Abstract
Aberrant activation of the Janus Kinase (JAK)/Signal Activator of Transcription (STAT) pathway has been implicated in glioblastoma (GBM) progression. To develop a therapeutic strategy to inhibit STAT-3 signaling, we have evaluated the effects of AZD1480, a pharmacological inhibitor of JAK1 and JAK2. In this study, the in vitro efficacy of AZD1480 was tested in human and murine glioma cell lines. AZD1480 treatment effectively blocks constitutive and stimulus-induced JAK1, JAK2 and STAT-3 phosphorylation in both human and murine glioma cells, and leads to a decrease in cell proliferation and induction of apoptosis. Furthermore, we utilized human xenograft GBM samples as models for the study of JAK/STAT-3 signaling in vivo, since human GBM samples propagated as xenografts in nude mice retain both the hallmark genetic alterations and the invasive phenotype seen in vivo. In these xenograft tumors, JAK2 and STAT-3 are constitutively active, but levels vary among tumors, which is consistent with the heterogeneity of GBMs. AZD1480 inhibits constitutive and stimulus-induced phosphorylation of JAK2 and STAT-3 in these GBM xenograft tumors in vitro, downstream gene expression and inhibits cell proliferation. Furthermore, AZD1480 suppresses STAT-3 activation in the glioma-initiating cell population in GBM tumors. In vivo, AZD1480 inhibits the growth of subcutaneous tumors and increases survival of mice bearing intracranial GBM tumors by inhibiting STAT-3 activity, indicating that pharmacological inhibition of the JAK/STAT-3 pathway by AZD1480 should be considered for study in the treatment of patients with GBM tumors.
Keywords: AZD1480, glioma, STAT-3, JAK1, JAK2
Introduction
Glioblastoma (GBM) is a challenging disease to treat (1, 2). Patients diagnosed with GBM have a median survival of 12–14 months, and most tumors have an aggressive rate of recurrence and resistance to existing treatments. Aberrant activation of signaling pathways has been implicated in GBM tumor progression including receptor tyrosine kinases such as EGFR and PDGF (1–4). Activation of the PI3-K pathway is also a common feature of GBM due to frequent loss of PTEN that causes dysregulated PI3-K activity and an increase in downstream Akt signaling (5). Other pathways implicated in GBM initiation and/or progression include PKC (6), MAPK (7), Wnt (8), NF-κB (9, 10), and the Notch and Hedgehog pathways (11). Constitutive activation of the Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) pathway is linked to GBM tumor promotion and maintenance by promoting cell growth while inhibiting apoptosis (12).
The JAK/STAT pathway is involved in inflammation, proliferation, and invasion/migration (13). Activation of this pathway involves binding of a cytokine to its receptor, which leads to tyrosine phosphorylation of intracellular corresponding JAK kinases. This allows for recruitment and phosphorylation of STAT transcription factors. Phosphorylated STAT proteins dimerize, translocate to the nucleus and initiate gene transcription. Cytokines of the IL-6 family, including IL-6, Oncostatin M (OSM), Leukemia Inhibitory Factor, Ciliary Neurotrophic Factor, and IL-11, are potent activators of the JAK/STAT pathway, utilizing JAK1 and JAK2, and activating predominantly STAT-3 (13). Common gene targets of STAT-3 include pro-survival molecules such as Bcl-2, Bcl-xL, Survivin, cIAP2 and VEGF (13). STAT-3 is phosphorylated on tyrosine 705 and serine 727, and phosphorylation of both residues is required for maximal STAT-3 transcriptional activity (14).
The link between inflammation and cancer has been well established, and the JAK/STAT pathway, especially STAT-3, has been implicated in multiple cancers (15, 16). STAT-3 is upregulated and aberrantly activated in many cancers including breast, colon, prostate, and GBM (17, 18), yet STAT-3 has a very low frequency of mutation (19). Aberrant activation of STAT-3 may be due to stimuli in the GBM microenvironment, such as IL-6, or by loss of negative regulators. IL-6 family members including IL-6 and OSM are upregulated in GBMs and corresponding tumor microenvironment (20). IL-6 gene amplification events occur in 40–50% of GBM patients, which is associated with decreased patient survival (21). Recently, it was demonstrated that STAT-3 is a major transcription factor responsible for the mesenchymal subtype of GBMs (22). This subtype correlates with a more malignant phenotype and poor outcome compared to other GBM subtypes (23, 24).
AZD1480, an ATP competitive inhibitor of JAK1 and JAK2, was recently shown to inhibit the growth of solid tumors including breast, ovarian and prostate (25). AZD1480 inhibited constitutive and IL-6 induced STAT-3 activation and subsequent nuclear translocation. The ability of AZD1480 to effectively limit tumor volume was attributed to inhibition of STAT-3. In this study, we sought to determine the efficacy and potential anti-tumor effects of AZD1480 in GBMs, which have not been previously studied. We demonstrate that AZD1480 effectively inhibits JAK1,2/STAT-3 signaling in two human glioma cell lines, a murine glioma cell line, and human GBM xenografts. This inhibition of STAT-3 activation leads to a decrease in glioma cell proliferation and induction of apoptosis. In vivo, AZD1480 inhibited the growth of GBM xenografts propagated subcutaneously through decreased STAT-3 signaling. More importantly, AZD1480 treated mice bearing intracranial GBM xenografts had significantly longer survival times compared to vehicle treated mice. Although future studies are necessary, this is the first report of the anti-tumor effects of AZD1480 in GBM, which demonstrate a therapeutic benefit for targeting JAK/STAT-3 signaling in GBMs.
