In Vitro and in Vivo Targeting of Chronic Lymphocytic Leukemia Using CX-4945, a Clinical-Stage CK2-Specific Inhibitor.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2897-2897 ◽  
Author(s):  
Leila R. Martins ◽  
Paulo Lúcio ◽  
Alice Melao ◽  
Bruno A. Cardoso ◽  
Ryan Stansfield ◽  
...  

Abstract Abstract 2897 Specific inhibition of signaling elements essential for chronic lymphocytic leukemia (CLL) cell survival offers great promise for the design of improved therapies against this still incurable malignancy. The serine/threonine protein kinase CK2 is frequently upregulated in cancer, and mounting evidence implicates CK2 in tumorigenesis. Here, we evaluated whether CK2 is a valid target for therapeutic intervention in CLL, by testing the efficacy of CX-4945, a potent and highly specific orally available ATP-competitive inhibitor of CK2 that is undergoing phase I clinical trials for solid tumors and multiple myeloma. We previously showed that CK2 phosphorylates and thereby inactivates PTEN in primary T-ALL and CLL cells, leading to the hyperactivation of PI3K signaling pathway, and consequently promoting leukemia cell survival (Silva et al, JCI 2008; Martins et al, Blood 2010). Therefore, we first analyzed the impact of CX-4945 on PTEN phosphorylation and PI3K pathway activation. Incubation of CLL cells with 20 μM CX-4945 for 2h resulted in striking downregulation of PTEN phosphorylation, indicative of increased PTEN activity, and a concomitant decrease in the activity of PI3K downstream targets Akt and PKC, as determined by Akt (S473), PKCβ (S660) and PKCδ (T550) phosphorylation in both MO1043 and primary CLL cells collected and isolated to >90% purity from the peripheral blood of untreated patients. Importantly, we confirmed that Akt phosphorylation on the CK2 direct target site (S129) was also inhibited by CX-4945. Next, we evaluated the functional impact of the CK2 inhibitor on CLL cell viability. Primary CLL cells (n=11) were cultured with 10 and 20 μM CX-4945. Both drug concentrations exerted clear pro-apoptotic effects in all cases (P<0.0001 for each dose, 2-tailed paired Student's t test), as determined by Annexin V-APC/7-AAD staining. Moreover, the effect of CX-4945 was time- and dose-dependent in 4 out of 4 cases that were more thoroughly analyzed. Similar results were obtained using MEC1, MEC2, WaC3CD5, JVM3 and MO1043 cell lines whose IC50 ranged between 3.1 and 5.8μM. Notably, although co-culture with OP9 stromal cells promoted primary leukemia cell survival, it did not prevent CX-4945-mediated apoptosis of CLL cells. Most importantly, CX-4945 induced a stronger decrease in the viability of CLL cells from patients with higher percentage of malignant cells in the blood (R2=0.4176, P=0.0317, n=11, Pearson correlation), Binet stage B/C (P=0.0424, n=10, 2-tailed unpaired Student's t test) or higher plasma β2 microglobulin levels (P=0.0239, n=9). Furthermore, CLL cells with a higher proliferation rate (LDT < 12 months) were also more sensitive to CX-4945 (P=0.0007, n=11). In accordance, the need for treatment positively correlated with the sensitivity to CX-4945 (R2=0.4504, P = 0.0238). These observations suggest that treatment with CK2 inhibitors may be especially beneficial to patients with more advanced or aggressive disease. The promising results obtained in vitro prompted us to assess the impact of CX-4945 on CLL tumor development in vivo. We implanted MO1043 CLL cells subcutaneously into Swiss nude mice. At day 3, all animals presented palpable tumor masses of approximately 150 mm3, and were randomly assigned into 4 groups (n=6 per group) to receive either CX-4945 alone (75mg/kg, bid, p.o.), fludarabine alone (34mg/kg, i.p., 5 days + 2 days rest, every week), the combination of both drugs, or vehicle control. A significant delay in tumor growth was observed in all of the treatment groups when compared to the control group (P<0.0001, 2-way ANOVA). Notably, CX-4945 was as effective as fludarabine when used as a single agent, and the combination of the two drugs was significantly more effective than fludarabine alone (P=0.0375). All treatments were well tolerated as evidenced by the maintenance of body weight and the inexistence of signs of overt toxicity. Overall, our data indicate that pharmacological inhibition of CK2 is a promising therapeutic strategy in CLL that may be of special benefit to patients with aggressive and advanced stage disease. Moreover, our studies pave the way to the development of clinical trials using CX-4945 or other CK2 antagonists to manage CLL. Disclosures: Stansfield: Cylene Pharmaceuticals Inc.: Employment. Drygin:Cylene Pharmaceuticals Inc.: Employment.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3116-3116 ◽  
Author(s):  
Danelle F. James ◽  
Maryann R. Betty ◽  
Ruzbeh Mosadeghi ◽  
Thomas J. Kipps

