Diffuse intrinsic pontine gliomas (DIPG) are an incurable childhood brain cancer for which novel treatments are needed. DIPGs are characterized by a mutation in the H3 histone (H3K27M), resulting in loss of H3K27 methylation and global gene dysregulation. TRX-E-009–1 is a novel anticancer agent with preclinical activity demonstrated against a range of cancers. We examined the antitumor activity of TRX-E-009–1 against DIPG neurosphere cultures and observed tumor-specific activity with IC50s ranging from 20 to 100 nmol/L, whereas no activity was observed against normal human astrocyte cells. TRX-E-009–1 exerted its anti-proliferative effect through the induction of apoptotic pathways, with marked increases in cleaved caspase 3 and cleaved PARP levels, while also restoring histone H3K27me3 methylation. Co-administration of TRX-E-009–1 and the histone deacetylase (HDAC) inhibitor SAHA extended survival in DIPG orthotopic animal models. This antitumor effect was further enhanced with irradiation. Our findings indicate that TRX-E-009–1, combined with HDAC inhibition, represents a novel, potent therapy for children with DIPG.

Diffuse intrinsic pontine glioma (DIPG) is an aggressive brainstem tumor and the most common form of high-grade glioma in children, with a median survival of less than 1 year (1, 2). DIPG is uniformly fatal and remains the leading cause of childhood brain tumor-related death (3). There are no established systemic therapies, and focal irradiation remains the standard treatment (4, 5). However, the application of autopsy and biopsy sampling has aided in understanding the molecular landscape of DIPG tumors over the past decade (6, 7). Approximately 80% of DIPG tumors harbor a mutation in the H3 histone (H3K27M), leading to loss of H3K27 trimethylation (H3K27me3) and aberrant gene expression (8). Moreover, the development of patient-derived cultures and orthotopic models allows the preclinical testing of a variety of epigenetic, targeted agents, and immunotherapies (1, 9, 10).

One of the defining characteristics of DIPG tumors is the infiltrative and invasive growth patterns that contribute to limited therapeutic response (11). Tumor migration, cell division, and structural integrity depend upon microtubules composed of dynamic tubular polymers of α- and β-tubulin (12, 13). Blocking the dynamics of microtubule assembly and disassembly results in disruption of cellular division, mitotic arrest, and induction of apoptosis, thus making this a potential strategy for cancer therapy (14). Trilexium (TRX-E-009–1) is a third-generation benzopyran compound that can cross the blood–brain barrier (BBB) and reach brain tissue. Benzopyran is a naturally occurring compound that has been shown to display different biological effects such as antimicrobial, anti-HIV, anti-inflammatory, anti-coagulant, and antitumor (15–17). As a novel anticancer therapy, TRX-E-009–1 has been shown to initiate various mechanisms of cell death via several pathways, including through inhibition of microtubule polymerization (18). TRX-E-009–1 has been shown to significantly reduce tumor volume in preclinical models of melanoma (19).

Recent therapeutic strategies targeting epigenetic alterations by using epigenetic modifiers have shown promise in preclinical models of pediatric brain cancer (20). These treatment strategies often aim at restoring the normal function of histones by reverting the repressed trimethylation or acetylation phenotype (21). A comprehensive study using a chemical screen in patient-derived DIPG neurospheres along with integrated computational modelling, has highlighted histone deacetylases (HDAC) as promising targets (9). HDACs have been shown to interact and deacetylate microtubules in vivo (22). This led us to a combinatorial approach to enhance efficacy.

Our study demonstrates the antitumor potential of TRX-E-009–1 in combination with HDAC inhibitor SAHA. The combination of both agents inhibited cell proliferation and clonogenic potential and induced G2–M arrest and apoptosis. Furthermore, this combination also markedly enhanced survival in an orthotopic model of DIPG when combined with irradiation. These preclinical results support the translation of microtubule-targeting compounds into the clinic for DIPG therapy.

Cell culture and reagents

Patient-derived DIPG cultures were kindly provided by our collaborators at Stanford University and St John of Hope Hospital, respectively. The cells were cultured in DMEM/F12 and neurobasal media (1:1) supplemented with heparin, B27, human EGF, human FGF, PDGF-AA, PDGF-BB, HEPES, nonessential amino acids, antibiotic/antimycotic, pyruvate and glutamax. Normal human astrocytes and MRC5 cells were purchased from Lonza, and cultured according to the manufacturer's instructions. All cells were maintained at 37°C in 5% CO2 and humidified atmosphere. Cell lines were validated at the Garvan Molecular Genetics using short tandem repeat DNA profiling and were routinely confirmed as being mycoplasma negative using MycoAlert Mycoplasma Detection Kit from Lonza. TRX-E-009–1 (solutol and liposomal formulations) were provided by Novogen. Solutol formulation was used to assess anti-clonogenic effects in combination with irradiation while all remaining experiments were performed with the liposomal formulation.

