Abstract
The tumor microenvironment (TME) plays an essential role in the development, growth, and survival of the malignant B-cell clone in chronic lymphocytic leukemia (CLL). Within the proliferation niches of lymph nodes, bone marrow, and secondary lymphoid organs, a variety of phenotypically and functionally altered cell types, including T cells, natural killer cells, monocytes/macrophages, endothelial and mesenchymal stroma cells, provide crucial survival signals, along with CLL-cellinduced suppression of antitumor immune responses. The B-cell receptor pathway plays a pivotal role in mediating the interaction between CLL cells and the TME. However, an increasing number of additional components of the multifactorial TME are being discovered. Although the majority of therapeutic strategies employed in CLL hitherto have focused on targeting the leukemic cells, emerging evidence implies that modulation of microenvironmental cells and CLL-TME interactions by novel therapeutic agents significantly affect their clinical efficacy. Thus, improving our understanding of CLL-TME interactions and how they are affected by current therapeutic agents may improve and guide treatment strategies. Identification of novel TME interactions may also pave the road for the development of novel therapeutic strategies targeting the TME. In this review, we summarize current evidence on the effects of therapeutic agents on cells and interactions within the TME. With a growing demand for improved and personalized treatment options in CLL, this review aims at inspiring future exploration of smart drug combination strategies, translational studies, and novel therapeutic targets in clinical trials.
Introduction
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by the clonal expansion of CD5+/CD19+ malignant B cells, and displays a heterogeneous pathology with chromosomal aberrations, recurrent mutations, and microenvironmental involvement.1 Although characterized by an accumulation of malignant cells in peripheral blood, CLL develops in protective niches and proliferation centers within the bone marrow, lymph nodes, the spleen and, more rarely, the liver.2 These tissues allow close interactions between malignant cells and various host cells constituting the tumor microenvironment (TME). The survival and growth of CLL cells is highly dependent on support from these surrounding microenvironmental cells that include T cells, monocytes/macrophages, endothelial and mesenchymal stroma cells, and natural killer (NK) cells.2-5 The complex crosstalk between CLL cells and these essential microenvironmental components is still poorly defined but studies have revealed how these interactions support disease progression and drug resistance.6-9 For an extensive and detailed overview of the CLL-TME constituents and interactions, we refer the reader to previously published reviews,3-5 as a complete review of the CLL TME is beyond the scope of this review. However, key components and interactions relevant for the contents of this review are briefly highlighted here.
The T-cell compartment in CLL has a complex dual role since it can exert both pro-tumor as well as anti-CLL cytotoxic activity.10 Recruited CD4+ T helper cells (Th cells) within proliferation centers provide tumor support through CD40/CD40 ligand (CD40L) co-stimulation and cytokine signaling.11,12 In the peripheral blood of patients, T-cell numbers are increased with skewing towards cytotoxic CD8+ T cells and enriched effector cell subpopulations. 13 Both CD4+ and CD8+ T-cell subpopulations exhibit functional defects including impaired immune synapse formation with antigen-presenting cells, impaired cytokine production, degranulation, and antitumor cytotoxicity. 14-16 Furthermore, T cells in CLL show increased expression of markers of chronic activation and exhaustion, such as programmed cell death protein 1 (PD-1),13,16 contributing to inhibited effector function and impaired immunological synapse formation.15,16 Patients with CLL also have elevated numbers of regulatory T cells (Treg), a subset of immunosuppressive T cells that constitute significant suppressors of antitumor T-cell responses.17 Thus, Th cells play an important supportive role in CLL, whereas the accumulation of Treg and exhausted cytolytic T cells prevent effective anti-CLL effector functions.
Similarly, myeloid cells in CLL play both tumor-supportive and immunosuppressive roles. These cells include nurse-like cells (NLC), which constitute an essential tumor-supporting component of the TME. NLC, generated in vitro, protect CLL cells from spontaneous and druginduced apoptosis, promote migration, and aid recruitment of tumor-supportive T cells.18-20 Importantly, NLC reveal a strong resemblance to tumor-associated macrophages infiltrating lymph node tissue in CLL.21 In contrast, myeloid cells with immunosuppressive properties, termed myeloid-derived suppressor cells (MDSC), accumulate in the peripheral blood of CLL patients.22 In vitro, CLL-induced MDSC suppress T-cell effector function and promote Treg differentiation.23 Thus, MDSC represent a significant immunosuppressive component within the CLL-TME.
Co-culturing CLL cells with bone marrow-derived stromal cells or endothelial cells abrogates the spontaneous apoptosis of CLL cells in vitro, highlighting the supportive role of stromal cells in the CLL-TME.24 Stromal cells mediate lymphocyte trafficking and homing, and promote CLL survival and proliferation by inducing expression of proangiogenetic and anti-apoptotic proteins.19,24 Thus, the CLL-TME constitutes a complex cellular and molecular network that contributes to tumor survival and immune suppression.
The B-cell receptor pathway is a central mechanism by which CLL cells maintain their crucial interaction with the TME. It consists of an antigen-binding transmembrane immunoglobulin connected to downstream regulators including spleen tyrosine kinase (SYK), Bruton tyrosine kinase (BTK), and phosphoinositide-3-kinase δ (PI3Kδ) (Figure 1A). B-cell receptor signaling, recently reviewed elsewhere,25 promotes proliferation, survival, and migration of the malignant clone. Stimulation of B-cell activating factor receptor (BAFF-R) by its ligand B-cell activating factor (BAFF) provided by, for example, NLC in the TME, also promotes important pro-survival and growth signals. 26 Furthermore, through direct cell-cell contact by coexpressed adhesion molecules such as lymphocyte function- associated antigen-1 (LFA-1) and intercellular adhesion molecule-1, and chemokine signaling via the CXC motif chemokine receptor (CXCR)4/CXC ligand (CXCL)12 axis, TME constituents, such as NLC and stromal cells, aid migration and homing of CLL cells into protective niches.4,18,19,24
Reciprocally, CLL cells release cytokines including interleukin (IL)-6 and IL-10,27,28 chemokines such as CCL2,12 and extracellular vesicles,4,29 through which they recruit and alter microenvironmental cells, thus inducing a tumor-supportive niche. The above highlighted CLL-TME constituents and interactions are summarized in Online Supplementary Figure S1.
The immune-subversive milieu preventing the host immune system from eliminating CLL cells also entails a state of clinical immune dysfunction, manifested as an increased risk of infections and autoimmune conditions in patients with CLL.30 Thus, the CLL-TME is not merely a “silent” support system for malignant cells, but contributes significantly to clinical presentation and disease aggressiveness.
The majority of therapeutic strategies employed hitherto have been designed to target the survival axes of CLL cells, as exemplified by the development of inhibitor drugs targeting the B-cell receptor pathway. However, as our knowledge on the mechanisms of action is expanding, there is emerging evidence that targeted agents modulate immune TME cells and interactions, which likely profoundly influences clinical responses. These effects occur both indirectly, through elimination of CLL cells and/or disruption of critical CLL-TME interaction pathways, and directly, through inhibition of targets within the specific TME cells (Figure 1B). Furthermore, some novel treatment modalities rely directly on the engagement and activation of microenvironmental cells for their anti-CLL activity (Figure 1B).
In order to improve tailored treatment options for patients with CLL, and ultimately improve the clinical course of the disease, a better understanding of how current novel therapies affect the CLL-TME is warranted. Here we review the current knowledge on how novel targeted therapies modulate CLL-TME cells and their interactions. We discuss implications for future treatment strategies and the development of combination therapy, and highlight potential novel therapeutic targets that warrant future exploration.
BTK inhibitors
The introduction of small molecule inhibitors of BTK, a TEC family kinase that plays a crucial role downstream of B-cell receptor signaling, has shifted the paradigm for CLL treatment during the past decade. Ibrutinib (PCI-32765) was the first oral covalent BTK inhibitor to be approved for CLL by the Food and Drug Administration. Secondgeneration BTK inhibitors, acalabrutinib and zanubrutinib, are currently being introduced into clinical use.31,32 BTK inhibition by ibrutinib inhibits activation-induced proliferation and induces apoptosis of CLL cells.33 However, a growing number of studies describe effects of ibrutinib on several components of the TME.
