scholarly journals Glucocorticoids and rituximab in vitro: synergistic direct antiproliferative and apoptotic effects

Blood ◽  
2002 ◽  
Vol 100 (5) ◽  
pp. 1765-1773 ◽  
Author(s):  
Andrea L. Rose ◽  
Barbara E. Smith ◽  
David G. Maloney

Rituximab, a chimeric human immunoglobulin G1(IgG1) anti-CD20 monoclonal antibody has been shown to mediate cytotoxicity in malignant B cells via several mechanisms in vitro. These include direct antiproliferative and apoptotic effects, complement-dependent cytotoxicity (CDC), and antibody-dependent cell-mediated cytotoxicity (ADCC). Glucocorticoids (GCs) are often administered in conjunction with rituximab in chemotherapeutic regimens or as premedication to reduce infusion-related symptoms. The effects of GCs on CDC and ADCC, and the direct apoptotic and antiproliferative effects of rituximab are unknown. Therefore, we evaluated these mechanisms in 9 B-cell non-Hodgkin lymphoma (B-NHL) cell lines using rituximab and GCs. Rituximab and dexamethasone induced synergistic growth inhibition in 6 B-NHL cell lines. Dexamethasone and rituximab induced significant G1 arrest in 9 of 9 cell lines. The combination of rituximab and dexamethasone resulted in supra-additive increases in phosphatidylserine exposure and hypodiploid DNA content in 5 and 3 B-NHL cell lines, respectively. CDC and ADCC were neither impaired nor enhanced when dexamethasone and rituximab were administered concurrently. However, preincubation of both effector and tumor cells with dexamethasone reduced specific lysis in ADCC assays in 4 B-NHL cell lines. Preincubation of tumor cell lines with dexamethasone significantly increased cell sensitivity to CDC in 3 B-NHL cell lines. We conclude that the addition of dexamethasone to rituximab results in supra-additive cytotoxicity with respect to its direct antiproliferative and apoptotic effects, induces a cell-dependent increased sensitivity to rituximab-induced CDC, and has minimal negative impact on ADCC when used simultaneously with rituximab.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3499-3499 ◽  
Author(s):  
Gabriel G Vega ◽  
Mario I Vega ◽  
Sara Huerta-Yepez ◽  
Ali Jazirehi ◽  
Hector Mayani ◽  
...  