Cell viability assay

For cell viability assay, DIPG cells were seeded into 96-well plates (100 μL per well) and incubated for 72 hours to form neurospheres, whereas NHAs and MRC5 cells were incubated for 24 hours prior to adding the drugs. After incubation, cells were treated with various concentrations of TRX-E-009–1 and SAHA for an additional 72 hours. Resazurin reduction was then used to measure cell viability. Synergistic drug interactions were determined using CalcuSyn software (Biosoft) developed by Chou and Talalay where CI values of less than 1 indicated synergism, CI values more than 1 showed antagonism and CI equal to 1 represented additive effect. IC50 values were determined by nonlinear regression analysis by Graphpad Prism 6 software (Applied BioSystems).

Clonogenic assay

Long-term clonogenic assays were used to assess the effects of TRX-E-009–1, SAHA, and irradiation on the clonogenic activity of DIPG cells. First, 24-well plates were coated by media containing 0.5% soft agar. After 24 hours, cell suspensions in media containing 0.33% agar and drugs or DMSO as a vehicle were added to the bottom layer. Depending on the growth rate of DIPG cultures, 1,000 to 2,000 cells were seeded in each well. Cells were maintained at 37°C in a humidified 5% CO2 atmosphere. After 2 to 3 weeks, the colonies were stained using 5 mg/mL 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and counted using ImageJ software.

Cell-cycle analysis

SU-DIPGXVII cells were treated with TRX-E-009–1 for 24 hours. Cells were then collected and fixed with 70% ice-cold ethanol and stained with 25 μg/mL propidium iodide (Sigma-Aldrich, Catalog No. P4864) and 2 μg/mL DNase free RNase (Roche Diagnostics) in 0.1% BSA/PBS. Cells were resuspended in cold 0.1% BSA/PBS and were run on FACSCalibur Flow Cytometer (BD Biosciences). Data were analyzed using Flow analysis software (Flow Jo).

Western blot analysis

Following treatment, HSJD-DIPG007 cells were lysed using RIPA buffer supplemented with protease and phosphatase inhibitors and protein concentration was determined using the Pierce BCA Analysis Kit per manufacturer's instructions. Thirty micrograms of protein was then incubated for 5 minutes at 95°C in Laemmli loading buffer containing 10% DTT (Bio-Rad). Electrophoresis was conducted at 80V for 2 hours and proteins were transferred to nitrocellulose membranes (Bio-Rad) at 85V for 1 hour. Membranes were blocked in 5% skim milk in PBS containing 0.05% Tween 20 (PBST) and then incubated with primary antibodies diluted in 5% BSA in PBST containing overnight at 4°C [anti-gamma H2A.X (1:1,000; Abcam, #ab26350), p21 Waf1/Cip1 (1:1,000; Cell Signaling Technology, #2946), cleaved caspase-3 (1:1,000; Cell Signaling Technology, #9579), cleaved PARP (1:1,000; Cell Signaling Technology, #5625), GAPDH (1:2,000; Cell Signaling Technology, #2118), β-actin (1:1,000; Cell Signaling Technology, #8457), H3K27me3 (1:1,000; Cell Signaling Technology, #9733), H3K27ac (1:1,000; Cell Signaling Technology, #8173), acetyl-α-tubulin (1:1,000; Cell Signaling Technology, #3971), HDAC6 (1:1,000; Cell Signaling Technology, #7558)]. Membranes were then washed 5× using PBST and incubated with secondary antibody in PBST for 1 hour at room temperature. Proteins were visualized by chemiluminescence (Super Signal; Pierce).