Changes in total T-cell numbers induced by ibrutinib are controversial, as studies have documented both increased and decreased total T-cell numbers in patients treated with ibrutinib.34-36 This discrepancy may be due to differences in treatment duration and disease status at the time of follow-up, as well as differences between cohorts of patients. Increased T-cell numbers were observed during the first 6 months of treatment in one study,35 while a decrease and normalization of T-cell numbers were found in studies with longer follow-up.33,34,36 This may suggest a correlation between T-cell dynamics and CLL tumor burden during ibrutinib treatment. It was previously demonstrated that T-cell receptor repertoire diversity increased in patients upon ibrutinib treatment, which correlated with disease response and lower infection rates.34 Interestingly, an increase in clonal T cells during ibrutinib treatment, which could be linked to residual CLL disease persistence and the co-occurrence of anti-CLL T-cell clones, was reported recently,37 suggesting that residual disease may maintain certain, specific anti-CLL T-cell clones. Thus, reduced tumor burden as an indirect effect of ibrutinib likely contributes significantly to normalization of the majority of the T-cell repertoire along with T-cell numbers. Ibrutinib exhibits off-target activity against IL-2-inducible T-cell kinase (ITK), a TEC kinase signaling downstream of the Tcell receptor, which plays a role in T-cell activation, cytokine release, and proliferation.38 The second-generation BTK inhibitors, acalabrutinib and zanubrutinib, have increased BTK selectivity but an insignificant inhibitory effect on ITK.39,40 In contrast to ibrutinib, treatment with acalabrutinib and zanubrutinib did not alter patients’ Tcell numbers; however, the follow-up time in these studies was limited to 6-7 months, when residual disease may still be present.35,40 Thus, further studies are warranted to clarify the potential contribution of direct ITK inhibition to the changes in T-cell numbers seen with ibrutinib.
It was also demonstrated that ibrutinib restored T-cell proliferation and degranulation,36 enhanced T-cell lytic immune synapse function,41 and reversed the exhaustion/chronic activation T-cell phenotype illustrated by PD-1 downregulation,33,35 supporting the concept that ibrutinib improves T-cell function. Similarly, treatment with acalabrutinib and zanubrutinib downregulated T-cell PD-1 expresssion.35,40 Thus, reduced exhaustion phenotypes could be due to indirect removal of tumor burden by all three BTK inhibitors. However, improved T-cell functionality may also be due to differential effects on CD4+ and CD8+ subsets, which could be linked to direct off-target activity of ibrutinib.
ITK has particular importance for Th2 T-cell polarization as well as for the development of Treg. Ibrutinib promoted Th1 polarization in a CLL mouse model,38 but this has been more challenging to detect in patients receiving therapy.35 Furthermore ex vivo ibrutinib treatment of γδ T cells from CLL patients promoted a Th1 phenotype leading to improved antitumor effector function, indicating effects due to direct off-target ITK inhibition.42 Ibrutinib treatment also reduced the fraction of Treg in CLL patients,43 while treatment with acalabrutinib did not affect Treg numbers, further indicating direct off-target ITK inhibition by ibrutinib.35,43 Reduced numbers of CD4+ IL-17 producing Th cells (Th17 cells) in ibrutinib-treated patients, as well as reduced Th17 differentiation in vitro, have also been demonstrated, recapitulating findings from ITK knockout mice.33 As for Treg, acalabrutinib did not affect Th17-cell numbers.35 However, contradictory findings, with increased Th17 T cells in patients with CLL receiving ibrutinib, have been reported.35 This is perhaps due to complex CD4+ subset changes which are related to time on therapy and prior treatment history in study cohorts. Additionally, although current data support ibrutinib-mediated direct ITK inhibition in both Th17 and Treg subsets, given their antagonizing roles,44 indirect effects on Th17 T cells due to reductions of Treg may “dominate” the direct effects, and contribute to this compartment expanding.
Inhibition of B-cell receptor signaling leading to redistribution of CLL cells from sanctuary niches into the peripheral blood is a hallmark of ibrutinib treatment,45 the mechanism of which is, in part, disruption of microenvironmental interactions. Bone marrow specimens from ibrutinib- treated patients revealed disruption of (tumor-associated) macrophage-CLL cell contacts, with macrophage cellular protrusions contracting during therapy, likely reflecting a loss of NLC pro-survival signaling.33 Ibrutinib has been shown to block BTK and downstream transcription factors within macrophages, resulting in downregulated expression of the chemokines CXCL12 and CXCL13, thus suggesting a direct effect of ibrutinib on macrophages.46 The reduced levels of these chemokines further compromised adhesion and migration of malignant B cells in vitro.46 In accordance, ibrutinib-mediated inhibition of the migratory response of CLL cells towards these chemokines was demonstrated.33,47 Thus, direct effects of ibrutinib on macrophages seem to mediate inhibited CLL-cell chemotaxis and adhesion, thereby likely contributing significantly to the clinical reduction in lymph node and spleen size, and concomitant peripheral lymphocytosis.45 Contrariwise, unfavorable effects of ibrutinib, including impaired phagocytosis in macrophages and neutrophils, and inhibited NK-cell activation and suppressed antibody-dependent cellular cytotoxicity by NK cells have been demonstrated, likely related to direct inhibition of BTK and ITK by ibrutinib in these cells. This may have important clinical implications for combination treatment with CD20 antibodies.48
Reduction of MDSC and a concomitant increase in classical monocytes were recently demonstrated in patients with CLL after 12 months of ibrutinib treatment,36 and were likely due to both direct effects of BTK inhibition in MDSC,49 and indirect effects induced through reduced tumor burden. Given their suppressive effect on T-cell function,23 a reduction of MDSC may also further contribute indirectly to improved T-cell/immune functions. Moreover, ibrutinib abrogates the adherence of vascular cell adhesion molecule-1-positive CLL cells to fibronectin on stromal cells, thereby further reducing the ability of CLL cells to remain in the protective tissue niches.47,50 Although ibrutinib produces impressive clinical results, treatment resistance is emerging,6 and residual disease remains a challenge. In vitro studies have demonstrated a protective effect of NLC in the presence of ibrutinib, thereby implying a role for NLC in contributing to residual disease and the development of ibrutinib resistance.7 Furthermore, it was demonstrated that ibrutinib-resistant subclones harboring BTK mutations promote proliferation of BTK wild-type cells during ongoing ibrutinib treatment through paracrine stimulation, further implying a role of microenvironment crosstalk in the development of resistance. 8
A number of studies seem to point toward improved clinical immune function due to the TME modulations mediated by ibrutinib.51,52 This issue, however, remains controversial, as there is still a lack of data demonstrating reduced risk of infections compared to prior ibrutinib treatment. However, studies do indicate that restoration of immune phenotypes and function establish after longterm treatment.33,34,36,51 This is in line with previous realworld data demonstrating that infectious adverse events in patients with CLL treated with ibrutinib are most frequent during the first 6 months, after which infection rates decline.53 Thus, the long-term indirect effects of ibrutinib due to reduced tumor burden and disrupted CLL-TME crosstalk may allow the various immune cell compartments to re-establish normal host immunity; however, further studies on this matter are warranted.
Continued investigation of the impact of BTK inhibitors on the TME compartments is warranted in order to provide tailored treatment strategies to improve clinical outcome (residual and progressive disease) and immune function in patients with CLL, while evading emergence of drug resistance. The most important effects of BTK inhibitors on the TME are summarized in Table 1 and illustrated in Figure 2.