Abstract Abstract 3499 Rituximab (chimeric anti-CD20 mAb) (Rtx) has been successfully used in the treatment of patients with B-cell non-Hodgkin's lymphomas (B-NHLs). The combination treatment with chemotherapy results in achieving high response rates and prolongation of survival. However, a subset of patients does not initially respond to treatment and many responding patients relapse and no longer respond to further treatments. Currently, there are no alternative therapies for resistant patients. The mechanism of resistance in vivo is not clear. However, we have explored a potential mechanism by developing in vitro several clones of Rtx-resistant (RR) variants for several B-NHL cell lines and characterized their properties. Briefly, unlike the parental wild-type, the RR clones express CD20 but no longer respond to treatment with Rtx or combination with cytotoxic drugs. Further, these clones overexpress the activity of several survival/anti-apoptotic pathways [1]. It is not known whether chemical modification of Rtx might be necessary to exert its activity and signaling on the RR clones. Hence, a recent report demonstrated that a fusion protein consisting of Rtx and human IFN-α (anti-CD20-hIFN-α) exhibited superior activity over Rtx, IFN-α, or combination of Rtx and IFN-α, and exhibited significant anti-proliferative and apoptotic effects in vitro with several B-NHL cell lines and in vivo an anti-tumor xenograft response [2]. These findings prompted us to investigate the effect of anti-CD20-hIFN-α on the RR clones. We hypothesized that anti-CD20-hIFN-α may exert an anti-proliferative and apoptotic effects on the RR clones and may also synergize when used in combination with chemotherapy. In this study, we used the B-NHL line Ramos (Burkitt) and 2F7 (AIDS-related) and their respective Ramos RR1 and 2F7 RR1 clones as models. We examined the effects of anti-CD20-hIFN-α and Rtx on the wild-type and RR clones following treatment with IgG isotype control, Rtx, anti-CD20-hIFN-α, CDDP (10 mg/ml) and Treanda® (Bendamustine) (5 mg/ml), as well as combinations. Treatment of 2F7 with single agents alone had no cytotoxic effect; however, treatment with the combination of Rtx and CDDP or Treanda® or anti-CD20-hIFN-α plus CDDP or Treanda® resulted in significant cytotoxicity. Treatment of Ramos resulted in similar findings observed with 2F7, however, the anti-CD20-hIFN-α alone was significantly cytotoxic to Ramos cells. Importantly, whereas treatment of 2F7 RR1 or Ramos RR1 with Rtx or Rtx plus CDDP or Treanda® had no cytotoxic effects (as expected), the treatment with the anti-CD20-hIFN-α alone had significant cytotoxicity and synergy was observed when used in combination with CDDP or Treanda®. In all of the above experiments, the level of cytotoxicity was a function of the antibody concentration used (range 10–30 μg/ml). The mechanism by which anti-CD20-hIFN-α signals the RR clones for cytotoxicity and sensitization was examined. Preliminary findings show that treatment of the RR clones with anti-CD20-hIFN-α inhibits the activity of p38MAPK survival pathway and also inhibits the anti-apoptotic gene products, Bcl-2/BclXL and upregulates the pro-apoptotic expression of Bax. These findings established, for the first time, that modification of Rtx by fusion with IFN-α was cytotoxic on the RR clones and synergized with chemotherapy. The findings also show, unlike Rtx that, anti-CD20-hIFN-α signals the RR cells and inhibits survival/antiapoptotic pathways leading to direct cytotoxicity and chemo-sensitization. The molecular signaling mediated by anti-CD20-hIFN-α on the cell membrane of RR cells leading to inhibition of survival pathways will be presented. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 113 (5) ◽  
pp. 1062-1070 ◽  
Author(s):  
David M. Goldenberg ◽  
Edmund A. Rossi ◽  
Rhona Stein ◽  
Thomas M. Cardillo ◽  
Myron S. Czuczman ◽  
...  

Abstract Veltuzumab is a humanized anti-CD20 monoclonal antibody with complementarity-determining regions (CDRs) identical to rituximab, except for one residue at the 101st position (Kabat numbering) in CDR3 of the variable heavy chain (VH), having aspartic acid (Asp) instead of asparagine (Asn), with framework regions of epratuzumab, a humanized anti-CD22 antibody. When compared with rituximab, veltuzumab has significantly reduced off-rates in 3 human lymphoma cell lines tested, aswell as increased complement-dependent cytotoxicity in 1 of 3 cell lines, but no other in vitro differences. Mutation studies confirmed that the differentiation of the off-rate between veltuzumab and rituximab is related to the single amino acid change in CDR3-VH. Studies of intraperitoneal and subcutaneous doses in mouse models of human lymphoma and in normal cynomolgus monkeys disclosed that low doses of veltuzumab control tumor growth or deplete circulating or sessile B cells. Low- and high-dose veltuzumab were significantly more effective in vivo than rituximab in 3 lymphoma models. These findings are consistent with activity in patients with non-Hodgkin lymphoma given low intravenous or subcutaneous doses of veltuzumab. Thus, changing Asn101 to Asp101 in CDR3-VH of rituximab is responsible for veltuzumab's lower off-rate and apparent improved potency in preclinical models that could translate into advantages in patients.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3117-3117 ◽  
Author(s):  
Cuc Davis ◽  
Emily Stepanchick ◽  
Khaja Syed ◽  
Amy Axel ◽  
Brett Hall ◽  
...  