Tubulin polymerization assay

The effect of the TRX-E-009–1 on cell-free tubulin polymerization was conducted using a Tubulin Polymerization Assay Kit (Cytoskeleton, #BK006P) as per manufacturers’ instructions. Tubulin polymerization assay on HSJD-DIPG007 cells was conducted as described previously (23). Briefly, HSJD-DIPG007 cells were seeded at a density of 1,000,000 cells/well in 6-well plates. Cells were incubated at 37°C for 3 days and then were either left untreated or treated with TRX-E-009–1. Cells were collected after 24 hours following treatment. Briefly, cells were washed with PBS twice and centrifuged at 3,000 rpm for 5 minutes at 4°C. Cells were then lysed in protease inhibitor and Hypotonic buffer 1 (20 mmol/L Tris-HCl, pH 6.8, 1 mmol/L MgCl2, 2 mmol/L EGTA, 0.5% Nonidet P-40, 2 mmol/L PMSF, 200 U/mL Aprotinin, 100 μg/mL soybean trypsin inhibitor, 5 mmol/L e-amino caproic acid, and 1 mmol/L benzamidine) and incubated at 37°C in the dark. Samples were then vortexed and centrifuged at 16,000 × g for 10 minutes. Supernatant (depolymerized tubulin) was then separated from the cell pellet (polymerized tubulin) and transferred into new tubes. The cell pellets were resuspended in hypotonic buffer 2 containing 10 mmol/L Tris, pH 7.5, 1.5 mmol/L MgCl2, 10 mmol/L KCl, 0.5% Nonidet P-40, and the protease inhibitor. Western blot analysis was conducted as described above using α-Tubulin (1:1,000, Cell Signaling Technology; 11H10 #2125).

Animal models

All animal experiments were performed in accordance with the Animal Care and Ethics Committee of the University of New South Wales and the Australian Code of Practice for Care and Use of Animals for Scientific Purposes. All mice were maintained in a temperature-controlled environment with a 12-hour light/dark cycle. DIPG orthografts were generated as described previously (24). Briefly, female NOD/SCID and female balb c/nude mice (7–8 weeks, Animal Resources Centre) were stereotactically injected with 200,000 HSJD-DIPG007 or SU-DIPGVI in 2 μL of Matrigel in the 4th ventricle/pons. All animals were monitored daily for weight loss and neurological symptoms due to drug treatments and/or tumor engraftment. If ataxia, circling, head tilting, and/or weight loss of >20% occurred, the mice were humanely euthanized.

In vivo drug treatment

For a single-agent study, Balb/c nude mice received 50 mg/kg TRX-E-009–1 intravenously thrice a week for 4 weeks. Treatment started 4 weeks post-intracranial HSJD-DIPG007 cells inoculation and upon drug treatment.

The combination study treatment commenced at 5 weeks after intracranial injection of the cells. At week 5, specific cohorts were treated with irradiation at 2Gy per day for 4 days. TRX-E-009–1 and SAHA treatment as single agents or combination started on week 6 where TRX-E-009–1 was given three times a week (M/W/F) intravenously at 40 mg/kg, and SAHA was administered intraperitoneally at the dosage of 35 mg/kg 5 days per week (M-F). The treatment continued for four weeks, and 24 hours after the final dose, two animals were euthanized, and brain samples were subjected to IHC. Control animals received the vehicles of both drugs. Plasma and brain tissue samples were collected from the animals two hours after the last administration of TRX-E-009–1. Pharmacokinetic (PK) analysis of the collected samples was outsourced to Jubilant Biosys.

IHC

Excised brain tissues were fixed in formalin, embedded in paraffin, and 5 μmol/L sections were cut and mounted on Superfrost slides. The slides were dried in a 60°C oven for 1 hour and deparaffinized in xylene for 20 minutes. Slides were then re-hydrated with 100%, 90%, 70% ethanol, and MilliQ water. Antigen retrieval was performed by immersing the slides in 100 mmol/L tri-sodium citrate buffer at pH 6 and heating for 20 minutes with a cooling interval of 10 minutes following every 5 minutes of heating. The endogenous peroxidases were then inactivated by immersing the slides in 3% hydrogen peroxide. Nonspecific binding of immunoglobulin was blocked by 2.5% BSA, 2.5% FCS, and 0.2% Triton-X for 1 hour at room temperature. Slides were then incubated with primary antibodies [SSRP1 (1:1,000; Origene, #TA308461), SUPT16H (1:1,000; Thermo Fisher Scientific, #MA5–27214), Ki67 (1:1,000; Abcam, #ab209897), H3K27me3 (1:500; Abcam, #ab6002), H3K27ac (1:500; Abcam, #ab177178), anti-gamma H2A.X (1:1,000; Abcam, #ab11174)] at 4°C overnight followed by incubation with secondary antibodies for 1 hour at room temperature. Finally, the slides were washed in MilliQ water and dehydrated sequentially with 70%, 90%, and 100% ethanol.