PI3K inhibitors
In addition to BTK, PI3K constitutes another critical component of the B-cell receptor signaling pathway (Figure 2). Idelalisib is a selective inhibitor of PI3Kδ, the PI3K isoform generally restricted to hematopoietic cell types,54 and was the first PI3K inhibitor approved for CLL treatment. In preclinical studies, idelalisib induced caspase-dependent apoptosis of primary CLL cells and also reduced their chemokine secretion, independently of cytogenetics or IgHV mutational status.55,56 Although treatment of autologous T cells and NK cells with idelalisib does not induce apoptosis in these cells, it does decrease their production of inflammatory cytokines (IL-6, IL-10, tumor necrosis factorα [TNF-α], interferon [IFN]-γ) and activation-induced molecules (CD40L).55 These changes could potentially have effects on both pro-tumor and antitumor immune functions. In addition, idelalisib antagonizes the CLL pro-survival functions of TNF-α and CD40L.55 The effect of idelalisib on the Treg subset has been a focus of previous studies, as inactivation of PI3Kδ in mice impaired Treg-mediated immune tolerance, enhancing CD8+ T-cell mediated cytotoxic responses towards tumor cells.57 Interestingly, PI3Kδinhibition in a CLL mouse model resulted in reduced numbers and maturation of Tregs; however, this did not result in enhanced antitumor CD8+ T-cell function, likely due to concomitant direct inhibition of PI3Kδ downstream of Tcell receptor signaling.58
Similar to the effects of ibrutinib, idelalisib seems to interfere with CXCL12-mediated chemotaxis, and abrogates adhesion of CLL cells to stromal cells, suggesting an indirect mechanism through disrupting the protection of CLL cells provided by the TME.9,56 This correlates with clinical findings of reduced lymphadenopathy and splenomegaly concomitant with lymphocytosis and significantly reduced levels of CLL-related chemokines.59 It has been demonstrated that idelalisib impairs neutrophil function ex vivo,60 which together with changes in cytotoxic T-cell subsets and strong suppression of Treg, likely contribute to the increased immune-related adverse events and increased risk of infections observed upon idelalisib treatment in clinical trials.61 The next-generation PI3K inhibitor, duvelisib, a dual inhibitor of PI3K isoforms δ and γ, was recently approved for the treatment of relapsed/refractory CLL. Similar to idelalisib, treatment with duvelisib entails increased risk of immune-related toxicities and infections in patients with CLL,62 likely also due to strong direct inhibitory effects on Treg and cytotoxic T-cell effector function as demonstrated in a CLL mouse model.63 In contrast, another next-generation PI3K inhibitor, umbralisib, with dual PI3Kδ/casein kinase-1-ε(CK1ε) inhibitory activity, did not modulate Treg function, which was associated with lower toxicity in a murine model.63 Thus, the disadvantageous direct effects of idelalisib and duvelisib on T-cell subsets contributing to a risk of infections and toxicity, which have hampered their clinical usage, could potentially be mitigated with the use of umbralisib due to altered PI3K specificity. The most important effects of PI3K inhibition on the TME are summarized in Table 1 and illustrated in Figure 2.
BCL-2 inhibitors
The anti-apoptotic regulatory protein B-cell lymphoma 2 (BCL-2) is constitutively upregulated in several lymphomas including CLL, hence playing a dominant role in blocking apoptotic signaling and promoting survival in these malignancies.64 Venetoclax (ABT-199), a selective BCL-2 inhibitor, demonstrated the ability to induce rapid apoptosis in primary CLL cells in vitro and in xenograft models65 (Figure 3). In clinical trials, venetoclax alone or combined with an anti-CD20 antibody has achieved deep and durable undetectable minimal residual disease in patients with CLL.64,66 However, while leukemic cells are highly dependent on BCL-2, the dependence of nonleukemic cells on this protein seems to vary substantially. The high prevalence of grade 3/4 neutropenia among patients treated with venetoclax likely reflects the relatively marked dependency of granulopoiesis on BCL-2.67 T-cell homeostasis also depends on BCL-2, however with variable impact on different T-cell subsets. While murine naïve T cells were found to be highly dependent on BCL- 2, the protein was dispensable for memory T cells.68 Coherently, a decrease in naïve T-cell subsets and increased memory T cells have also been reported in both mice and healthy human subjects receiving venetoclax69 (Figure 3). A study on CLL patients treated with venetoclax and the CD20 antibody obinutuzumab documented decreased numbers of normal B cells, NK cells, and T cells, including Tregs, in the peripheral blood. In addition, a decrease in the exhausted/chronically activated PD1+ Tcell phenotype was observed, along with improved NKcell function, and reductions of the levels of elevated inflammatory cytokines70 (Figure 3). The authors interpreted these changes as being indirect effects due to eradication of the leukemic cells, and any direct effects on these cells by venetoclax were not investigated. Critically, the TME also appears to play a role in venetoclax resistance. In a previous study, in vitro CD40/CD40L co-stimulation strongly reduced sensitivity to venetoclax through upregulation of other anti-apoptotic proteins, such as myeloid cell leukemia 1 (MCL-1) and B-cell lymphoma extra large (BCL-XL), in CLL cells.71 The varying dependency on BCL- 2 among different microenvironmental cell types, as well as between patients, warrants further investigation, in order to optimize the advantages of targeting the apoptotic pathway in malignant cells, and utilize potential immunomodulatory effects in the immune TME while minimizing disadvantageous on-target-but-off-leukemic effects leading to adverse events.
Immunomodulatory drugs
Lenalidomide is an immunomodulatory drug (IMiD) widely used to treat multiple myeloma. Despite having no direct cytotoxicity against CLL cells in vitro,72 clinical activity in patients with CLL has been demonstrated,73,74 supporting anti-CLL immunomodulatory effects in the TME as a principle mode of action. In vitro, lenalidomide induces downregulation of CLL immune checkpoint receptors on T cells, suggesting treatment-induced immune activation or reversal of exhaustion.15 Moreover, lenalidomide treatment of autologous T cells and CLL cells triggers repair of T-cell dysfunction. This results in improved synapse formation, granzyme B- and IL-21- mediated cytotoxicity, enhanced CD8+ T-cell effector killing, and restored LFA-1-mediated T-cell motility.14,75–77 Supporting this, in vivo samples from treated patients revealed changes in the composition of the T-cell subpopulations and their cytokine production.78 Lenalidomide also affects CLL monocytes/NLC. The presence of lenalidomide impaired migration of CLL-supportive monocytes towards CCL2, CCL3, and CXCL12 in in vitro chemotaxis assays.79 The same study demonstrated downregulation of genes associated with pro-survival signals for CLL cells and impaired protective ability of NLC.79 Moreover, CLL-induced immunosuppression was reversed by lenalidomide, with improved phagocytotic activity, cytokine production, T-cell stimulatory and proliferative activity.79 Lenalidomide has produced clinical responses as monotherapy,74 in combination with rituximab or with chemotherapy,80 and as maintenance following chemotherapy.73 However, increased risk of toxicities and infections with treatment remains a concern,73 potentially reflecting potent activation of the immune TME with this class of drug. Thus, the place and dosing regimen for lenalidomide in clinical practice remain unclear. A novel option emerging for CLL therapy are next-generation cereblon E3 ligase modulators (CELMoD), with avadomide recently investigated in a preclinical study. Avadomide stimulated T-cell activation, the expression of immunostimulatory chemokines, and the formation of lytic synapses with CLL cells by triggering inflammatory IFN type I and II signaling in previously exhausted T cells from patients.81 The potential and optimal roles of IMiD and CELMoD in the context of the CLL-TME remain to be determined; however, the favorable immunomodulatory effects on the T-cell/NK-cell compartments imply a role for IMiD and CELMoD in developing novel combination treatment strategies. The most important effects of IMiD/CELMoD on the TME are summarized in Table 1 and illustrated in Figure 3.
Immune checkpoint blockade
The PD-1:PD-L1 is an immune checkpoint pathway used by tumor cells to inhibit T cells and escape immune surveillance. Thus, this pathway constitutes an attractive therapeutic target (Figure 4).82 Blocking PD-L1 in CLLtransplanted mice resulted in repressed disease development and restored T-cell immune effector functions including improved cytotoxicity, cytokine production, and immune synapse formation.83 Despite this, the sparse clinical data on immune checkpoint blockade (ICB) in CLL are disappointing. In a phase II study of the PD-1 blocking antibody drug, pembrolizumab, four out of nine patients with Richter transformation showed clinical response to treatment, whereas none of the 16 CLL patients responded.84 The clinical efficacy of ICB-based therapy correlates with upregulated levels of tumor PDL1 expression that is associated with an “inflamed” microenvironment with the presence of activated cytotoxic tumor-infiltrating T cells attempting to engage tumor cells, which can be unleashed as checkpoint inhibitory signals are abrogated.85 PD-L1 expression on CLL cells is relatively low, likely reflecting low activity of cytolytic T cells.81,82 Furthermore, the immunosuppressive state of the TME in CLL, with profoundly exhausted effector T cells that exhibit multiple functional defects, likely contributes significantly to the lack of clinical response to checkpoint inhibitor monotherapy in CLL patients. Consistent with this, a recent study of patients’ lymph node biopsies has provided evidence for a noninflamed microenvironment in CLL, incorporating low numbers of CD8+ T cells, low PD-L1 expression and profound T-cell exhaustion.81 Thus, strategies that can subvert the strong immunosuppressive pressure of the CLLTME and overcome T-cell dysfunction may be necessary to sensitize CLL to ICB immunotherapy and develop therapeutic options for CLL patients. Further research to unravel the complex immunosuppression in the CLLTME is warranted in order to develop and optimize immuno-oncology treatments.