Materials and Methods
Reagents and Cells
AZD1480 (Supplemental Fig. 1), a JAK1/2 inhibitor, was synthesized and provided by AstraZeneca (25, 26). Antibodies to phosphorylated STAT-3 (Y705 and S727), phosphorylated JAK1 (Y1022/1023), CD133 and Caspase-3 were from Cell Signaling Technologies (Beverly, MA); JAK1, JAK2, phosphorylated JAK2 (Y1007/1008), Cyclin A and Survivin from Santa Cruz (Santa Cruz, CA); STAT-3 and PARP from BD Transduction Laboratories (San Jose, CA); and GAPDH from AbCam (Cambridge, MA). Monoclonal antibodies to Bcl-2 and Bcl-xL were a generous gift of Dr. Tong Zhou (UAB). OSM, IL-6, and soluble IL-6R were purchased from R&D Systems (Minneapolis, MN). U87-MG, U251-MG and 4C8 cells were maintained as previously described (9). U251-MG cells were authenticated and are the same as the parent line of Dr. Darrell Bigner (Duke University). U87-MG cells were purchased from ATCC and are authentic and consistent with the STR profile in the ATCC database. 4C8 is a transgenic mouse line and possesses markers consistent with the strain of origin, B6D2F1. Primary astrocyte cultures from C57BL/6 mice were established as described (27).
Immunoblotting
Cells were harvested and lysed in RIPA buffer with protease inhibitors. Protein concentration was determined using the Pierce BCA Assay. Equivalent amounts of protein were run on SDS-PAGE gels, and then transferred onto nitrocellulose membranes. After blocking with 5% milk in Tris Buffered Saline (0.01% Tween) for 1 h, primary antibodies were incubated overnight at 4°C followed by 1 h with biotinylated HRP secondary antibody, and developed with chemiluminescent ECL (Pierce, Rockford, IL), as described (28).
Cell Proliferation Assay
Cells were plated in 96 well plates at a density of 1 × 104 cells/well and the WST-1 Cell Proliferation Assay (Roche) was performed as described (28).
Soft Agar Growth Assay
A bottom layer of 0.4% agarose and DMEM/F12 with 10% FBS was poured and allowed to solidify. The top layer of agarose was allowed to reach 42°C and 7.5 × 103 U251-MG cells were added to the agarose/media solution and poured onto the bottom layer. Appropriate concentrations of AZD1480 were added to both agarose/media layers. Cells were incubated at 37°C for 4 weeks to form colonies followed by staining with 0.005% crystal violet. The numbers of colonies were imaged and quantified using the Gel Dock imager and Quantity One Software (BioRad).
Xenograft GBM Tumors
Human GBM xenograft tumors were maintained by the UAB Brain Tumor Core Facility with the approval (APN # 100908862) of the UAB Institutional Animal Care and Use Committee. Human GBM xenografts were analyzed by the Heflin Genomics Core Facility using the Applied Biosystems AmpF1STR system to screen 15 different STR markers, and determined to have identical STR patterns to that of the original patient's tumor from which they were derived. Xenograft tumors were dissociated into single cells for brief cell culture analysis, snap frozen for protein isolation and immunoblotting, injected subcutaneously in the flank, or injected intracranially. Female athymic nude mice (6–8 weeks old) were used for all experiments. Flank tumors were removed, washed with PBS, minced, and disaggregated. Cells were passed through a 40 µm filter and plated in Neurobasal media with FBS (10%), Amphotericin (1%), B27 Supplement, Gentamycin (0.25%), L-glutamine (260 mM), EGF (10 ng/ml), and FGF (10 ng/ml) and cultured as spheroids in suspension (29). Xenograft tumor cells were separated based on cell surface CD133 separation using the CD133 MicroBead kit (Miltenyi Biotec, Auburn, CA). Populations were verified by immunoblotting for CD133. Xenograft flank tumors were removed and snap frozen in liquid nitrogen and lysed in RIPA lysis buffer with protease inhibitors using a tissue homogenizer, and 30 µg of protein was immunoblotted. For subcutaneously injected tumor experiments, xenograft tumors were roughly disaggregated and minced. Approximately 100 (xenograft X1066) or 200 µl (xenograft X1046) of tumor slurry was injected subcutaneously into the flanks of athymic nude mice. Tumor volume was measured using calipers and calculated using the following equation: v = (0.5 × longest diameter × shortest diameter2). On day 6, mice were randomized to vehicle control or AZD1480. Treatment was administered intraperitoneally (IP) twice a day (6–8 h apart) at 30 mg/kg per dose in sterile water. Dosing schedule included continual twice daily IP injections for the duration of the experiment. Mice were euthanized and tumors excised, divided, and snap frozen for analysis or formalin fixed and paraffin embedded. For intracranial injection, xenograft tumors were disaggregated into single cells, and approximately 5 × 105 cells in 5 µl of methylcellulose were injected 2 mm anterior and 1 mm lateral to the bregma at a depth of 2 mm over 2 min for adequate perfusion. Tumors were allowed to establish for 5 days before beginning once daily oral gavage treatment of AZD1480 (50 mg/kg per mouse) in methylcellulose or vehicle on day 6. Treatment schedule consisted of 5 days of treatment followed by 2 days of rest for a total of 3 weeks. All mice were euthanized at moribund.
Phosphorylated JAK2 ELISA Assay
Approximately 65 µg of lysates from snap frozen xenograft samples were analyzed for phosphorylated JAK2 levels using the JAK2 [pYpY1007/1008] ELISA (Invitrogen).