Abstract Introduction: Ibrutinib is a first-in-class, oral, once-daily, small molecule that covalently binds to and inhibits Bruton's tyrosine kinase (BTK), an essential enzyme in the B-cell receptor signaling pathway. Ibrutinib has shown single-agent efficacy and good tolerability in patients with various B-cell malignancies, and has been approved for treatment of previously treated chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL). In addition, ibrutinib is also being evaluated in combination with several other anticancer agents, including the anti-CD20 monoclonal antibodies rituximab and ofatumumab, as well as obinutumumab (GA-101), a next-generation anti-CD20 antibody recently approved to treat CLL. A previous report suggested that the combination of ibrutinib and rituximab under certain conditions results in antagonistic effects on ADCC, a possible mechanism of action of rituximab (Kohrt HE, et al. Blood. 2014;123:1957-1960). However, clinical data in high-risk CLL in combination with rituximab showed high response rates compared with available data on the single agents (Burger JA, et al. Blood. 2012;120 (21):187). As the variance could be due to experimental conditions that do not replicate well the unique pharmacokinetics (PK; such as short half-life and lower Cmax; Advani RH, et al. JCO. 2013;31:88-94) and selectivity profile of ibrutinib under clinical dosing conditions, we modified the assay to better mimic the physiological PK profile, and further, to test these possible effects of ibrutinib in combination with the newer anti-CD20 antibody obinutumumab. Methods:Peripheral blood mononuclear cells (PBMC) isolated from different donors were used as effector cells in this study, in addition to the purified natural killer (NK) cells used previously, and were treated as follows: (1) no treatment for 24 hrs (control), (2) pretreatment with 200nM ibrutinib for 2hrs (2 hr T), (3) pretreatment with 200nM ibrutinib for 2 hours with a 24-hour washout period (2hr T/24 hr WO) which mimics the physiological PK profile of ibrutinib, (4) pretreatment with 200nM ibrutinib for 24 hours (24 hr C). ADCC assays were then conducted on target lymphoma cell lines (Ramos and Daudi) with rituximab (10 µg/ml) or obinutuzumab (10 µg/ml) alone or in combination with 200nM ibrutinib for 3 hours. Percent-specific lysis was measured using the Calcein AM assay. BTK occupancy was measured as described previously (Honigberg LA, et al. PNAS. 2010;107:13075-13080). Cytokine levels were measured using the 10-plex Proinflammatory Panel 1 from Meso Scale Discovery per recommended procedures. Results: Continuous exposure to ibrutinib reduced rituximab-mediated ADCC in the studied lymphoma cell lines. However, under physiologically-relevant conditions where cells were exposed to ibrutinib for 2 hrs and then washed off, the antagonistic effect on rituximab-mediated ADCC was minimized. Even with a 2 hr exposure, BTK was still fully occupied at 24 hrs after washoff, suggesting that the chronic effect may be due at least in part to other targets of ibrutinib (Honigberg LA, et al. PNAS. 2010;107:13075-13080) not continuously inhibited under physiological conditions. Obinutuzumab induced a higher level of ADCC than seen with rituximab in the same cell lines, and ibrutinib pretreatment had minimal effects on obinutuzumab-mediated ADCC, especially at 2 hr exposure with or without ibrutinib washout. Figure 1 shows typical data in Daudi cells, derived from a Burkitt's patient and not appreciably killed by ibrutinib at 200nM in 24 hrs. Cytokine measurements showed that interferon-gamma was reduced by chronic exposure to ibrutinib but this effect was also mitigated by 2 hr ibrutinib exposure followed by 24 hr washoff. Figure 1. ADCC Induced by Rituximab (Left) and Obinutuzumab (GA-101, Right) in Daudi Cells. 2 hr T and 24 hr C Represent Continuous Pre-Exposure to Ibrutinib (200 nm), While 2hr T/24 hr WO Represents the 2 hr Exposure to Ibrutinib Followed by 24 hr Washout Period Figure 1. ADCC Induced by Rituximab (Left) and Obinutuzumab (GA-101, Right) in Daudi Cells. 2 hr T and 24 hr C Represent Continuous Pre-Exposure to Ibrutinib (200 nm), While 2hr T/24 hr WO Represents the 2 hr Exposure to Ibrutinib Followed by 24 hr Washout Period Conclusions: These results demonstrate that, when administered under physiologically-relevant conditions in vitro, the effects of ibrutinib on anti-CD20 antibody-induced ADCC are substantially mitigated, particularly with obinutuzumab (GA-101). Disclosures Davis: Janssen: Employment. Syed:Janssen R&D: Employment. Axel:Janssen: Employment. Hall:Janssen: Employment. Sasser:Janssen R&D: Employment. Balasubramanian:Janssen R&D: Employment, Equity Ownership.