Abstract Lenalidomide (3-(4-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione)) is an agent approved for treatment of patients with del 5q myelodysplastic syndromes and previously treated multiple myeloma. Lenalidomide has been found in early clinical trials to have potential therapeutic activity in patients with relapsed chronic lymphocytic leukemia (CLL). The mechanism(s) whereby this drug is active in CLL is unknown. In particular, studies to date have not found lenalidomide to have any direct cytotoxic activity on CLL cells in vitro. This has stimulated speculation that this agent might adversely affect the positive influence of the microenvironment on leukemia-cell survival. We and others have observed that cells found in the leukemia microenvironment can support CLL-cell survival in vitro. One such type of cells are nurse-like cells (NLC), which can differentiate from the CD14-positive blood mononuclear cells of CLL patients into large, round adherent cells that can attract and support CLL cell survival in vitro for weeks, if not longer. We evaluated the effects of lenalidomide on primary leukemia-cell survival in vitro when the CLL cells from different patients (N=21) were cultured alone or together with NLC generated as previously described [Tsukada Blood 2002]. We assessed the in-vitro activity of lenalidomide on primary CLL cells from 21 patients, in duplicate in a series of 6 experiments. Lenalidomide at concentrations of 0.1μM-200μM did not significantly impact the survival of CLL cells that were cultured alone for up to 12 days. Analysis of cell surface markers revealed increased expression of CD38 at 36 hours in 5/5 lenalidomide treated CLL samples compared with untreated cells (MFIR 5.7 +/− .86 vs. 3.4 +/− .83 p=.003). We observed sustained upregualtion of CD40 and regulation of CXCR4 in the majority of cells treated with lenalidomide. When cultured with NLC, the survival of CLL cells was comparable to or significantly higher than that of CLL cells cultured alone 62.4% vs. 51% (+/−3% SEM n=21 p [<] 0.0005). The addition of lenalidomide at concentrations of 0.1μM and greater to co-cultures of NLC and CLL cells caused specific reductions in CLL cell survival to levels similar to or lower than that of CLL cells cultured without NLC. In the presence of NLC, lenalidomide at 1μM reduced CLL cell viability compared to control (41.5% vs. 56% +/−4% p [<] 0.0005 paired student t test n=13). For most patients the levels of CLL cell viability on days 4 through 8 in the co-cultures with lenalidomide was significantly lower than those of CLL cells co-cultured with NLC in the absence of lenalidomide. As such, this study reveals that physiologic concentrations of lenalidomide might abrogate the protective influence of NLC on CLL cell survival in vitro and potentially in vivo. Conceivably, those patients who have leukemia cells displaying a high dependency on NLC for survival in vitro also might be most likely to experience a favorable clinical response to treatment with lenalidomide. This hypothesis will be tested in a prospective manner with a planned clinical trial evaluating lenalidomide for treatment of CLL through the CLL Research Consortium.


2006 ◽  
Vol 18 (2) ◽  
pp. 189
Author(s):  
A. Harvey ◽  
M. Lane ◽  
J. Thompson

Collection of embryos exposes them to a number of stresses, including light, air, and changes in temperature. Improvement of holding media to reduce the impact of handling stresses on the embryo during in vivo collection and transfer is therefore beneficial to ensure maintenance of viability following transfer. The aim of this study was to compare the effect of holding IVP-derived blastocysts at 25°C in Emcare I (ECMI, Emcare, Dallas, TX, USA) with those held in Emcare II (ECMII), a proprietry formulation designed to reduce in vitro-induced stress. In vitro-produced bovine embryos were generated using standard protocols. Blastocysts were randomly allocated to either ECMI or ECMII (ICPBio, Aukland, New Zealand) on Day 7 and were held at 25°C for a period of 24 h, after which they were cultured in Cook Bovine Blast (Cook Australia, Brisbane, Australia) supplemented with 10% fetal calf serum for 48 h. At 24 and 48 h, embryos were scored for hatching, and a cohort removed for TUNEL staining at each time point. Differences were analyzed by Student's t-test. At both 24- and 48-h culture, hatching rates tended to be higher for embryos held in ECMII than in ECMI (Table 1). The level of apoptosis at 48 h was reduced in blastocysts held in ECMII (P = 0.06). Moreover, the total cell number of hatched blastocysts at 48 h was significantly increased (1.5-fold) in those held in ECMII (P = 0.01). Results suggest that the formulation of ECMII improves the ability of IVP bovine blastocysts to re-expand and hatch following an imposed stress (25°C for 24 h). Furthermore, ECMII improves overall embryo quality through a reduction in the percentage of cells undergoing apoptosis as well as through increased cell numbers, evident 48 h following cessation of the stress. We suggest that Emcare II reduces the impact of (or increases the embryo's tolerance to and recovery from) an imposed stress, which, although severe in the present study, may provide improved outcomes following embryo transfer in field situations. Table 1. Hatching and apoptosis of blastocysts held at 25°C for 24 h in Emcare I or Emcare II This work was supported with funding by ICPBio (NZ).