Statistical analysis

All statistical analyses were performed using GraphPad Prism 6 software. For in vitro and IHC experiments, one-way or two-way ANOVAs (Tukey test and Dunnett multiple comparison test) were used. A Log-rank (Mantel–Cox) test was employed for survival analyses of orthotopic xenografts. Images were analyzed using ImageJ Software (ImageJ). P values <0.05 were considered statistically significant.

Data availability

The data are available in the Article, Supplementary Information, or available from the authors upon request.

Tubulin polymerization inhibitor TRX-E-009–1 exhibits potent activity against patient-derived DIPG cultures

To determine whether targeting tubulin polymerization is an effective strategy against DIPG, TRX-E-009–1 was tested in cytotoxicity assays against six DIPG patient-derived cultures. TRXE-009–1 potently decreased DIPG cell viability, with IC50 value ranging from 0.1 to 0.8 μmol/L, whereas minimal effects were observed on human fibroblast cells (MRC5) and normal human astrocytes (NHA) indicating wide therapeutic index (Fig. 1A). Likewise, in a soft agar colony formation assay on three DIPG cultures, TRX-E-009–1 potently reduced clonogenicity at submicromolar concentrations (Fig. 1B).

Figure 1.

TRX-E-009–1 has anticancer effect against patient-derived DIPG cells. A, Patient-derived DIPG cell lines are sensitive to TRX-E-009–1 compared with normal healthy astrocytes (NHA) and normal lung fibroblast (MRC5) cells. TRX-E-009–1 impairs TRX-E-009–1 impairs clonogenic activity in three DIPG cell lines (B), which is further potentiated in HSJD-DIPG007 cells (C) when combined with irradiation in a dose-dependent manner. B and C,n = 4 independent experiments performed in duplicate. Statistical analysis was calculated using one-way ANOVA between cohorts and for treated and untreated samples (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001). C, Synergy scores were calculated by Calcusyn software.

Figure 1.

TRX-E-009–1 has anticancer effect against patient-derived DIPG cells. A, Patient-derived DIPG cell lines are sensitive to TRX-E-009–1 compared with normal healthy astrocytes (NHA) and normal lung fibroblast (MRC5) cells. TRX-E-009–1 impairs TRX-E-009–1 impairs clonogenic activity in three DIPG cell lines (B), which is further potentiated in HSJD-DIPG007 cells (C) when combined with irradiation in a dose-dependent manner. B and C,n = 4 independent experiments performed in duplicate. Statistical analysis was calculated using one-way ANOVA between cohorts and for treated and untreated samples (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001). C, Synergy scores were calculated by Calcusyn software.

Close modal

Given that radiotherapy is currently the only effective treatment for DIPG, we next sought to determine if the effect of TRX-E-009–1 could be further enhanced when combined with irradiation. When HSJD-DIPG007 cells were treated with either 0.5 or 1 μmol/L TRX-E-009–1 alone, or in combination with 2, 4, and 6 Gy irradiation, a significant and synergistic decrease in colony formation was observed for all combinations. TRX-E-009–1 combined with irradiation at 2 and 4 Gy synergistically reduced colony numbers indicated by CI values <1, whereas combination with 1 μmol/L TRX-E-009–1 and 6 Gy abrogated colony formation altogether (Fig. 1C). Together, these results show that TRX-E-009–1 is highly potent against DIPG neurosphere cultures and that this effect is enhanced through combination with irradiation.

TRX-E-009–1 treatment increases p21 protein expression and induces apoptosis

Previous studies have shown that TRX-E-009–1 induces mitotic arrest in various cell lines (19). Upon treating HSJD-DIPG007 cells with TRX-E-009-1, we observed a marked increase in p21 protein—a marker associated with linking DNA damage to cell-cycle arrest, suggesting that the cell-cycle arrest in DIPG cells was mediated by p21 over-expression (Fig. 2A; Supplementary Fig. S1).

Figure 2.

Mechanism of action of TRX-E-009-1 on DIPG cells. A, Treatment of HSJD-DIPG007 cells with 1 and 5 μmol/L TRX-E-009-1 leads to increased protein expression of p21, c-PARP, and c-Caspase 3. Western blot analysis is representative of two independent experiments. B, HSJD-DIPG007 cells were treated with TRX-E-009-1 at 1, 5, and 10 μmol/L to observe the effect on polymerized and depolymerized α-tubulin levels. Western blot analysis is representative of three independent experiments. C, TRX-E-009-1 restores H3K27 trimethylation and increases H3K27 acetylation in treated HSJD-DIPG007 cells. Western blot analysis is representative of two independent experiments. D, TRX-E-009–1 significantly extends survival in the orthotopic mouse model of HSJD-DIPG007. Treatment commenced 4 weeks after intracranial injection of HSJD-DIPG007 cells 50 mg/kg TRX-E-009–1 intravenously three times a week (M, W, F) for 4 weeks. Statistical analysis has been performed using the log-rank (Mantel–Cox) test. E, TRX-E-009–1 drug concentration in the brainstem region of treated mice 2 hours posttreatment. Data are presented as mean values ± SEM from brain samples collected from n = 2 in each cohort.