2019 ◽  
Vol 20 (19) ◽  
pp. 4740 ◽  
Author(s):  
Katrin Pansy ◽  
Julia Feichtinger ◽  
Barbara Ehall ◽  
Barbara Uhl ◽  
Miriam Sedej ◽  
...  

In tumor cells of more than 20 different cancer types, the CXCR4-CXCL12-axis is involved in multiple key processes including proliferation, survival, migration, invasion, and metastasis. Since data on this axis in diffuse large B cell lymphoma (DLBCL) are inconsistent and limited, we comprehensively studied the CXCR4-CXCL12-axis in our DLBCL cohort as well as the effects of CXCR4 antagonists on lymphoma cell lines in vitro. In DLBCL, we observed a 140-fold higher CXCR4 expression compared to non-neoplastic controls, which was associated with poor clinical outcome. In corresponding bone marrow biopsies, we observed a correlation of CXCL12 expression and lymphoma infiltration rate as well as a reduction of CXCR4 expression in remission of bone marrow involvement after treatment. Additionally, we investigated the effects of three CXCR4 antagonists in vitro. Therefore, we used AMD3100 (Plerixafor), AMD070 (Mavorixafor), and WKI, the niacin derivative of AMD070, which we synthesized. WK1 demonstrated stronger pro-apoptotic effects than AMD070 in vitro and induced expression of pro-apoptotic genes of the BCL2-family in CXCR4-positive lymphoma cell lines. Finally, WK1 treatment resulted in the reduced expression of JNK-, ERK1/2- and NF-κB/BCR-target genes. These data indicate that the CXCR4-CXCL12-axis impacts the pathogenesis of DLBCL and represents a potential therapeutic target in aggressive lymphomas.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2522-2522 ◽  
Author(s):  
Nishitha Reddy ◽  
Raymond Cruz ◽  
Francisco Hernandez-Ilizaliturri ◽  
Joy Knight ◽  
Myron S. Czuczman