T-cell-based therapy
Chimeric antigen receptor (CAR) T cells have emerged as a powerful therapeutic option designed to transfer high numbers of tumor-targeted effector T cells into the TME to overcome a paucity of endogenous cytolytic T cells. Briefly, autologous T cells are genetically modified to express CAR with specificity for specific tumor antigens, such as CD19 in B-cell malignancies, thus creating an adoptive T-cell-mediated cytotoxic response (Figure 4).86 CAR T cells combine the effects of T-cell and antibodymediated immune responses by triggering T-cell activation with granule exocytosis upon antigen binding.87 Despite the first successful CAR T-cell trial being reported in CLL, few clinical trials have subsequently reported efficacy of CAR T cells in CLL.88 CLL-induced T-cell dysfunction, as well as understudied lymphoid TME barriers, likely reduce the efficacy of this approach in CLL. A recent study revealed that CAR T cells from CLL patients responding well to CAR T-cell therapy expressed upregulated genes associated with T-cell memory. Furthermore, enriched T-memory subsets prior to CAR T-cell generation correlated with sustained remissions. Meanwhile, CAR T cells from non-responders had upregulated genes associated with effector T-cell differentiation, apoptosis and exhaustion, thus emphasizing that T-cell fitness is crucial for the efficacy of CAR T cells.89 Due to the multitude of (successful) treatment options for CLL currently, CAR T-cell therapy may first become a relevant option in treating multi-relapsed disease, and preliminary reports from current clinical trials of CD19-targeted CAR T-cell therapy in patients with multi-relapsed CLL show somewhat encouraging results.90 However, paradoxically, Tcell exhaustion in CLL is demonstrated to worsen with progressive disease,13 thus pointing towards a need for options that improve T-cell function prior to the manufacture of CAR T cells or during treatment. Furthermore, it was recently elucidated that CLL cells can directly impair CAR T-cell function and induce an exhausted phenotype through the release of plasma extracellular vesicles. 91 Thus, a meaningful role for CAR T-cell therapy in CLL may rely on the ability of current and/or future therapies to successfully target the TME and improve T-cell fitness in patients with CLL, prior to the CAR T-cell treatment, during preparation of the product, and after its administration.
A novel therapeutic approach that could constitute an alternative to CAR T-cell therapy is off-the-shelf bispecific CD19/CD3 or CD20/CD3 antibody treatment. Bispecific antibodies simultaneously engage CD3 on T cells and CD19 or CD20 on target B cells, and thereby redirect T cells to recognize CLL cells, facilitating synapse formation and, thus, T-cell-mediated antitumor responses (Figure 4). Preclinical studies using bispecific antibodies have demonstrated antileukemic activity against CLL cells in vitro and in xenograft models.92 Thus, bispecific antibodies may constitute a promising T-cell-based immunotherapeutic approach for CLL, alone, or in combination with TME-modulating agents that help improve T-cell function.
Developing combination strategies targeting the chronic lymphocytic leukemia – tumor microenvironment
It is becoming evident that improving clinical responses (residual and progressive disease), overcoming toxicity, infection risk, as well as drug resistance, likely require strategies aimed at reshaping the immunosubversive, protumor TME state. Our improved understanding of the direct and indirect CLL-TME modulations by novel therapeutic agents in recent years provides a unique opportunity to optimize CLL treatment with strategic drug combinations that target multiple CLL-TME interactions to achieve therapeutic synergy while controlling toxicity.
Monoclonal antibodies targeting the B-cell surface protein CD20 have been the backbone of standard chemoimmunotherapy regimes used to treat CLL for decades, although they are rarely used as a monotherapy in CLL.1 Major mechanisms of action of anti-CD20 antibodies are activation of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis, which rely on engaging the antitumor activity of NK cells and monocytes/macrophages within the immune TME.1,48,93 The direct inhibitory effects of ibrutinib on macrophage phagocytosis and NK-cell activation may therefore interfere with the therapeutic efficacy of anti-CD20 treatment. 48 Compared to ibrutinib as a single agent, adding an anti-CD20 antibody to ibrutinib was associated with faster remissions and lower levels of residual disease in a clinical trial, although it was not demonstrated that the combination improved progression-free survival.94 Thus, whether this combination is beneficial remains debatable. In contrast, combining anti-CD20 antibodies with venetoclax seems to improve the phagocytosis of CLL cells by macrophages in vitro,93 and reverse venetoclax resistance induced by TME signaling.71 Interestingly, although venetoclax plus anti-CD20 treatment produces impressive clinical responses in clinical trials, a recent retrospective study including real-world data demonstrated comparable efficacy between venetoclax as a single agent and venetoclax plus anti-CD20 combination treatment in high risk relapsed/refractory CLL patients.95 Thus, further validating prospective studies are warranted to determine whether the addition of an anti-CD20 antibody to venetoclax is truly necessary.
The addition of venetoclax to ibrutinib constitutes another approach aiming to provide improved duration and depth of remissions as well as to enable fixed-duration treatment, which has already, in part, demonstrated success in clinical trials.96 Similarly, the PI3K inhibitor, duvelisib, increases sensitivity of CLL cells to venetoclax, providing the rationale for duvelisib-venetoclax combination treatment, currently being investigated in clinical trials.97
However, the biggest challenges ahead involve finding strategic combinations that overcome T-cell dysfunction, improve the efficacy of T-cell-based therapies and ICB in CLL, and work towards curative therapy. Data from a human xenograft model support the ability of ibrutinib to enhance CAR T-cell function when administered concurrently. 98 Similarly, another murine study indicated that PI3Kδ inhibition during CAR T-cell production may have a positive effect on engraftment and antitumor activity.99 Consistently with this, a clinical pilot study recently demonstrated high response rates in relapsed/refractory CLL patients receiving ibrutinib concomitant with CD19- targeted CAR T-cell therapy, and lower toxicities compared to those in patients treated without concomitant ibrutinib.100 Furthermore, T cells from ibrutinib-treated CLL patients seem to exhibit improved in vitro anti-CLL activity combined with bispecific antibodies.101
The lack of clinical activity of anti-PD-1 monotherapy in CLL,84 has highlighted the need to incorporate ICB therapies into more powerful combinations to unleash the power of antitumor immune cells. Studies of PD- 1:PD-L1 blockade combined with ibrutinib have demonstrated enhanced CD8+ T-cell function along with improved disease control in a CLL mouse model.102 However, preliminary clinical results have indicated that coupling anti-PD-1 with ibrutinib may not increase response rates in patients.103 PI3K inhibition improved the anticancer effect of ICB through modulatory effects on MDSC in a solid cancer in vitro model,104 thus highlighting additional roles of PI3K inhibition in modulation of the TME which could be exploited. The relative expansion of memory T-cell subsets due to direct effects of venetoclax on other, more BCL-2-dependent T-cell subsets, support a role for venetoclax in combination with ICB. In a recent sold cancer murine study, venetoclax augmented the antitumor effect of anti-PD-1 and anti-PD-L1 inhibitors in vivo.69
Through their potent activation of T cells, CELMoD and IMiD could represent strong complementary treatment partners for combination (immune)therapy.14,15,73,75,76,80,81 It has been demonstrated preclinically that the CELMoD avadomide could sensitize CLL to anti-PD-1 or anti-PD-L1 immunotherapy.81 By inducing inflammatory interferon type I and II signaling in previously exhausted T cells from patients, avadomide stimulated the proliferation and release of chemokines by T cells which recruited additional CD8+ T cells, upregulated PD-L1 in the immune TME, and enhanced lytic synapse formation.81
Even more powerful combinations could include pairing ICB with CAR T cells or bispecific antibodies to increase tumor infiltrating T cells, or dual ICB combinations to overcome additional inhibitory barriers. T-cell bispecific antibodies combined with an anti-PD-L1 antibody showed enhanced antitumor efficacy compared to either given alone in a solid cancer mouse model.105 Furthermore, a recent CLL murine study demonstrated that anti-PD1 ICB combined with inhibition of the immune checkpoint receptor lymphocyte-antigen gene 3 (LAG3) was able to decrease tumor load significantly, while either as monotherapy had little effect.106
Thus, developing combination immunotherapy could represent a powerful strategy for deepening targeted drug (e.g., BTK inhibitor- and/or venetoclax)-induced responses and working towards curative therapy in CLL.