Annexin V/PI Staining
U251-MG cells were stained with Annexin V and propidium iodide using Clontech (Mountain View, CA) ApoAlert Annexin V-FITC Apoptosis Kit, and examined by flow cytometry. The percentage of Annexin V-positive and propidium iodide-positive cells was determined by FlowJo 7.5.5 software.
Quantitative RT-PCR
Total RNA was isolated using TRIzol® (Invitrogen), and approximately 1 µg of RNA per sample was used to generate cDNA by reverse transcription for PCR. Pre-designed Taqman primers (Applied Biosystems) were used to obtain quantitative PCR results using the Applied Biosytems StepOnePlus Real-Time PCR System Thermal Cycling Block and corresponding software analysis for data quantification (StepOneSoftware v2.1, Applied Biosystems). The following Taqman primers and the corresponding Gene Ref # were used: human c-Myc (Hs99999003_m1), human IL-6 (Hs00174131_m1) and human SOCS-3 (Hs02330328_s1). Eukaryotic 18s rRNA (Hs99999901_s1) was used as an endogenous control.
Statistical Analysis
Student’s t-test and Mann-Whitney Rank Sum tests were performed for comparison of two values, ANOVA analysis was performed on appropriate multi-variable analyses using the Bonferonni test, and the Log-Rank test was used for Kaplan-Meier survival curves. p<0.05 was considered statistically significant.
Results
AZD1480 inhibits constitutive STAT-3 and JAK2 activation in glioma cells
We sought to determine the inhibitory effect of AZD1480 on JAK/STAT-3 signaling in GBM tumor cells and potential anti-tumor effects. Two human glioma cell lines (U251-MG and U87-MG) as well as a murine glioma cell line (4C8) that all exhibit constitutive STAT-3 activation were used to determine the effects of AZD1480. Treatment of glioma cells with AZD1480 at 1 µM blocked constitutive STAT-3 and JAK2 phosphorylation in all three glioma cell lines beginning as early as 30 min and lasting for at least 16 h (Fig. 1A). Comparable results were observed using 0.5 µM AZD1480 (data not shown). This demonstrates that AZD1480 inhibits constitutive activation of STAT-3 in GBM cell lines.
Figure 1. AZD1480 Inhibits JAK/STAT-3 Signaling in Glioma Cells.
A, Cells were treated with AZD1480 for the indicated times, lysed and immunoblotted with the indicated antibodies. B, Cells were treated with AZD1480 for the indicated times, and the WST-1 cell proliferation assay performed. The 48 and 72 h time points were statistically significant. Data represent mean ± SD, replicates of three (**, p<0.001; ANOVA). C, U251-MG cells were treated with AZD1480 for 48 h followed by Annexin V/PI staining and examined by flow cytometry. Data represent mean ± SD, replicates of three (*, p<0.05; **, p<0.001; ANOVA). Representative of two experiments. D, U251-MG cells were treated with AZD1480 for the indicated times, lysed and immunoblotted with the indicated antibodies. E, U251-MG cells were plated in 0.4% soft agar without or with AZD1480. Data represent mean ± SD (**, p<0.001; ANOVA). Representative of three experiments.
AZD1480 treatment elicits functional anti-tumor effects in glioma cells
Inhibition of STAT-3 signaling can decrease proliferation and induce apoptosis of glioma cells (30). U251-MG and 4C8 glioma cells were treated with AZD1480, which led to an inhibition of proliferation at a concentration of 10 µM (Fig. 1B). This was also demonstrated using the U87-MG cell line (data not shown). More importantly, we evaluated the ability of AZD1480 to inhibit proliferation of murine primary astrocytes and found no inhibitory effect at either a 1 or 10 µM dose (Fig. 1B). This suggests that the functional effect of AZD1480 is specific to tumor cells without affecting normal glial cells. U251-MG cells were treated with AZD1480 (1 and 10 µM) for 48 h, stained with Annexin V and PI and analyzed by flow cytometry. AZD1480 induced apoptosis in a dose-dependent manner as seen by the increase in the percentage of Annexin V/PI positivity (Fig. 1C). The ability of AZD1480 to induce cell death was also evaluated by immunoblotting for the presence of cleaved poly (ADP-ribose) polymerase (PARP). Treatment with AZD1480 induced the cleavage of PARP (PARPc) at 24 h, indicating induction of cell death (Fig. 1D). A common characteristic of transformed or malignant cells is the ability to grow in soft agar (31). We therefore determined the ability of AZD1480 to affect U251-MG growth as colonies in soft agar. Cells were plated in 0.4% agarose with media in the absence or presence of AZD1480 and colonies were stained and counted after 4 weeks. In a dose-dependent manner, AZD1480 prevented glioma cells from forming colonies (Fig. 1E).