Abstract Background: Lenalidomide is a potent thalidomide analogue shown to activate both the innate and adoptive immune system, inhibit angiogenesis, and modify the tumor microenvironment. While lenalidomide has received approval by the U.S. Federal Drug Administration (FDA) for the treatment of various hematological conditions, ongoing clinical trials are addressing its role in the treatment of B-cell lymphomas. There is a dire need to develop novel well-tolerated, therapies which combine various target-specific agents such as lenalidomide and monoclonal antibodies (mAbs). We previously demonstrated that lenalidomide is capable of expanding natural killer (NK) cells in a human-lymphoma-bearing SCID mouse model and improve rituximab anti-tumor activity in vivo. Methods: In our current work we studied the effects of lenalidomide on the biological activity of a panel of mAbs against various B-cell lymphomas, utilizing various rituximab-sensitive (RSCL) and rituximab-resistant cell lines (RRCL) generated in our laboratory from Raji and RL cell lines. Functional assays including antibody-dependant cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CMC) were performed to demonstrate changes in sensitivity to rituximab. RSCL and RRCL (1′105 cells/well) were exposed to either lenalidomide (5 μg/ml) or vehicle with or without mAb at a final concentration of 10μg/ml. The mAb panel consisted of two anti-CD20 mAbs: rituximab (Biogen IDEC, Inc.) and hA20, a humanized anti-CD20 mAb (Immunomedics, Inc.); an anti-CD80 mAb (galixumab, Biogen IDEC Inc.), and an anti-CD52 antibody (Alemtuzumab, Berlex Inc.). Changes in DNA synthesis and cell proliferation were determined at 24 and 48 hrs by [3H]-thymidine uptake. For ADCC/CMC studies, NHL cells were exposed to lenalidomide or vehicle for 24 hrs and then labeled with 51Cr prior to treatment with one of various mAbs (10 mg/ml) and peripheral blood mononuclear cells (Effector: Target ratio, 40:1) or human serum, respectively. 51Cr-release was measured and the percentage of lysis was calculated. Changes in antigen (CD20, CD80, and CD52) expression following in vitro exposure to lenalidomide were studied by multicolor flow cytometric analysis. Results: Concomitant in vitro exposure of various RSCL and RRCL cells to lenalidomide and either galixumab, hA20 or alemtuzumab for 24 hrs resulted in improved anti-tumor activity when compared to controls. In addition, pre-incubation of both RSCL and RRCL with lenalidomide rendered cells more susceptible to alemtuzumab-, hA20- and galixumab-mediated ADCC and CMC. No antigen modulation (i.e., upregulation) was observed following in vitro exposure of lenalidomide to NHL cell lines, suggesting an alternative mechanism involved in the improvement antitumor activity observed. Conclusions: Our data suggest that the augmented antitumor effect of lenalidomide is not limited to its combination with rituximab, but also that it augments the antiproliferative and biological activity of alemtuzumab, hA20 and galixumab. Furthermore, these interactions are observed even in our RRCL. Future studies will be directed towards evaluating whether similar activity will be seen in vivo using a human lymphoma-bearing SCID mouse model. (Supported by USPHS grant PO1-CA103985 from the National Cancer Institute.)


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2508-2508
Author(s):  
Andrei Ivanov ◽  
Mark S. Cragg ◽  
Tim M. Illidge

Abstract Radioimmunotherapy using radiolabeled anti-CD20 antibodies (mAb) is an effective new treatment in non-Hodgkin lymphoma with high response rates. However, the molecular mechanisms behind these impressive clinical responses are poorly understood. To elucidate these mechanisms we studied the signaling events evoked in a panel of lymphoma cell lines following treatment with anti-CD20 mAb alone or in combination with irradiation. In all three lymphoma cell-lines tested a synergistic cytotoxic effect was observed when the anti-CD20 mAb B1 was combined with irradiation. The additive effect seen with B1 mAb and radiation was not observed with Rituximab and could be reversed with MEK inhibitors U0126 and PD98059 as well as siRNA targeting MEK1 or 2. Moreover, addition of U0126 reversed the decrease in clonogenic survival triggered by treatment with B1 and irradiation. To further probe the mechanism of this synergistic cell death we used cell lines over-expressing BCL2 or crmA, to block mitochondrial and death receptor pathways, respectively. Although BCL2 and crmA over-expression mediated protection against radiation alone, it had no impact on the increased cytotoxicity induced by B1+irradiation. Morphological studies revealed gross vacuolization of the cytoplasm, yet relatively well preserved nuclei in cells treated with B1+irradiation. Taken together our data indicate that activation of the MAPK cascade is an important factor that contributes to the synergistic effect of anti-CD20 (B1) antibody and irradiation and provides important new insights into how this treatment may work in the clinic.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3101-3101
Author(s):  
Aditya Veldurthy ◽  
Michaela Patz ◽  
Christian P. Pallasch ◽  
Clemens M. Wendtner ◽  
Michael Hallek ◽  
...  

