M
Kimberley
 
F
Phan
 
U
, et al. 
Development and characterization of APRIL antagonistic monoclonal antibodies for treatment of B-cell lymphomas
Blood
2011
, vol. 
117
 
25
(pg. 
6856
-
6865
)
52
Chiorazzi
 
N
Rai
 
KR
Ferrarini
 
M
Chronic lymphocytic leukemia
N Engl J Med
2005
, vol. 
352
 
8
(pg. 
804
-
815
)
53
Chu
 
CC
Catera
 
R
Hatzi
 
K
, et al. 
Chronic lymphocytic leukemia antibodies with a common stereotypic rearrangement recognize nonmuscle myosin heavy chain IIA
Blood
2008
, vol. 
112
 
13
(pg. 
5122
-
5129
)
54
Catera
 
R
Silverman
 
GJ
Hatzi
 
K
, et al. 
Chronic lymphocytic leukemia cells recognize conserved epitopes associated with apoptosis and oxidation
Mol Med
2008
, vol. 
14
 
11-12
(pg. 
665
-
674
)
55
Rosenwald
 
A
Alizadeh
 
AA
Widhopf
 
G
, et al. 
Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia
J Exp Med
2001
, vol. 
194
 
11
(pg. 
1639
-
1647
)
56
Guarini
 
A
Chiaretti
 
S
Tavolaro
 
S
, et al. 
BCR ligation induced by IgM stimulation results in gene expression and functional changes only in IgV H unmutated chronic lymphocytic leukemia (CLL) cells
Blood
2008
, vol. 
112
 
3
(pg. 
782
-
792
)
57
Burger
 
JA
O'Brien
 
S
Fowler
 
N
, et al. 
The Bruton's tyrosine kinase inhibitor, PCI-32765, is well tolerated and demonstrates promising clinical activity in chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL): an update on ongoing phase 1 studies [Abstract]
Blood
2010
, vol. 
116
 
21
pg. 
32
 
58
Furman
 
RR
Byrd
 
JC
Brown
 
JR
, et al. 
CAL-101, an isoform-selective inhibitor of phosphatidylinositol 3-kinase P110{delta}, demonstrates clinical activity and pharmacodynamic effects in patients with relapsed or refractory chronic lymphocytic leukemia [Abstract]
Blood
2010
, vol. 
116
 
21
pg. 
31
 
59
Friedberg
 
JW
Sharman
 
J
Sweetenham
 
J
, et al. 
Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia
Blood
2010
, vol. 
115
 
13
(pg. 
2578
-
2585
)
60
Honigberg
 
LA
Smith
 
AM
Sirisawad
 
M
, et al. 
The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
29
(pg. 
13075
-
13080
)
61
Lannutti
 
BJ
Meadows
 
SA
Herman
 
SE
, et al. 
CAL-101, a p110{delta} selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability
Blood
2011
, vol. 
117
 
2
(pg. 
591
-
594
)
62
Herman
 
SE
Gordon
 
AL
Hertlein
 
E
, et al. 
Bruton's tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765
Blood
2011
, vol. 
117
 
23
(pg. 
6287
-
6296
)
63
Burger
 
JA
Chemokines and chemokine receptors in chronic lymphocytic leukemia (CLL): from understanding the basics toward therapeutic targeting
Semin Cancer Biol
2010
, vol. 
20
 
6
(pg. 
424
-
430
)