AZD1480 prevents stimulus-induced phosphorylation of STAT-3 and downstream gene transcription
Cytokines present in the tumor microenvironment contribute to the malignancy and continual circuitry maintaining tumor growth and proliferation (16). Two members of the IL-6 family, OSM and IL-6, were used to activate JAK1,2/STAT-3 in glioma cell lines. AZD1480 prevented OSM-induced activation of JAK1,2/STAT-3 in a dose-dependent manner in all three glioma cell lines (Fig. 2A). Due to the greatly enhanced phosphorylation of STAT-3 following OSM stimulation, we have provided an appropriately exposed blot revealing the constitutive STAT-3 phosphorylation (Supplemental Fig. 2A). This inhibition was also observed following IL-6 stimulation (Supplemental Fig. 2B). To determine if inhibition of STAT-3 phosphorylation correlated with inhibition of downstream gene expression, we tested the effect of AZD1480 on three targets of STAT-3: SOCS-3, c-Myc, and IL-6. Upon OSM stimulation, AZD1480 significantly prevented OSM-induced expression of SOCS-3, c-Myc, and IL-6 mRNA as shown by quantitative RT-PCR (Fig. 2B). AZD1480 inhibition of STAT-3 target genes was also verified using IL-6 as a stimulus (data not shown). We also tested the ability of AZD1480 to inhibit the NF-κB pathway, as a selectivity control. U87-MG glioma cells were incubated with AZD1480 (1 µM) for 2 h followed by treatment with TNF-α. Pre-treatment with AZD1480 does not inhibit TNF-α-induced NF-κB p65 phosphorylation or expression of IL-8, a NF-κB driven gene (Supplemental Figs. 2C&D), supporting the absence of pleiotropic effects of AZD1480 on signaling pathways in glioma cells.
Figure 2. AZD1480 Inhibits Stimulus-induced Phosphorylation of JAK1, JAK2 and STAT-3 and Prevents Downstream Gene Transcription.
A, Cells were treated with the indicated doses of AZD1480 for 2 h prior to a 15 min stimulation with OSM (1 ng/ml). Cells were lysed and immunoblotted with the indicated antibodies. B, U251-MG cells were treated with 1 µM AZD1480 for 2 h prior to stimulation with OSM (1 ng/ml) for 1 h. Quantitative RT-PCR was performed as described in the Methods. Data represent mean ± SD, replicates of three (**, p<0.001, Student’s t-test). Representative of two experiments.
Human xenograft GBM tumors exhibit constitutive JAK2/STAT-3 activation
Human GBM xenograft tumors propagated in the flank of athymic nude mice retain the hallmark mutations seen in GBM (29). We tested several xenografts for activation of JAK2/STAT-3 signaling, and found that STAT-3 is phosphorylated on both tyrosine and serine residues in all xenograft samples tested (Fig. 3A). We also analyzed the levels of phosphorylated JAK2 by ELISA and found it to be activated as well (Fig. 3B). As expected, the levels of activation vary among tumors, which is also similar to human GBM heterogeneity (29). This is the first report of activated JAK2/STAT-3 in human GBM xenografts. The xenografts have been further analyzed for the following parameters: EGFR amplification/mutation, NF-κB status, molecular subtype, and % CD133+ cells (Supplemental Table I). EGFR amplification varied amongst the xenograft tumors, while all had activated NF-κB, as assessed by immunoblotting for serine 276 phosphorylated p65 (data not shown). Important information has emerged regarding the identification and characterization of four subtypes of GBMs: Classical, Mesenchymal, Proneural, and Neural (24). Several of the xenografts studied have been analyzed for their genetic signatures, and have been classified as Proneural (GBM15, GBM12), Classical (GBM6), and Mesenchymal (GBM10) (Supplemental Table I). Lastly, the proportion of glioma-initiating cells (GIC), as assessed by staining for CD133 positive cells is shown (Supplemental Table I). These results reveal a striking heterogeneity in the percentage of CD133 positive cells in the xenografts. Based on our initial profiling results of JAK2/STAT-3 status among the GBM xenografts, we selected X1066, X1016, and X1046 that display high levels of activated STAT-3 to more extensively evaluate the anti-tumor role of AZD1480.
Figure 3. Human Xenograft GBM Tumors Exhibit Constitutive JAK2/STAT-3 Activation.
A&B, Fresh snap frozen xenograft tumor samples were lysed and immunoblotted with the indicated antibodies (A) or analyzed for phosphorylated JAK2 levels using ELISA (B).
We next determined the ability of AZD1480 to affect JAK2/STAT-3 signaling in the GBM xenografts. AZD1480 effectively blocks constitutive STAT-3 and OSM-induced JAK1,2/STAT-3 signaling in X1066 xenograft tumor cells (Fig. 4A). Constitutive STAT-3 phosphorylation was inhibited with 1 µM AZD1480 as early as 0.5 h and as little as 0.5 µM inhibited OSM-induced STAT-3 phosphorylation. Inhibition of constitutive and OSM-induced STAT-3 activation was confirmed in Xenografts X1046 and X1016, and also by utilizing IL-6 as a stimulus (data not shown). AZD1480 prevented OSM-induced transcription of the STAT-3 target genes SOCS-3, c-Myc, and IL-6 (Fig. 4B). Xenograft X1016 tumor cell proliferation in cell culture was also inhibited by 10 µM AZD1480 (Fig. 4C). These experiments validate AZD1480 as an effective inhibitor of JAK/STAT-3 signaling in human GBM xenografts. There have been reports of STAT-3 activation in GICs (32, 33). Xenograft X1066 was separated based on cell surface CD133 expression. We found that AZD1480 inhibited constitutive and OSM-induced STAT-3 phosphorylation in both CD133 negative and CD133 positive cell populations (Fig. 4D). This demonstrates the potential for AZD1480 to inhibit STAT-3 activation not only in resident tumor cells, but also in the GIC population in GBMs.
Figure 4. AZD1480 Inhibits JAK/STAT-3 Signaling in Human Glioblastoma Xenograft Tumors In Vitro.