Abstract Introduction: Dasatinib is an ATP competitive, dual-specific Src-Abl kinase inhibitor used for the treatment of imatinib-resistant Bcr-Abl-positive leukemias. Originally it was developed from the aminothiazole scaffold as an inhibitor of Src family kinases (SFKs). Since the aberrant expression and activity of the SFK Lyn seems to enhance the survival of CLL cells, dasatinib is currently tested in clinical studies for CLL. In order to explore the anti-leukemic potential of dasatinib we tested its effects on freshly isolated CLL cells and on CLL cell lines. Methods and Results: In freshly isolated CLL cells, dasatinib showed a dose and time dependent reduction of global tyrosine phosphorylation which was paralleled by a decreased phosphorylation of the activating tyrosine residue of SFKs. The comparison of the inhibitory effect of several protein tyrosine kinase inhibitors on overall tyrosine phosphorylation in cellular lysates of freshly isolated CLL cells showed that 0.1 μM dasatinib had a more pronounced effect than 5 μM of nilotinib, or 10 μM of PP1 or imatinib. In the CLL cell lines, Mec1 and JVM-3, 0.1 μM of dasatinib appeared to interfere with survival signaling, since decreased levels of the activated phosphorylated forms of Akt, Erk1/2 and p38 were observed after exposure to dasatinib for 2 hours. In these cell lines, dasatinib treatment increased p53 protein levels and induced PARP cleavage and caspase activity. Pro-apoptotic effects of dasatinib were observed by an increase of annexin V-positive cells and a decrease of metabolic activity measured as XTT reduction, alone and in combination with fludarabine. Among six patient samples, two clones were particularly sensitive to dasatinib and fludarabine-resistant. While in these two samples the apoptosis induction by 5 μM dasatinib surpassed that by 5 μM fludarabine, the same dose of the two drugs led to similar apoptosis induction in the fludarabine-sensitive samples. Dasatinib treatment sensitized CLL cells for fludarabine-induced apoptosis. This enhancement of apoptosis induction was more pronounced in fludarabine-resistant patient samples and JVM-3 cells than in Mec1 and fludarabine-sensitive CLL cells. Conclusion: The Src-Abl kinase inhibitor dasatinib shows potent inhibitory effects on the survival of CLL cells in vitro.


2012 ◽  
Vol 30 (5_suppl) ◽  
pp. 463-463
Author(s):  
Alaaeldin Shablak

463 Background: Combination of certain immunotherapies with TKI’s has been used with success in the treatment of metastatic renal cell carcinoma (MRCC) patients. Here we evaluate the in vitro effects of HDIL-2 and pazopanib or sunitinib on the PBMCs’s function and RCC cell lines apoptosis. Methods: PBMCs isolated from healthy donors or RCC patients were treated with different HDIL-2/TKI combination and their proliferation was assessed with CFSE labelling assay. Furthermore, using CD107a degranulation or chromium release assays, the function of these treated cells was evaluated. The BRdU assay was used to examine the effects of these drugs combinations on the proliferation of different RCC cell lines. Results: While HDIL2/sunitinib combination did not inhibit the proliferation of various immune cells induced by HDIL-2 alone, combination of HDIL-2/pazopanib did, on the other hand, inhibit these cells proliferation. Interestingly, none of these combinations had a negative impact on the functional properties of these cells. Significant proportion of RCC cell lines treated with pazopanib underwent apoptosis, while the proportions of apoptotic cells post HDIL-2 or sunitinib treatment were not different from the background. Furthermore, the combination of HDIL-2/pazopanib did not inhibit the pazopanib induced RCC apoptosis. Conclusions: These preliminary data showed that combination of HDIL-2 and either pazopanib or sunitinib could recruit different anticancer mechanism that could enhance the treatment efficacy. Further assessment in this direction is warranted.


Sarcoma ◽  
2013 ◽  
Vol 2013 ◽  
pp. 1-11 ◽  
Author(s):  
Carol H. Lin ◽  
Yi Guo ◽  
Samia Ghaffar ◽  
Peter McQueen ◽  
Jonathan Pourmorady ◽  
...  