Future perspectives: novel targetable tumor microenvironment interactions
The CLL-TME constitutes a landscape of potential targetable pathways. Antibodies interrupting the CXCL12/CXCR4 interaction have demonstrated anti-CLL activity in vitro and in mouse models, and have been tested in phase I clinical trials for multiple myeloma, but have not yet been further explored in CLL.107 The BAFF/BAFF-R axis constitutes another attractive CLL-TME interaction to target. An anti-BAFF-R antibody blocked protective survival signaling in CLL cells and enhanced antibody-dependent cellular cytotoxicity in vitro, and also enhanced efficacy of ibrutinib in a CLL mouse model.108 Targeting of an IL- 10-producing CD38hi regulatory B cell-like CLL subset is also currently under investigation.27 Although aimed at CLL cells, the anti-leukemic potential here would be mediated indirectly by abrogating IL-10-mediated immunosuppression. The “don’t eat me” signal regulatory protein (SIRP)1α/CD47 axis, co-expressed by macrophages and malignant cells, respectively, in various lymphoid malignancies including CLL, constitutes a mechanism of myeloid immune tolerance. It is currently being explored as a potential target in lymphoma, and may also constitute an important macrophage-CLL interaction with targetable potential.109 Furthermore, MDSC-derived indoleamine 2,3- dioxygenase (IDO) has been explored as a target in other cancers, as an antitumor vaccination antigen.110 Mediating strong immunosuppressive effects in the CLL-TME, IDO may constitute another potentially attractive interaction to target in CLL.
In conclusion, the heterogeneous course of CLL is driven by (i) the genetic complexity and (ii) diverse and complex tissue TME interactions, including (iii) antigenic drive. The functionally and phenotypically skewed cell types within the TME not only promote CLL itself, but also compromise the induction of adequate immune responses towards developing and progressing CLL clones, as well as infectious agents. Thus, further exploration of the impact of different therapies on critical microenvironmental interactions is warranted. This review was intended to outline our current understanding of how CLL-TME interactions are influenced by current CLL therapies, with the view to encourage continued mapping of the CLL-TME for more specific future targeting of CLL-TME crosstalk. The inclusion of translational correlative studies assessing immunological and TME changes within clinical trials should inform development of novel combination therapies beyond BTK inhibitors and BCL-2 inhibitors. These include, but are not limited to, checkpoint inhibitors, T-cell based therapies, and TME modulation overcoming the inherent immune exhaustion in CLL. Such strategies together with further understanding of the TME should, eventually, lead to improved and more personalized treatment options aiming for a clinical cure with reduced burden of adverse events for patients with CLL.
Footnotes
- Received August 6, 2020
- Accepted March 31, 2021
Correspondence
Disclosures
None of the authors received funds or any other direct contribution to support the submitted work. There are no patents or copyrights relevant to this work. RS received research support from AstraZeneca, and travel grants from Abbvie outside of this work. CN received research support and/or personal consultancy fees from Abbvie, AstraZeneca, Janssen, Roche, Danish Cancer Society and Novo Nordisk Foundation (grant NNF16OC0019302) outside of this work. SJ, AR and PP have no conflicts of interest to disclose.
Contributions
RS and SJ wrote the first draft of the review under supervision of CN; the final version of the review was written by all five authors. All authors approved the final version.
References
- Hallek M. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol. 2019; 94(11):1266-1287. https://doi.org/10.1002/ajh.25595PubMedGoogle Scholar
- Herishanu Y, Pérez-Galán P, Liu D. The lymph node microenvironment promotes Bcell receptor signaling, NF-κB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood. 2011; 117(2):563-574. https://doi.org/10.1182/blood-2010-05-284984PubMedPubMed CentralGoogle Scholar
- Burger JA, Ghia P, Rosenwald A, Caligaris-Cappio F. The microenvironment in mature B-cell malignancies: a target for new treatment strategies. Blood. 2009; 114(16):3367-3375. https://doi.org/10.1182/blood-2009-06-225326PubMedPubMed CentralGoogle Scholar
- Dubois N, Crompot E, Meuleman N, Bron D, Lagneaux L, Stamatopoulos B. Importance of crosstalk between chronic lymphocytic leukemia cells and the stromal microenvironment: direct contact, soluble factors, and extracellular vesicles. Front Oncol. 2020; 10:1-19. https://doi.org/10.3389/fonc.2020.01422PubMedPubMed CentralGoogle Scholar
- Caligaris-Cappio F, Bertilaccio MTS, Scielzo C. How the microenvironment wires the natural history of chronic lymphocytic leukemia. Semin Cancer Biol. 2014; 24:43-48. https://doi.org/10.1016/j.semcancer.2013.06.010PubMedGoogle Scholar
- George B, Chowdhury SM, Hart A. Ibrutinib resistance mechanisms and treatment strategies for B-cell lymphomas. Cancers (Basel). 2020; 12(5):1328. https://doi.org/10.3390/cancers12051328PubMedPubMed CentralGoogle Scholar
- Boissard F, Fournié JJ, Quillet-Mary A, Ysebaert L, Poupot M. Nurse-like cells mediate ibrutinib resistance in chronic lymphocytic leukemia patients. Blood Cancer J. 2015; 5(10):e355. https://doi.org/10.1038/bcj.2015.74PubMedPubMed CentralGoogle Scholar
- Chen JG, Liu X, Munshi M. BTK Cys481Ser drives ibrutinib resistance via ERK1/2 and protects BTK wild-type MYD88-mutated cells by a paracrine mechanism. Blood. 2018; 131(18):2047-2059. https://doi.org/10.1182/blood-2017-10-811752PubMedGoogle Scholar
- Davids MS, Deng J, Wiestner A. Decreased mitochondrial apoptotic priming underlies stroma-mediated treatment resistance in chronic lymphocytic leukemia. Blood. 2012; 120(17):3501-3509. https://doi.org/10.1182/blood-2012-02-414060PubMedPubMed CentralGoogle Scholar
- Roessner PM, Seiffert M. T-cells in chronic lymphocytic leukemia: Guardians or drivers of disease?. Leukemia. 2020; 34(8):2012-2024. https://doi.org/10.1038/s41375-020-0873-2PubMedPubMed CentralGoogle Scholar
- Os A, Bürgler S, Ribes AP. Chronic lymphocytic leukemia cells are activated and proliferate in response to specific T helper cells. Cell Rep. 2013; 4(3):566-577. https://doi.org/10.1016/j.celrep.2013.07.011PubMedGoogle Scholar
- van Attekum MHA, van Bruggen JAC, Slinger E. CD40 signaling instructs chronic lymphocytic leukemia cells to attract monocytes via the CCR2 axis. Haematologica. 2017; 102(12):2069-2076. https://doi.org/10.3324/haematol.2016.157206PubMedPubMed CentralGoogle Scholar
- Palma M, Gentilcore G, Heimersson K. T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers. Haematologica. 2017; 102(3):562-572. https://doi.org/10.3324/haematol.2016.151100PubMedPubMed CentralGoogle Scholar
- Ramsay AG, Johnson AJ, Lee AM. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest. 2008; 118(7):2427-2437. https://doi.org/10.1172/JCI35017PubMedPubMed CentralGoogle Scholar
- 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; 120(7):1412-1421. https://doi.org/10.1182/blood-2012-02-411678PubMedPubMed CentralGoogle Scholar
- Riches JC, Davies JK, McClanahan F. T cells from CLLpatients exhibit features of Tcell exhaustion but retain capacity for cytokine production. Blood. 2013; 121(9):1612-1621. https://doi.org/10.1182/blood-2012-09-457531PubMedPubMed CentralGoogle Scholar
- Giannopoulos K, Schmitt M, Kowal M, Wlasiuk P, Bojarska-Junka A, Dmoszynska A. Characterization of regulatory T cells in patients with B-cell chronic lymphocytic leukemia. Oncol Rep. 2008; 20(3):677-682. Google Scholar