A, Xenograft X1066 was disaggregated into single cells and treated with AZD1480 for the indicated times, or treated with the indicated doses for 2 h prior to stimulation with OSM (1 ng/ml) for 15 min. Cells were lysed and immunoblotted with the indicated antibodies. B, Xenograft X1016 cells were disaggregated into single cells, treated with 1 µM AZD1480 for 2 h prior to stimulation with OSM (1 ng/ml) for 1 h, and quantitative RT-PCR performed. Data represent mean ± SD, replicates of three (*, p<0.05; **, p<0.01 Student’s t-test). C, Xenograft X1016 was disaggregated into single cells, treated with AZD1480 for the indicated times, and the WST-1 cell proliferation assay was performed. The 48 and 72 h time points were statistically significant. Data represent mean ± SD, replicates of three (*, p< 0.01; **, p<0.001 ANOVA). D, Xenograft X1066 was disaggregated into single cells and separated based on cell surface expression of CD133. Separated cells were then treated with AZD1480 (1 µM) for 2 h followed by treatment with OSM (10 ng/ml) for 15 min, lysed and immunoblotted with the indicated antibodies.
Treatment with AZD1480 inhibits GBM tumor growth in vivo
Since the overall goal is to develop a potential therapeutic agent for GBM patients, we evaluated the ability of AZD1480 to inhibit glioma growth in vivo. We first tested AZD1480 using a subcutaneously implanted xenograft model. Xenograft X1046 was injected subcutaneously into athymic nude mice, and starting at day 6, mice received twice daily IP injections of AZD1480 (30 mg/kg per dose) or vehicle control for a total of 3 weeks. At day 29 all mice were euthanized and tumors removed for analysis. AZD1480 significantly inhibited subcutaneous tumor growth compared to vehicle treated mice (Fig. 5A). No significant weight loss or decrease in the total number of red blood cells was observed during AZD1480 treatment (data not shown). Tumors were analyzed by immunoblotting for effectiveness of AZD1480 on inhibition of STAT-3 phosphorylation. All tumors treated with AZD1480 had little or no STAT-3 tyrosine or serine phosphorylation compared to control treated tumors (Fig. 5B). The levels of phosphorylated JAK2 also appear slightly decreased in AZD1480 treated tumors. We also observed a decrease in several growth promoting proteins including Cyclin A, Bcl-2 and Survivin in the flank tumors treated with AZD1480, while Bcl-XL expression was not affected (Fig. 5C). This suggests that AZD1480 inhibition of tumor growth can be attributed to an inhibition of STAT-3 activity. Following the same protocol, we verified the inhibition of tumor growth by AZD1480 using another xenograft tumor, X1066 (Fig. 5D). At day 21, all mice were euthanized and flank tumors removed for analysis. Excised tumors were significantly smaller in weight than control treated tumors (Fig. 5E), and expression of IL-6 was also significantly decreased in AZD1480 treated tumors (Fig. 5F), consistent with the interpretation that AZD1480 is inhibiting tumor growth in vivo due to inhibition of STAT-3 signaling and subsequent gene transcription.
Figure 5. AZD1480 Inhibits In Vivo Growth of Subcutaneous Xenograft GBM Tumors by Inhibiting STAT-3 Activity.
A, Xenograft tumor X1046 was removed, disaggregated, roughly minced, and injected subcutaneously into athymic nude mice. Data represent mean ± SEM (*, p<0.05, Student’s t-test at day 29). B&C, Frozen tumor samples were homogenized, and 30 µg was immunoblotted with the indicated antibodies. D, Xenograft tumor X1066 was injected subcutaneously into athymic nude mice as described in (A). Data represent mean ± SEM (**, p<0.01, Student’s t-test at day 21). E, Excised flank tumors were weighed for analysis. Data represent mean ± SD (*, p<0.05, Student’s t-test). F, RNA was isolated from frozen flank tumor samples, followed by generation of cDNA, and quantitative RT-PCR. Data represent mean of group ± SD (*, p<0.01 Mann-Whitney Rank Sum test).
The ability of AZD1480 to inhibit tumor growth and increase survival in an intracranial model of glioma was next examined. Xenograft X1046 was stereotactically injected into the brains of 20 athymic nude mice. The tumor was allowed to establish for 5 days before starting treatment. On day 6, AZD1480 (50 mg/kg per mouse) or vehicle control was administered orally once a day for 3 weeks with the endpoint measuring survival. The mice treated with AZD1480 had significantly increased survival when compared to vehicle treated mice (Fig. 6A). The intracranial model of glioma was evaluated using another xenograft, X1016, as described above. As shown in Fig. 6B, mice receiving AZD1480 treatment survived significantly longer than those receiving vehicle control. It should be noted that xenograft X1046 is more sensitive to the effects of AZD1480 compared to xenograft X1016, which will be addressed in the Discussion.
Figure 6. The JAK Inhibitor AZD1480 Increases Survival of Mice Bearing Intracranial Xenograft GBM Tumors.
A&B, Xenograft tumor X1046 (A) or X1016 (B) was removed, disaggregated into single cells and injected intracranially into nude mice. Survival was measured and mice were euthanized upon moribund or by 180 days.
Discussion
Here we report our findings of AZD1480, a JAK1,2 inhibitor, and the anti-tumor effects in GBM tumors both in vitro and in vivo. AZD1480 inhibited constitutive and stimulus-enhanced JAK/STAT-3 signaling in three established GBM cell lines. AZD1480 also reduced the expression of several downstream gene targets of STAT-3; c-Myc, SOCS3, and IL-6, and elicited anti-tumor functional effects in glioma cells as seen by a decrease in proliferation, inhibition of soft agar colony formation and an induction of apoptosis.