Osteosarcoma (OS) is the most common primary bone malignancy with a high propensity for local invasion and distant metastasis. Despite current multidisciplinary treatments, there has not been a drastic change in overall prognosis within the past 2 decades. Dickkopf-3 protein (Dkk-3/REIC) has been known to inhibit canonical Wnt/β-catenin pathway, and its expression has been shown to be downregulated in OS cell lines. Usingin vivoandin vitrostudies, we demonstrated that Dkk-3-transfected 143B cells inhibited tumorigenesis and metastasis in an orthotopic xenograft model of OS. Inoculation of Dkk-3-transfected 143B cell lines into nude mice showed significant decreased tumor growth and less metastatic pulmonary nodules (88.7%) compared to the control vector.In vitroexperiments examining cellular motility and viability demonstrated less anchorage-independent growth and decreased cellular motility for Dkk-3-transfected 143B and SaOS2 cell lines compared to the control vector. Downstream expressions of Met, MAPK, ALK, and S1004A were also downregulated in Dkk-3-transfected SaOS2 cells, suggesting the ability of Dkk-3 to inhibit tumorigenic potential of OS. Together, these data suggest that Dkk-3 has a negative impact on the progression of osteosarcoma. Reexpressing Dkk-3 in Dkk-3-deficient OS tumors may prove to be of benefit as a preventive or therapeutic strategy.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2943-2943 ◽  
Author(s):  
Nicholas Heaney ◽  
Francesca Pellicano ◽  
Lisa Crawford ◽  
Sandra Irvine ◽  
Tessa L. Holyoake

Abstract CML is a disease of the haemopoietic stem cell (HSC) caused by the constitutively active tyrosine kinase Bcr-Abl. The current first-line therapy for CML is imatinib (IM), a tyrosine kinase inhibitor (TKI) effective in establishing durable disease control in the majority of patients in chronic phase. However, the quiescent leukaemic HSC (Ph+HSC) population is insensitive to the apoptotic effects of this and more potent TKIs - such as dasatinib or nilotinib. These TKIs exert potent anti-proliferative effects against CML stem and progenitor cells, resulting in accumulation of quiescent cells in vitro. This Ph+HSC reservoir of disease in treated patients is now recognised to be a critical target if we wish to achieve complete eradication of residual disease. Bcr-Abl+ cells have abnormally high levels of proteasome activity and expression and in common with other cancer cells have been shown to be relatively more sensitive to the effects of proteasome inhibitors (PI) than their non-malignant counterparts. We have assessed the potential role for PI in CML using Bcr-Abl+ cell lines (K562 and Ba/F3 with mutant/wild type Bcr-Abl) and Ph+HSC obtained from leukapheresis products of patients, taken with consent, at time of diagnosis with CML. We have demonstrated apoptotic and anti-proliferative effects of PI (MG132 and bortezomib) in wild type and mutated Bcr-Abl+ cell lines (including T315I) and in Ph+HSC (n=7) by viable cell counts, thymidine incorporation and flow cytometry for annexin V and 7AAD. The apoptotic effects also occurred in the quiescent Ph+HSC fraction with an IC50 of 5–7.5nM (bortezomib)- similar to that seen in the total CD34+ cell population and lower than that previously reported for cells from multiple myeloma patients. These results were compared to parallel FACS-sorted cell populations from non-CML Ph- leukapheresis products and a therapeutic window was seen. CFSE tracking of cell division in CML samples in vitro indicated depletion of the quiescent fraction, in contrast to the accumulation of these cells demonstrated with IM, nilotinib or dasatinib, either alone or in combination. The intracellular effects of exposure of these cell populations to bortezomib were assessed by quantitatively measuring the intracellular accumulation of polyubiquitinated proteins seen following PI treatment. Interestingly no clear effect on Bcr-Abl activity was demonstrated with PI treatment of Ph+HSC (measured by immunoblotting with antibody to phosphorylated Crkl), though in Bcr-Abl+ cell lines detectable activity was reduced. Drug combination experiments (Calcusyn) were performed with bortezomib and IM or dasatinib to assess the effect of simultaneous inhibition of Bcr-Abl and the proteasome - the combination of a TKI with a PI resulted in additive effects, however no clear synergistic effects of significance were seen. Our results indicate that PI are capable of inducing CML stem cell specific apoptosis, suggesting the proteasome may be a relevant target for eradication of residual disease in CML following TKI therapy.


Sign in / Sign up

Export Citation Format

Share Document