Blood ◽  
2012 ◽  
Vol 119 (20) ◽  
pp. 4708-4718 ◽  
Author(s):  
Anja Troeger ◽  
Amy J. Johnson ◽  
Jenna Wood ◽  
William G. Blum ◽  
Leslie A. Andritsos ◽  
...  

Abstract Trafficking of B-cell chronic lymphocytic leukemia (CLL) cells to the bone marrow and interaction with supporting stromal cells mediates important survival and proliferation signals. Previous studies have demonstrated that deletion of Rhoh led to a delayed disease onset in a murine model of CLL. Here we assessed the impact of RhoH on homing, migration, and cell-contact dependent interactions of CLL cells. Rhoh−/− CLL cells exhibited reduced marrow homing and subsequent engraftment. In vitro migration toward the chemokines CXCL12 and CXCL13 and cell-cell interactions between Rhoh−/− CLL cells and the supporting microenvironment was reduced. In the absence of RhoH the distribution of phosphorylated focal adhesion kinase, a protein known to coordinate activation of the Rho GTPases RhoA and Rac, appeared less polarized in chemokine-stimulated Rhoh−/− CLL cells, and activation and localization of RhoA and Rac was dysregulated leading to defective integrin function. These findings in the Rhoh−/− CLL cells were subsequently demonstrated to closely resemble changes in GTPase activation observed in human CLL samples after in vitro and in vivo treatment with lenalidomide, an agent with known influence on microenvironment protection, and suggest that RhoH plays a critical role in prosurvival CLL cell-cell and cell-microenvironment interactions with this agent.


Blood ◽  
2012 ◽  
Vol 119 (5) ◽  
pp. 1182-1189 ◽  
Author(s):  
Sabine Ponader ◽  
Shih-Shih Chen ◽  
Joseph J. Buggy ◽  
Kumudha Balakrishnan ◽  
Varsha Gandhi ◽  
...  

Abstract B-cell receptor (BCR) signaling is a critical pathway in the pathogenesis of several B-cell malignancies, including chronic lymphocytic leukemia (CLL), and can be targeted by inhibitors of BCR-associated kinases, such as Bruton tyrosine kinase (Btk). PCI-32765, a selective, irreversible Btk inhibitor, is a novel, molecularly targeted agent for patients with B-cell malignancies, and is particularly active in patients with CLL. In this study, we analyzed the mechanism of action of PCI-32765 in CLL, using in vitro and in vivo models, and performed correlative studies on specimens from patients receiving therapy with PCI-32765. PCI-32765 significantly inhibited CLL cell survival, DNA synthesis, and migration in response to tissue homing chemokines (CXCL12, CXCL13). PCI-32765 also down-regulated secretion of BCR-dependent chemokines (CCL3, CCL4) by the CLL cells, both in vitro and in vivo. In an adoptive transfer TCL1 mouse model of CLL, PCI-32765 affected disease progression. In this model, PCI-32765 caused a transient early lymphocytosis, and profoundly inhibited CLL progression, as assessed by weight, development, and extent of hepatospenomegaly, and survival. Our data demonstrate that PCI-32765 effectively inhibits CLL cell migration and survival, possibly explaining some of the characteristic clinical activity of this new targeted agent.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 803-803
Author(s):  
Leila R. Martins ◽  
Paulo Lúcio ◽  
Paula Gameiro ◽  
Maria Gomes Silva ◽  
Joao T Barata