We performed studies using primary human GBM samples that are maintained as subcutaneously propagated xenograft tumors (29). A panel of 8 xenograft tumors was examined, and we found that JAK2 and STAT-3 activation was evident in all tumors, albeit the levels of activation vary among tumors. This heterogeneity is similar to what is seen in patient human samples (18). Both STAT-3 residues were phosphorylated in the xenografts, suggesting the presence of a transcriptionally active STAT-3 protein (14). Several of the xenografts were tested for responsiveness to AZD1480. AZD1480 effectively inhibited constitutive and stimulus-induced STAT-3 signaling, gene expression, and significantly inhibited proliferation of the xenograft cells.
Activated STAT-3 induces the expression of a wide array of genes that promote anti-apoptotic behavior, drug resistance, cell migration and invasion, angiogenesis, and evasion of anti-tumor immunity (13). AZD1480 potently inhibited IL-6 and OSM induction of c-Myc and SOCS3 in glioma cells and GBM xenograft tumors. Of interest was the observation that expression of IL-6 was also inhibited by AZD1480. IL-6 has traditionally been considered to be an NF-κB responsive gene, particularly in response to TNF-α (34). NF-κB is constitutively activated in GBMs, and associated with apoptotic resistance and poor disease prognosis (12, 28, 35). The elevated levels of IL-6 detected in many cancers have been thought to result from activation of the NF-κB pathway (36). Our findings demonstrate that IL-6 and OSM activation of STAT-3 promotes IL-6 expression by GBM cells, indicating that IL-6 is also a STAT-3 target gene. Both NF-κB and STAT-3 activate IL-6, as well as other genes that promote cell survival, growth, angiogenesis, invasiveness and motility (12, 36). The complex cross-talk between the NF-κB and JAK/STAT pathways is beginning to be elucidated, and data illustrate that the JAK/STAT/NF-κB axis is critical for tumor progression (36–38). Given the inter-dependency of the two pathways, inhibitors such as AZD1480 may attenuate NF-κB activation in vivo in the tumor microenvironment, as well as suppressing the JAK/STAT pathway. This remains to be evaluated in GBM.
The cancer stem cell hypothesis with regards to GBMs remains a complicated and challenging issue (39, 40), although it is clear that GICs are critical for tumor propagation, angiogenesis, invasion and therapeutic resistance. CD133 was originally identified to be a “restrictive” initiating cell marker for GBM and necessary for tumorigenesis (41, 42). However, reports have illustrated that CD133 negative cells are also tumorigenic in vivo, demonstrating that cell surface markers to identify cancer initiating cell populations are more complicated and dynamic than originally thought (43, 44). In our studies, we did not wish to restrict the cancer initiating cell population to cells which express CD133, as we realize that other markers, such as SSEA-1 may be important (45). We revealed that AZD1480 is an effective inhibitor of STAT-3 signaling in both populations of GICs, regardless of CD133 expression status. The importance of STAT-3 in maintenance of GICs phenotype has been recently elucidated (32, 33, 44). Our results indicate that AZD1480 can target the GIC population in addition to resident tumor cells, thus having the potential to be a very effective therapeutic agent for patients with GBM.
In vivo, we found that AZD1480 inhibited xenograft tumor growth in a flank model using xenografts X1046 and X1066. This inhibition of growth correlated with decreased STAT-3 activation, indicating that AZD1480 treatment is preventing the transcriptional activity of STAT-3. This was accompanied by a decrease in expression of Cyclin A, Bcl-2, Survivin, and IL-6. In orthotopic tumor models in which GBM xenograft cells were intracranially injected, AZD1480 treated mice displayed significantly longer survival times than vehicle treated mice. It should be noted that the mice were only treated for a total of three weeks, thus, longer duration of AZD1480 treatment may yield an even greater increase in survival of the mice. These findings are also suggestive that AZD1480, administered orally, has efficacy in the central nervous system. We also observed that in the intracranial model, xenograft X1046 was more sensitive to AZD1480 therapy compared to X1016. One noticeable difference between the two xenografts is that X1016 has amplified EGFR, while X1046 does not (Supplemental Table I; data not shown). One hypothesis is that GBM tumors with amplified EGFR will require combination therapy with JAK and EGFR inhibitors for optimal response. Monotherapy of GBM patients with EGFR inhibitors does not provide improved radiographic responses or survival benefits (46), emphasizing a need for combination cancer therapies.
The current therapy for GBM tumors includes partial surgical resection, radiation and chemotherapy, as it has been shown that treatment with radiation and the DNA alkylating agent temozolomide significantly increased survival in patients (47). However, these tumors eventually recur yielding these advances ultimately unsuccessful. Combination therapies, including receptor tyrosine kinase inhibitors and anti-angiogenic agents, are currently being explored as therapeutic approaches against the invasive and resistant nature of these tumors (1). In fact, preclinical studies combining STAT-3 inhibitors with tyrosine kinase inhibitors, including EGFR and Src, report synergistic anti-tumor effects (48). Our results, along with other investigative reports, suggest AZD1480 may potentially be an effective anti-tumor agent when combined with current therapies available for GBM.
Supplementary Material
Acknowledgements
We thank the UAB Brain Tumor Animal Models Core Facility for assistance with all xenograft in vivo experiments, and AstraZeneca for generously providing AZD1480 and critical reading of the manuscript.
Financial support: T32AR007450 (B.C.M.), T32NS048039 (B.C.M), R01NS057563 (E.N.B), R01NS050665 (E.N.B.), R01CA138517 (S.E.N.), P50CA097247 (G.Y.G) from the National Institutes of Health, and CaRES Support (J.Y.M.) from the Cancer Research Experiences for Students (CaRES), University of Alabama at Birmingham.