- 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; 96(8):2655-2663. https://doi.org/10.1182/blood.V96.8.2655.h8002655_2655_2663PubMedGoogle Scholar
- 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; 94(11):3658-3667. https://doi.org/10.1182/blood.V94.11.3658.423k11_3658_3667PubMedGoogle Scholar
- Borge M, Nannini PR, Morande PE. CXCL12 is a costimulator for CD4+ T cell activation and proliferation in chronic lymphocytic leukemia patients. Cancer Immunol Immunother. 2013; 62(1):113-124. https://doi.org/10.1007/s00262-012-1320-7PubMedGoogle Scholar
- Filip AA, Ciseł B, Koczkodaj D, Wa̧sik-Szczepanek E, Piersiak T, Dmoszyńska A. Circulating microenvironment of CLL: are nurse-like cells related to tumor-associated macrophages?. Blood Cells Mol Dis. 2013; 50(4):263-270. https://doi.org/10.1016/j.bcmd.2012.12.003PubMedGoogle Scholar
- Gustafson MP, Abraham RS, Lin Y. Association of an increased frequency of CD14+ HLA-DR lo/neg monocytes with decreased time to progression in chronic lymphocytic leukaemia (CLL). Br J Haematol. 2012; 156(5):674-676. https://doi.org/10.1111/j.1365-2141.2011.08902.xPubMedPubMed CentralGoogle Scholar
- Jitschin R, Braun M, Büttner M. CLLcells induce IDOhi CD14+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote TRegs. Blood. 2014; 124(5):750-760. https://doi.org/10.1182/blood-2013-12-546416PubMedGoogle Scholar
- 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; 91(7):2387-2396. https://doi.org/10.1182/blood.V91.7.2387PubMedGoogle Scholar
- Burger JA, Wiestner A. Targeting B cell receptor signalling in cancer: preclinical and clinical advances. Nat Rev Cancer. 2018; 18(3):148-167. https://doi.org/10.1038/nrc.2017.121PubMedGoogle Scholar
- Nishio M, Endo T, Tsukada N. Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF-1α. Blood. 2005; 106(3):1012-1020. https://doi.org/10.1182/blood-2004-03-0889PubMedPubMed CentralGoogle Scholar
- Manna A, Kellett T, Aulakh S. Targeting CD38 is lethal to Breg-like chronic lymphocytic leukemia cells and Tregs, but restores CD81 T-cell responses. Blood Adv. 2020; 4(10):2143-2157. https://doi.org/10.1182/bloodadvances.2019001091PubMedPubMed CentralGoogle Scholar
- Alhakeem SS, McKenna MK, Oben KZ. Chronic lymphocytic leukemia–derived IL-10 suppresses antitumor immunity. J Immunol. 2018; 200(12):4180-4189. https://doi.org/10.4049/jimmunol.1800241PubMedPubMed CentralGoogle Scholar
- Smallwood DT, Apollonio B, Willimott S. Extracellular vesicles released by CD40/IL-4-stimulated CLL cells confer altered functional properties to CD4+ T cells. Blood. 2016; 128(4):542-552. https://doi.org/10.1182/blood-2015-11-682377PubMedGoogle Scholar
- Ravandi F, O’Brien S. Immune defects in patients with chronic lymphocytic leukemia. Cancer Immunol Immunother. 2006; 55(2):197-209. https://doi.org/10.1007/s00262-005-0015-8PubMedGoogle Scholar
- Sharman JP, Egyed M, Jurczak W. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): a randomised, controlled, phase 3 trial. Lancet. 2020; 395(10232):1278-1291. https://doi.org/10.1016/S0140-6736(20)30262-2PubMedPubMed CentralGoogle Scholar
- Tam CS, Robak T, Ghia P. Efficacy and safety of zanubrutinib in patients with treatment- naive chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) with del(17p): initial results from arm C of the Sequoia (BGB-3111-304) trial. Blood. 2019; 134(Suppl_1):499. https://doi.org/10.1182/blood-2019-125394PubMedGoogle Scholar
- Niemann CU, Herman SEM, Maric I. Disruption of in vivo chronic lymphocytic leukemia tumor-microenvironment interactions by ibrutinib - findings from an investigator- initiated phase II study. Clin Cancer Res. 2016; 22(7):1572-1582. https://doi.org/10.1158/1078-0432.CCR-15-1965PubMedPubMed CentralGoogle Scholar
- Yin Q, Sivina M, Robins H. Ibrutinib therapy increases T cell repertoire diversity in patients with chronic lymphocytic leukemia. J Immunol. 2017; 198(4):1740-1747. https://doi.org/10.4049/jimmunol.1601190PubMedPubMed CentralGoogle Scholar
- Long M, Beckwith K, Do P. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest. 2017; 127(8):3052-3064. https://doi.org/10.1172/JCI89756PubMedPubMed CentralGoogle Scholar
- Solman IG, Blum LK, Hoh HY. Ibrutinib restores immune cell numbers and function in first-line and relapsed/refractory chronic lymphocytic leukemia. Leuk Res. 2020; 97:106432. https://doi.org/10.1016/j.leukres.2020.106432PubMedGoogle Scholar
- Baptista MJ, Basumallik N, Herman SEM. Ibrutinib increases the clonality of TCR repertoire in patients with chronic lymphocytic leukemia. Blood. 2018; 132(Suppl 1):238. https://doi.org/10.1182/blood-2018-99-109847PubMedGoogle Scholar
- Dubovsky JA, Beckwith KA, Natarajan G. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013; 122(15):2539-2549. https://doi.org/10.1182/blood-2013-06-507947PubMedPubMed CentralGoogle Scholar
- Herman SEM, Montraveta A, Niemann CU. The Bruton tyrosine kinase (BTK) inhibitor acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia. Clin Cancer Res. 2017; 23(11):2831-2841. https://doi.org/10.1158/1078-0432.CCR-16-0463PubMedPubMed CentralGoogle Scholar
- Zou YX, Zhu HY, Li XT. The impacts of zanubrutinib on immune cells in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma. Hematol Oncol. 2019; 37(4):392-400. https://doi.org/10.1002/hon.2667PubMedGoogle Scholar
- Papazoglou D, Lesnick CE, Wang V, Kay NE, Shanafelt TD, Ramsay AG. Ibrutinib-based therapy improves anti-tumor T cell killing function allowing effective pairing with anti-PD-L1 immunotherapy compared to traditional FCR chemoimmunotherapy; implications for therapy and correlative immune functional data from the phase III. Blood. 2018; 132(Suppl 1):236. https://doi.org/10.1182/blood-2018-99-118333PubMedGoogle Scholar
- De Weerdt I, Hofland T, Lameris R. Improving CLL Vγ9Vδ2-T-cell fitness for cellular therapy by ex vivo activation and ibrutinib. Blood. 2018; 132(21):2260-2272. https://doi.org/10.1182/blood-2017-12-822569PubMedGoogle Scholar
- Podhorecka M, Goracy A, Szymczyk A. Changes in T-cell subpopulations and cytokine network during early period of ibrutinib therapy in chronic lymphocytic leukemia patients: the significant decrease in T regulatory cells number. Oncotarget. 2017; 8(21):34661-34669. https://doi.org/10.18632/oncotarget.16148PubMedPubMed CentralGoogle Scholar
- Jadidi-Niaragh F, Ghalamfarsa G, Memarian A. Downregulation of IL-17-producing T cells is associated with regulatory T cell expansion and disease progression in chronic lymphocytic leukemia. Tumor Biol. 2013; 34(2):929-940. https://doi.org/10.1007/s13277-012-0628-4PubMedGoogle Scholar
- Herman SEM, Niemann CU, Farooqui M. Ibrutinib-induced lymphocytosis in patients with chronic lymphocytic leukemia: correlative analyses from a phase II study. Leukemia. 2014; 28(11):2188-2196. https://doi.org/10.1038/leu.2014.122PubMedPubMed CentralGoogle Scholar
- Ping L, Ding N, Shi Y. The Bruton’s tyrosine kinase inhibitor ibrutinib exerts immunomodulatory effects through regulation of tumorinfiltrating macrophages. Oncotarget. 2017; 8(24):39218-39229. https://doi.org/10.18632/oncotarget.16836PubMedPubMed CentralGoogle Scholar
- De Rooij MFM, Kuil A, Geest CR. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012; 119(11):2590-2594. https://doi.org/10.1182/blood-2011-11-390989PubMedGoogle Scholar
- Da Roit F, Engelberts PJ, Taylor RP. Ibrutinib interferes with the cell-mediated anti-tumor activities of therapeutic CD20 antibodies: implications for combination therapy. Haematologica. 2015; 100(1):77-86. https://doi.org/10.3324/haematol.2014.107011PubMedPubMed CentralGoogle Scholar