Abstract Abstract 803 Characterization of the molecular mechanisms that regulate viability of B cell chronic lymphocytic leukemia (CLL) cells may provide novel insights into the biology of this incurable malignancy and reveal prognostic markers and therapeutic targets. In particular, specific inhibition of signaling elements essential for leukemia cell survival offers great promise for the design of more efficient and selective therapies. The protein serine/threonine kinase CK2 is frequently upregulated in different cancers and mounting evidence implicates CK2 in tumorigenesis. In the present study, we evaluated whether CK2 may play a significant role in CLL. Peripheral blood mononuclear cells (PBMCs) from CLL patients (n=15) and healthy controls (n=7 ) were initially compared by Western blot densitometry analysis. CK2α, but not CK2β, was significantly upregulated in primary CLL patient samples (P=0.0003; Unpaired t-test), and CK2α expression clearly correlated with CK2 in vitro kinase activity (P value=0.0165). To test the functional significance of CK2 overexpression in CLL, we cultured PBMCs collected from leukemia patients (n=53) or purified CLL cells (n=7) in the presence of the CK2-specific small molecule inhibitors TBB and DRB. As determined by Annexin V-APC/7AAD staining and analysis of procaspase 3 cleavage, both TBB and DRB clearly promoted apoptosis of CLL cells (CD19+CD5+) in a time- and dose-dependent manner. Importantly, neither CK2 antagonist induced significant cell death in the normal T-cell population (CD19-CD3+) present in each PBMC patient sample. Consequently, the percentage of normal T-cells dramatically increased upon TBB/DRB treatment. Further, B-cells from healthy controls (n=7) were not affected by TBB or DRB, confirming the selectivity of the CK2 inhibitors towards leukemia cells. Notably, although co-culture with OP9 stromal cells promoted primary leukemia cell survival in vitro, it did not prevent apoptosis of CLL cells mediated by CK2 inhibition. The pro-apoptotic effect of TBB/DRB did not correlate with clinical parameters such as lymphocyte doubling time, ZAP-70 expression or IGHV mutational status. Interestingly, there was a significant association between the percentage of CLL cells in the peripheral blood and sensitivity to CK2 inhibition (e.g. 25μM TBB, P=0.0043), and treatment with 12.5μM TBB correlated with Binet stage (P=0.0043). We previously showed that CK2 phosphorylates and thereby inactivates PTEN in primary T-ALL cells leading to the hyperactivation of PI3K signaling pathway (Silva et al, JCI 2008). Here, we found that primary CLL samples displayed higher P-PTEN/PTEN ratios than healthy controls (P=0.0034), which correlated with CK2 expression (P=0.0064). Furthermore, treatment with TBB/DRB abrogated PTEN phosphorylation in 2 primary leukemia samples analyzed, suggesting that CK2 negatively regulates the activity of the tumor suppressor PTEN in CLL and raising the possibility that the pro-survival effect of CK2 may be dependent, at least in part, on the activation of PI3K pathway. Overall, our study suggests that CK2 plays an important role in the biology of CLL, and lays the groundwork for the inclusion of CK2 antagonists into future therapeutic options. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 113 (19) ◽  
pp. 4656-4666 ◽  
Author(s):  
David M. Lucas ◽  
Ryan B. Edwards ◽  
Gerard Lozanski ◽  
Derek A. West ◽  
Jungook D. Shin ◽  
...  

Abstract Therapeutic options for advanced B-cell acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL) are limited. Available treatments can also deplete T lymphocytes, leaving patients at risk of life-threatening infections. In the National Cancer Institute cell line screen, the structurally unique natural product silvestrol produces an unusual pattern of cytotoxicity that suggests activity in leukemia and selectivity for B cells. We investigated silvestrol efficacy using primary human B-leukemia cells, established B-leukemia cell lines, and animal models. In CLL cells, silvestrol LC50 (concentration lethal to 50%) is 6.9 nM at 72 hours. At this concentration, there is no difference in sensitivity of cells from patients with or without the del(17p13.1) abnormality. In isolated cells and whole blood, silvestrol is more cytotoxic toward B cells than T cells. Silvestrol causes early reduction in Mcl-1 expression due to translational inhibition with subsequent mitochondrial damage, as evidenced by reactive oxygen species generation and membrane depolarization. In vivo, silvestrol causes significant B-cell reduction in Eμ-Tcl-1 transgenic mice and significantly extends survival of 697 xenograft severe combined immunodeficient (SCID) mice without discernible toxicity. These data indicate silvestrol has efficacy against B cells in vitro and in vivo and identify translational inhibition as a potential therapeutic target in B-cell leukemias.