Footnotes
Conflict-of-interest disclosure: The following authors declare no conflicts of interest: B.C.M., J-Y.M., C.P.L., G.Y.G., H.Y., Y.Z., and S.E.N. E.N.B. is a scientific advisor for The Sontag Foundation and D.H. is a full-time employee of AstraZeneca, and holds company stock.
References
- 1.Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med. 2008;359:492–507. doi: 10.1056/NEJMra0708126. [DOI] [PubMed] [Google Scholar]
- 2.Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007;21:2683–2710. doi: 10.1101/gad.1596707. [DOI] [PubMed] [Google Scholar]
- 3.Nakada M, Nakada S, Demuth T, Tran NL, Hoelzinger DB, Berens ME. Molecular targets of glioma invasion. Cell Mol Life Sci. 2007;64:458–478. doi: 10.1007/s00018-007-6342-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brennan C, Momota H, Hambardzumyan D, Ozawa T, Tandon A, Pedraza A, et al. Glioblastoma subclasses can be defined by activity among signal transduction pathways and associated genomic alterations. PLoS One. 2009;4:e7752. doi: 10.1371/journal.pone.0007752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cheng CK, Fan QW, Weiss WA. PI3K signaling in glioma--animal models and therapeutic challenges. Brain Pathol. 2009;19:112–120. doi: 10.1111/j.1750-3639.2008.00233.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fan QW, Cheng C, Knight ZA, Haas-Kogan D, Stokoe D, James CD, et al. EGFR signals to mTOR through PKC and independently of Akt in glioma. Sci Signal. 2009;2:ra4. doi: 10.1126/scisignal.2000014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Demuth T, Reavie LB, Rennert JL, Nakada M, Nakada S, Hoelzinger DB, et al. MAP-ing glioma invasion: mitogen-activated protein kinase kinase 3 and p38 drive glioma invasion and progression and predict patient survival. Mol Cancer Ther. 2007;6:1212–1222. doi: 10.1158/1535-7163.MCT-06-0711. [DOI] [PubMed] [Google Scholar]
- 8.Pu P, Zhang Z, Kang C, Jiang R, Jia Z, Wang G, et al. Downregulation of Wnt2 and beta-catenin by siRNA suppresses malignant glioma cell growth. Cancer Gene Ther. 2009;16:351–361. doi: 10.1038/cgt.2008.78. [DOI] [PubMed] [Google Scholar]
- 9.Nozell S, Laver T, Moseley D, Nowoslawski L, De Vos M, Atkinson GP, et al. The ING4 tumor suppressor attenuates NF-kappaB activity at the promoters of target genes. Mol Cell Biol. 2008;28:6632–6645. doi: 10.1128/MCB.00697-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bredel M, Scholtens DM, Yadav AK, Alvarez AA, Renfrow JJ, Chandler JP, et al. NFKBIA deletion in glioblastomas. N Engl J Med. 2011;364:627–637. doi: 10.1056/NEJMoa1006312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Purow BW, Haque RM, Noel MW, Su Q, Burdick MJ, Lee J, et al. Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res. 2005;65:2353–2363. doi: 10.1158/0008-5472.CAN-04-1890. [DOI] [PubMed] [Google Scholar]
- 12.Atkinson GP, Nozell SE, Benveniste ET. NF-kappaB and STAT3 signaling in glioma: targets for future therapies. Expert Rev Neurother. 2010;10:575–586. doi: 10.1586/ern.10.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Aggarwal BB, Kunnumakkara AB, Harikumar KB, Gupta SR, Tharakan ST, Koca C, et al. Signal transducer and activator of transcription-3, inflammation, and cancer: how intimate is the relationship? Ann N Y Acad Sci. 2009;1171:59–76. doi: 10.1111/j.1749-6632.2009.04911.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wen Z, Zhong Z, Darnell JE., Jr Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell. 1995;82:241–250. doi: 10.1016/0092-8674(95)90311-9. [DOI] [PubMed] [Google Scholar]
- 15.Al Zaid Siddiquee K, Turkson J. STAT3 as a target for inducing apoptosis in solid and hematological tumors. Cell Res. 2008;18:254–267. doi: 10.1038/cr.2008.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809. doi: 10.1038/nrc2734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bowman T, Garcia R, Turkson J, Jove R. STATs in oncogenesis. Oncogene. 2000;19:2474–2488. doi: 10.1038/sj.onc.1203527. [DOI] [PubMed] [Google Scholar]
- 18.Brantley EC, Nabors LB, Gillespie GY, Choi YH, Palmer CA, Harrison K, et al. Loss of protein inhibitors of activated STAT-3 expression in glioblastoma multiforme tumors: implications for STAT-3 activation and gene expression. Clin Cancer Res. 2008;14:4694–4704. doi: 10.1158/1078-0432.CCR-08-0618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pilati C, Amessou M, Bihl MP, Balabaud C, Van Nhieu JT, Paradis V, et al. Somatic mutations activating STAT3 in human inflammatory hepatocellular adenomas. J Exp Med. 2011;208:1359–1366. doi: 10.1084/jem.20110283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Brantley EC, Benveniste EN. Signal transducer and activator of transcription-3: a molecular hub for signaling pathways in gliomas. Mol Cancer Res. 2008;6:675–684. doi: 10.1158/1541-7786.MCR-07-2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tchirkov A, Khalil T, Chautard E, Mokhtari K, Veronese L, Irthum B, et al. Interleukin-6 gene amplification and shortened survival in glioblastoma patients. Br J Cancer. 2007;96:474–476. doi: 10.1038/sj.bjc.6603586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Carro MS, Lim WK, Alvarez MJ, Bollo RJ, Zhao X, Snyder EY, et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature. 2010;463:318–325. doi: 10.1038/nature08712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–173. doi: 10.1016/j.ccr.2006.02.019. [DOI] [PubMed] [Google Scholar]