- Stiff A, Trikha P, Wesolowski R. Myeloid-derived suppressor cells express Bruton’s tyrosine kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment. Cancer Res. 2016; 76(8):2125-2136. https://doi.org/10.1158/0008-5472.CAN-15-1490PubMedPubMed CentralGoogle Scholar
- Herman SEM, Mustafa RZ, Jones J, Wong DH, Farooqui M, Wiestner A. Treatment with ibrutinib inhibits BTK- and VLA-4- dependent adhesion of chronic lymphocytic leukemia cells in vivo. Clin Cancer Res. 2015; 21(20):4642-4651. https://doi.org/10.1158/1078-0432.CCR-15-0781PubMedPubMed CentralGoogle Scholar
- Sun C, Tian X, Lee YS. Partial reconstitution of humoral immunity and fewer infections in patients with chronic lymphocytic leukemia treated with ibrutinib. Blood. 2015; 126(19):2213-2219. https://doi.org/10.1182/blood-2015-04-639203PubMedPubMed CentralGoogle Scholar
- Xiao Y, Zou P, Wang J, Song H, Zou J, Liu L. Lower phosphorylation of p38 MAPK blocks the oxidative stress-induced senescence in myeloid leukemic CD34(+)CD38 (- ) cells. J Huazhong Univ Sci Technolog Med Sci. 2012; 32(3):328-333. https://doi.org/10.1007/s11596-012-0057-zPubMedGoogle Scholar
- Aarup K, Rotbain EC, Enggaard L. Realworld outcomes for 205 patients with chronic lymphocytic leukemia treated with ibrutinib. Eur J Haematol. 2020; 105(5):646-654. https://doi.org/10.1111/ejh.13499PubMedGoogle Scholar
- Lannutti BJ, Meadows SA, Herman SEM. CAL-101, a p110δ selective phosphatidylinositol- 3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood. 2011; 117(2):591-594. https://doi.org/10.1182/blood-2010-03-275305PubMedPubMed CentralGoogle Scholar
- Herman SEM, Gordon AL, Wagner AJ. Phosphatidylinositol 3-kinase-δ inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood. 2010; 116(12):2078-2088. https://doi.org/10.1182/blood-2010-02-271171PubMedPubMed CentralGoogle Scholar
- Hoellenriegel J, Meadows SA, Sivina M. The phosphoinositide 3′-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood. 2011; 118(13):3603-3612. https://doi.org/10.1182/blood-2011-05-352492PubMedPubMed CentralGoogle Scholar
- Ali K, Soond DR, Piñeiro R. Inactivation of PI(3)K p110δ breaks regulatory T-cell-mediated immune tolerance to cancer. Nature. 2014; 510(7505):407-411. https://doi.org/10.1038/nature13444PubMedPubMed CentralGoogle Scholar
- Hanna BS, Roessner PM, Scheffold A. PI3Kδ inhibition modulates regulatory and effector T-cell differentiation and function in chronic lymphocytic leukemia. Leukemia. 2019; 33(6):1427-1438. https://doi.org/10.1038/s41375-018-0318-3PubMedGoogle Scholar
- Brown JR, Byrd JC, Coutre SE. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood. 2014; 123(22):3390-3397. https://doi.org/10.1182/blood-2013-11-535047PubMedPubMed CentralGoogle Scholar
- Alflen A, Stadler N, Aranda Lopez P. Idelalisib impairs TREM-1 mediated neutrophil inflammatory responses. Sci Rep. 2018; 8(1):1-10. https://doi.org/10.1038/s41598-018-23808-2PubMedPubMed CentralGoogle Scholar
- O’Brien SM, Lamanna N, Kipps TJ. A phase 2 study of idelalisib plus rituximab in treatment-naïve older patients with chronic lymphocytic leukemia. Blood. 2015; 126(25):2686-2694. https://doi.org/10.1182/blood-2015-03-630947PubMedPubMed CentralGoogle Scholar
- Flinn IW, Hillmen P, Montillo M. The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood. 2018; 132(23):2446-2455. https://doi.org/10.1182/blood-2018-05-850461PubMedPubMed CentralGoogle Scholar
- Maharaj K, Powers JJ, Achille A. The dual PI3Kδ/CK1ε inhibitor umbralisib exhibits unique immunomodulatory effects on CLL T cells. Blood Adv. 2020; 4(13):3072-3084. https://doi.org/10.1182/bloodadvances.2020001800PubMedPubMed CentralGoogle Scholar
- Valentin R, Grabow S, Davids MS. The rise of apoptosis: targeting apoptosis in hematologic malignancies. Blood. 2018; 132(12):1248-1264. https://doi.org/10.1182/blood-2018-02-791350PubMedGoogle Scholar
- Souers AJ, Leverson JD, Boghaert ER. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013; 19(2):202-208. https://doi.org/10.1038/nm.3048PubMedGoogle Scholar
- Roberts AW, Ma S, Kipps TJ. Efficacy of venetoclax in relapsed chronic lymphocytic leukemia is influenced by disease and response variables. Blood. 2019; 134(2):111-122. https://doi.org/10.1182/blood.2018882555PubMedPubMed CentralGoogle Scholar
- Leverson JD, Phillips DC, Mitten MJ. Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy. Sci Transl Med. 2015; 7(279):279ra40. https://doi.org/10.1126/scitranslmed.aaa4642PubMedGoogle Scholar
- Wojciechowski S, Tripathi P, Bourdeau T. Bim/Bcl-2 balance is critical for maintaining naive and memory T cell homeostasis. J Exp Med. 2007; 204(7):1665-1675. https://doi.org/10.1084/jem.20070618PubMedPubMed CentralGoogle Scholar
- Kohlhapp FJ, Haribhai D, Mathew R. Venetoclax increases intratumoral effector T cells and antitumor efficacy in combination with immune checkpoint blockade. Cancer Discov. 2020; 11(2):68-79. https://doi.org/10.1158/2159-8290.CD-19-0759PubMedGoogle Scholar
- De Weerdt I, Hofland T, De Boer R. Distinct immune composition in lymph node and peripheral blood of CLL patients is reshaped during venetoclax treatment. Blood Adv. 2019; 3(17):2642-2652. https://doi.org/10.1182/bloodadvances.2019000360PubMedPubMed CentralGoogle Scholar
- Thijssen R, Slinger E, Weller K. Resistance to ABT-199 induced by microenvironmental signals in chronic lymphocytic leukemia can be counteracted by CD20 antibodies or kinase inhibitors. Haematologica. 2015; 100(8):e302-305. https://doi.org/10.3324/haematol.2015.124560PubMedPubMed CentralGoogle Scholar
- Chanan-Khan AA, Chitta K, Ersing N. Biological effects and clinical significance of lenalidomide-induced tumour flare reaction in patients with chronic lymphocytic leukaemia: in vivo evidence of immune activation and antitumour response. Br J Haematol. 2011; 155(4):457-467. https://doi.org/10.1111/j.1365-2141.2011.08882.xPubMedPubMed CentralGoogle Scholar
- Fink AM, Bahlo J, Robrecht S. Lenalidomide maintenance after first-line therapy for high-risk chronic lymphocytic leukaemia (CLLM1): final results from a randomised, double-blind, phase 3 study. Lancet Haematol. 2017; 4(10):e475-e486. https://doi.org/10.1016/S2352-3026(17)30171-0PubMedGoogle Scholar
- Chen C, Paul H, Wang T. Long-term follow-up of a phase 2 trial of single agent lenalidomide in previously untreated patients with chronic lymphocytic leukaemia. Br J Haematol. 2014; 165(5):731-733. https://doi.org/10.1111/bjh.12785PubMedGoogle Scholar
- 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; 121(14):2704-2714. https://doi.org/10.1182/blood-2012-08-448332PubMedPubMed CentralGoogle Scholar
- Ramsay AG, Gribben JG. Immune dysfunction in chronic lymphocytic leukemia T cells and lenalidomide as an immunomodulatory drug. Haematologica. 2009; 94(9):1198-1202. https://doi.org/10.3324/haematol.2009.009274PubMedPubMed CentralGoogle Scholar
- Browning RL, Byrd WH, Gupta N. Lenalidomide induces interleukin-21 production by T cells and enhances IL21-mediated cytotoxicity in chronic lymphocytic leukemia B cells. Cancer Immunol Res. 2016; 4(8):697-707. https://doi.org/10.1158/2326-6066.CIR-15-0291PubMedPubMed CentralGoogle Scholar
- Aue G, Sun C, Liu D. Activation of Th1 immunity within the tumor microenvironment is associated with clinical response to lenalidomide in chronic lymphocytic leukemia. J Immunol. 2018; 201(7):1967-1974. https://doi.org/10.4049/jimmunol.1800570PubMedPubMed CentralGoogle Scholar
- Fiorcari S, Martinelli S, Bulgarelli J. Lenalidomide interferes with tumor-promoting properties of nurse-like cells in chronic lymphocytic leukemia. Haematologica. 2015; 100(2):253-262. https://doi.org/10.3324/haematol.2014.113217PubMedPubMed CentralGoogle Scholar