PLoS ONE ◽  
2013 ◽  
Vol 8 (10) ◽  
pp. e76607 ◽  
Author(s):  
Erin Hertlein ◽  
Kyle A. Beckwith ◽  
Gerard Lozanski ◽  
Timothy L. Chen ◽  
William H. Towns ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (20) ◽  
pp. 4223-4230 ◽  
Author(s):  
Martina Seiffert ◽  
Angela Schulz ◽  
Sibylle Ohl ◽  
Hartmut Döhner ◽  
Stephan Stilgenbauer ◽  
...  

Abstract Accumulation of leukemic cells in patients with chronic lymphocytic leukemia (CLL) is due to prolonged cell survival rather than increased proliferation. Survival of CLL cells depends on microenvironmental factors. Even though long-lived in vivo, CLL cells rapidly die by spontaneous apoptosis in vitro unless cocultured with stromal cells or their conditioned medium. In the present study, we show that survival of CLL cells is maintained in high cell density cultures, where the main prosurvival activity is delivered by monocytes. Cytokine array and enzyme-linked immunosorbent assay studies revealed increased expression of soluble CD14 by monocytes in the presence of CLL cells. The addition of recombinant soluble CD14 to primary CLL cells resulted in significantly increased cell survival rates, which were associated with higher activity nuclear factor κB. Quantification of serum levels of soluble CD14 revealed abnormally high levels of this protein in CLL patients, indicating a potential role of soluble CD14 in vivo. In summary, the presented data show that monocytes help in the survival of CLL cells by secreting soluble CD14, which induces nuclear factor κB activation in these cells, and that CLL cells actively shape their microenvironment by inducing CD14 secretion in accessory monocytes.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 588-588
Author(s):  
Davorka Messmer ◽  
Tomoyuki Endo ◽  
Bradley T. Messmer ◽  
Thomas J. Kipps

Abstract CD14+ blood mononuclear cells co-cultured with chronic lymphocytic leukemia (CLL) B cells differentiate into nurselike cells (NLCs) that in turn can support CLL-cell survival in vitro and possibly in vivo. These cells appear similar to lymphoma-associated macrophages, which were identified in secondary lymphoid tissue of patients with follicular lymphoma and appear more prevalent in patients with therapy-resistant disease. To investigate the relationship between NLC and macrophages, we performed studies on macrophages and NLCs that were induced to differentiate from CD14+ blood mononuclear cells in vitro. Consistent with prior studies, we found that NLCs express significantly higher levels of CD68 than macrophages, as assessed via cytoplasmic flow cytometry. However, Affymatrix U113A microarray analysis of gene expression by NLC, macrophages, and monocytes-derived dendritic cells (DCs) from 3 donors revealed major differences in gene expression between DCs versus macrophages or NLCs, but no major differences in gene expression profiles between NLCs and macrophages. Flow cytometric analyses of NLCs and macrophages revealed that these two cell types also shared similar expression levels of CD16, CD32, CD35, CD86, CD58, MHC-II, CD40, and CD54. However, using flow cytometry we found that NLCs (n=9) expressed significantly higher levels than macrophages of the B-cell activating factor belonging to the tumor necrosis factor family (BAFF). Deconvolution microscopy confirmed the differences in BAFF expression and also revealed that NLCs express higher levels of a proliferation-inducing ligand (APRIL) than macrophages. These are two key factors involved in promoting leukemia/lymphoma B cell survival. Moreover, NLCs maintained high-level expression of BAFF even when cultured apart from CLL cells in fresh medium. We investigated whether co-culture of differentiated macrophages with CLL cells could induce the macrophages to express high-levels of BAFF. Although such co-culture induced progressive increase in macrophages-expression of BAFF, the levels of BAFF induced after even 7 days of co-culture were lower than those noted for NLCs. We cultured CD14+ blood monocytes and CLL cells separated across a transwell membrane to determine whether a soluble factor(s) was responsible for the induction of high BAFF levels noted on NLCs. Following several days in culture, the cultured monocytes acquired expression levels of BAFF similar to those detected for NLCs. These studies indicate that monocytes can respond to a soluble factor(s) elaborated by CLL cells to assume properties similar to those of NLCs. Moreover they suggest that NLCs may be a peculiar type of terminally differentiated macrophages-like cells induced by the leukemia-cell population to have properties that promote CLL cell survival. Agents that can block the maturation of monocytes into NLCs or that inhibit the capacity of NLCs to promote leukemia-cell survival may be effective in the treatment of CLL and related lymphoid malignancies.


Sign in / Sign up

Export Citation Format

Share Document