- 24.Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110. doi: 10.1016/j.ccr.2009.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 2009;16:487–497. doi: 10.1016/j.ccr.2009.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ioannidis S, Lamb ML, Wang T, Almeida L, Block MH, Davies AM, et al. Discovery of 5-Chloro-N(2)-[(1S)-1-(5-fluoropyrimidin-2-yl)ethyl]-N(4)-(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (AZD1480) as a novel inhibitor of the Jak/Stat pathway. J Med Chem. 2011;54:262–276. doi: 10.1021/jm1011319. [DOI] [PubMed] [Google Scholar]
- 27.Ma X, Reynolds SL, Baker BJ, Li X, Benveniste EN, Qin H. IL-17 enhancement of the IL-6 signaling cascade in astrocytes. J Immunol. 2010;184:4898–4906. doi: 10.4049/jimmunol.1000142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhao X, Laver T, Hong SW, Twitty GB, Jr, Devos A, Devos M, et al. An NF-kappaB p65-cIAP2 link is necessary for mediating resistance to TNF-alpha induced cell death in gliomas. J Neurooncol. 2011;102:367–381. doi: 10.1007/s11060-010-0346-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Giannini C, Sarkaria JN, Saito A, Uhm JH, Galanis E, Carlson BL, et al. Patient tumor EGFR and PDGFRA gene amplifications retained in an invasive intracranial xenograft model of glioblastoma multiforme. Neuro Oncol. 2005;7:164–176. doi: 10.1215/S1152851704000821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Li GH, Wei H, Lv SQ, Ji H, Wang DL. Knockdown of STAT3 expression by RNAi suppresses growth and induces apoptosis and differentiation in glioblastoma stem cells. Int J Oncol. 2010;37:103–110. [PubMed] [Google Scholar]
- 31.Hamburger AW, Salmon SE. Primary bioassay of human tumor stem cells. Science. 1977;197:461–463. doi: 10.1126/science.560061. [DOI] [PubMed] [Google Scholar]
- 32.Sherry MM, Reeves A, Wu JK, Cochran BH. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells. 2009;27:2383–2392. doi: 10.1002/stem.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang H, Lathia JD, Wu Q, Wang J, Li Z, Heddleston JM, et al. Targeting interleukin 6 signaling suppresses glioma stem cell survival and tumor growth. Stem Cells. 2009;27:2393–2404. doi: 10.1002/stem.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Libermann TA, Baltimore D. Activation of interleukin-6 gene expression through the NF-kappa B transcription factor. Mol Cell Biol. 1990;10:2327–2334. doi: 10.1128/mcb.10.5.2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Weaver KD, Yeyeodu S, Cusack JC, Jr, Baldwin AS, Jr, Ewend MG. Potentiation of chemotherapeutic agents following antagonism of nuclear factor kappa B in human gliomas. J Neurooncol. 2003;61:187–196. doi: 10.1023/a:1022554824129. [DOI] [PubMed] [Google Scholar]
- 36.Grivennikov S, Karin M. Autocrine IL-6 signaling: a key event in tumorigenesis? Cancer Cell. 2008;13:7–9. doi: 10.1016/j.ccr.2007.12.020. [DOI] [PubMed] [Google Scholar]
- 37.Lee H, Herrmann A, Deng JH, Kujawski M, Niu G, Li Z, et al. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell. 2009;15:283–293. doi: 10.1016/j.ccr.2009.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lam LT, Wright G, Davis RE, Lenz G, Farinha P, Dang L, et al. Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma. Blood. 2008;111:3701–3713. doi: 10.1182/blood-2007-09-111948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Park DM, Rich JN. Biology of glioma cancer stem cells. Mol Cells. 2009;28:7–12. doi: 10.1007/s10059-009-0111-2. [DOI] [PubMed] [Google Scholar]
- 40.Stiles CD, Rowitch DH. Glioma stem cells: a midterm exam. Neuron. 2008;58:832–846. doi: 10.1016/j.neuron.2008.05.031. [DOI] [PubMed] [Google Scholar]
- 41.Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401. doi: 10.1038/nature03128. [DOI] [PubMed] [Google Scholar]
- 42.Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–5828. [PubMed] [Google Scholar]
- 43.Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, et al. CD133(+) and CD133(−) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007;67:4010–4015. doi: 10.1158/0008-5472.CAN-06-4180. [DOI] [PubMed] [Google Scholar]
- 44.Guryanova OA, Wu Q, Cheng L, Lathia JD, Huang Z, Yang J, et al. Nonreceptor tyrosine kinase BMX maintains self-renewal and tumorigenic potential of glioblastoma stem cells by activating STAT3. Cancer Cell. 2011;19:498–511. doi: 10.1016/j.ccr.2011.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Son MJ, Woolard K, Nam DH, Lee J, Fine HA. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell Stem Cell. 2009;4:440–452. doi: 10.1016/j.stem.2009.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Huang PH, Xu AM, White FM. Oncogenic EGFR signaling networks in glioma. Sci Signal. 2009;2:re6. doi: 10.1126/scisignal.287re6. [DOI] [PubMed] [Google Scholar]
- 47.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- 48.Lo HW, Cao X, Zhu H, Ali-Osman F. Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res. 2008;14:6042–6054. doi: 10.1158/1078-0432.CCR-07-4923. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