- Thompson PA, Keating MJ, Hinojosa C. Lenalidomide and rituximab in combination as initial treatment of chronic lymphocytic leukemia: initial results of a phase II study. Blood. 2014; 124(21):1988. https://doi.org/10.1182/blood.V124.21.1988.1988PubMedGoogle Scholar
- Ioannou N, Hagner PR, Stokes M. Triggering interferon signaling in T cells with avadomide sensitizes CLL to anti-PDL1/ PD-1 immunotherapy. Blood. 2020; 137(2):216-231. https://doi.org/10.1182/blood.2020006073PubMedPubMed CentralGoogle Scholar
- Brusa D, Serra S, Coscia M. The PD- 1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica. 2013; 98(6):953-963. https://doi.org/10.3324/haematol.2012.077537PubMedPubMed CentralGoogle Scholar
- McClanahan F, Hanna B, Miller S. PDL1 checkpoint blockade prevents immune dysfunction and leukemia development in a mouse model of chronic lymphocytic leukemia. Blood. 2015; 126(2):203-211. https://doi.org/10.1182/blood-2015-01-622936PubMedPubMed CentralGoogle Scholar
- Ding W, LaPlant BR, Call TG. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood. 2017; 129(26):3419-3427. https://doi.org/10.1182/blood-2017-02-765685PubMedPubMed CentralGoogle Scholar
- Yan X, Zhang S, Deng Y, Wang P, Hou Q, Xu H. Prognostic factors for checkpoint inhibitor based immunotherapy: an update with new evidences. Front Pharmacol. 2018; 9:1050. https://doi.org/10.3389/fphar.2018.01050PubMedPubMed CentralGoogle Scholar
- Kalos M, Levine BL, Porter DL. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011; 3(95):95ra73. https://doi.org/10.1126/scitranslmed.3002842PubMedPubMed CentralGoogle Scholar
- Laurin D, Marin V, Biagi E. Exploration of the lysis mechanisms of leukaemic blasts by chimaeric T-cells. J Biomed Biotechnol. 2010; 2010:234540. https://doi.org/10.1155/2010/234540PubMedPubMed CentralGoogle Scholar
- Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011; 365(8):725-733. https://doi.org/10.1056/NEJMoa1103849PubMedPubMed CentralGoogle Scholar
- Fraietta JA, Lacey SF, Orlando EJ. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018; 24(5):563-571. https://doi.org/10.1038/s41591-018-0010-1PubMedPubMed CentralGoogle Scholar
- Siddiqi T, Soumerai JD, Dorritie KA. Rapid undetectable MRD (uMRD) responses in patients with relapsed/refractory (R/R) chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) treated with lisocabtagene maraleucel (liso-cel), a CD19- directed CAR T cell product: updated results from transcend CLL 004, a phase 1/2 study including patients with high-risk disease previously treated with ibrutinib. Blood. 2019; 134(Suppl_1):503. https://doi.org/10.1182/blood-2019-127603PubMedGoogle Scholar
- Cox MJ, Lucien F, Sakemura R. Leukemic extracellular vesicles induce chimeric antigen receptor T cell dysfunction in chronic lymphocytic leukemia. Mol Ther. 2021; 29(5):1918-1932. Google Scholar
- Robinson HR, Qi J, Cook EM. A CD19/CD3 bispecific antibody for effective immunotherapy of chronic lymphocytic leukemia in the ibrutinib era. Blood. 2018; 132(5):521-532. https://doi.org/10.1182/blood-2018-02-830992PubMedPubMed CentralGoogle Scholar
- Elías EE, Almejún MB, Colado A. Autologous T-cell activation fosters ABT- 199 resistance in chronic lymphocytic leukemia: rationale for a combined therapy with SYK inhibitors and anti-CD20 monoclonal antibodies. Haematologica. 2018; 103(10):e458-e461. https://doi.org/10.3324/haematol.2018.188680PubMedPubMed CentralGoogle Scholar
- Burger JA, Sivina M, Jain N. Randomized trial of ibrutinib vs ibrutinib plus rituximab in patients with chronic lymphocytic leukemia. Blood. 2019; 133(10):1011-1019. https://doi.org/10.1182/blood-2018-10-879429PubMedPubMed CentralGoogle Scholar
- Mato AR, Roeker LE, Eyre TA. A retrospective comparison of venetoclax alone or in combination with an anti-CD20 monoclonal antibody in R/R CLL. Blood Adv. 2019; 3(10):1568-1573. https://doi.org/10.1182/bloodadvances.2019000180PubMedPubMed CentralGoogle Scholar
- Niemann CU, Levin M-D, Dubois J. Venetoclax and ibrutinib for patients with relapsed/refractory chronic lymphocytic leukemia. Blood. 2020; 137(8):1117-1120. https://doi.org/10.1182/blood.2020008608PubMedGoogle Scholar
- Patel VM, Balakrishnan K, Douglas M. Duvelisib treatment is associated with altered expression of apoptotic regulators that helps in sensitization of chronic lymphocytic leukemia cells to venetoclax (ABT- 199). Leukemia. 2017; 31(9):1872-1881. https://doi.org/10.1038/leu.2016.382PubMedPubMed CentralGoogle Scholar
- Fraietta JA, Beckwith KA, Patel PR. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016; 127(9):1117-1127. https://doi.org/10.1182/blood-2015-11-679134PubMedPubMed CentralGoogle Scholar
- Stock S, Übelhart R, Schubert ML. Idelalisib for optimized CD19-specific chimeric antigen receptor T cells in chronic lymphocytic leukemia patients. Int J Cancer. 2019; 145(5):1312-1324. https://doi.org/10.1002/ijc.32201PubMedGoogle Scholar
- Gauthier J, Hirayama AV, Purushe J. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood. 2020; 135(19):1650-1660. https://doi.org/10.1182/blood.2019002936PubMedPubMed CentralGoogle Scholar
- Long M, Williams E, Berard C. Ibrutinib treatment in CLL patients improves T cell function and blinatumomab redirected cytotoxicity. Blood. 2019; 134(Suppl_1):1049. https://doi.org/10.1182/blood-2019-125417PubMedGoogle Scholar
- Hanna BS, Yazdanparast H, Demerdash Y. Combining ibrutinib and checkpoint blockade improves CD8+ T-cell function and control of chronic lymphocytic leukemia in Eμ-TCL1 mice. Haematologica. 2021; 106(4):968-977. https://doi.org/10.3324/haematol.2019.238154PubMedPubMed CentralGoogle Scholar
- Younes A, Brody J, Carpio C. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non- Hodgkin lymphoma or chronic lymphocytic leukaemia: a phase 1/2a study. Lancet Haematol. 2019; 6(2):e67-e78. https://doi.org/10.1016/S2352-3026(18)30217-5PubMedGoogle Scholar
- Davis RJ, Moore EC, Clavijo PE. Anti- PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kδ/γ. Cancer Res. 2017; 77(10):2607-2619. https://doi.org/10.1158/0008-5472.CAN-16-2534PubMedPubMed CentralGoogle Scholar
- Sam J, Colombetti S, Fauti T. Combination of T-cell bispecific antibodies with PD-L1 checkpoint inhibition elicits superior anti-tumor activity. Front Oncol. 2020; 10:1-15. https://doi.org/10.3389/fonc.2020.575737PubMedPubMed CentralGoogle Scholar
- Wierz M, Pierson S, Guyonnet L. Dual PD1/LAG3 immune checkpoint blockade limits tumor development in a murine model of chronic lymphocytic leukemia. Blood. 2018; 131(14):1617-1621. https://doi.org/10.1182/blood-2017-06-792267PubMedPubMed CentralGoogle Scholar
- Kashyap MK, Amaya-Chanaga CI, Kumar D. Targeting the CXCR4 pathway using a novel anti-CXCR4 IgG1 antibody (PF-06747143) in chronic lymphocytic leukemia. J Hematol Oncol. 2017; 10(1):1-16. https://doi.org/10.1186/s13045-017-0435-xPubMedPubMed CentralGoogle Scholar
- McWilliams EM, Lucas CR, Chen T. Anti–BAFF-R antibody VAY-736 demonstrates promising preclinical activity in CLL and enhances effectiveness of ibrutinib. Blood Adv. 2019; 3(3):447-460. https://doi.org/10.1182/bloodadvances.2018025684PubMedPubMed CentralGoogle Scholar
- Chao MP, Alizadeh AA, Tang C. Anti- CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non- Hodgkin lymphoma. Cell. 2010; 142(5):699-713. https://doi.org/10.1016/j.cell.2010.07.044PubMedPubMed CentralGoogle Scholar
- Kjeldsen JW, Iversen TZ, Engell-Noerregaard L, Mellemgaard A, Andersen MH, Svane IM. Durable clinical responses and long-term follow-up of stage III-IV non-small-cell lung cancer (NSCLC) patients treated with IDO peptide vaccine in a phase I study - a brief research report. Front Immunol. 2018; 9:2145. https://doi.org/10.3389/fimmu.2018.02145PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.