Figure 2.

Mechanism of action of TRX-E-009-1 on DIPG cells. A, Treatment of HSJD-DIPG007 cells with 1 and 5 μmol/L TRX-E-009-1 leads to increased protein expression of p21, c-PARP, and c-Caspase 3. Western blot analysis is representative of two independent experiments. B, HSJD-DIPG007 cells were treated with TRX-E-009-1 at 1, 5, and 10 μmol/L to observe the effect on polymerized and depolymerized α-tubulin levels. Western blot analysis is representative of three independent experiments. C, TRX-E-009-1 restores H3K27 trimethylation and increases H3K27 acetylation in treated HSJD-DIPG007 cells. Western blot analysis is representative of two independent experiments. D, TRX-E-009–1 significantly extends survival in the orthotopic mouse model of HSJD-DIPG007. Treatment commenced 4 weeks after intracranial injection of HSJD-DIPG007 cells 50 mg/kg TRX-E-009–1 intravenously three times a week (M, W, F) for 4 weeks. Statistical analysis has been performed using the log-rank (Mantel–Cox) test. E, TRX-E-009–1 drug concentration in the brainstem region of treated mice 2 hours posttreatment. Data are presented as mean values ± SEM from brain samples collected from n = 2 in each cohort.

Close modal

To determine whether the cytotoxic effect of TRX-E-009–1 is due to the induction of apoptosis in DIPG cells, we examined the protein levels of two key regulators of apoptosis. Treatment of HSJD-DIPG007 cells with 1 and 5 μmol/L TRX-E-009–1 caused a significant increase in cleaved caspase-3 and cleaved PARP (Fig. 2A). Together, these results show that TRX-E-009–1 reduces DIPG cell proliferation and triggers apoptosis.

TRX-E-009–1 depolymerizes microtubules in HSJD-DIPG007 cells

TRX-E-009–1 has been shown to exert its cytotoxic activity through disruption of the microtubule network (19). Therefore, we evaluated the effect of TRX-E-009–1 treatment on the relative levels of polymerized (insoluble) and depolymerized (soluble) α-tubulin in HSJD-DIPG007 cells (Fig. 2B). In untreated cells, the ratio of polymerized to depolymerized α-tubulin was roughly 0.7, whereas with TRX-E-009–1 treatment (1, 5, 10 μmol/L), this ratio decreased to 0.1, 0.3, and 0.1 respectively.

These results were further confirmed in a cell-free assay measuring tubulin polymerization, where TRX-E-009–1 treatment visibly reduced the polymerization of tubulin compared with control. Reference compounds, paclitaxel (known to stimulate tubulin polymerization) and colchicine (known to bind to tubulin thus preventing polymerization) were also tested (Supplementary Fig. S2).

TRX-E-S009–1 is an epigenetic modifier, active against DIPG in vivo

Because flavonoids have epigenetic activities including histone modification and changing DNA methylation patterns in various cancers (25–27), we evaluated the effect of TRX-E-009–1 on H3K27me3 and H3K27ac proteins. TRX-E-009–1 increased the levels of both the acetylated and methylated forms of H3K27 in HSJD-DIPG007 cells (Fig. 2C). Given the potent cytotoxic activity of TRX-E-009–1, we subsequently evaluated its antitumor efficacy against an orthotopic patient-derived xenograft model of HSJD-DIPG007. Mice were treated at 4 weeks postimplantation with 50 mg/kg TRX-E-009–1 intravenously three times a week (M,W,F) for 4 weeks (Fig. 3B). TRX-E-009–1 treatment significantly extended survival from 72 to 78 days, P = 0.0013 (Fig. 2D). Analysis of brain samples from Balb/C nude mice xenografted with the same HSJD-DIPG007 cells, 2 hours after the last treatment of TRX-E-009–1, revealed good penetration of TRX-E-009–1 drug in the brainstem region at a dose of 1.07 μmol/L (Fig. 2E). The concentration of drug in the plasma was approximately 10-fold higher, with a brain-to-plasma ratio of 0.1 (Supplementary Fig. S3).

Figure 3.

Combination of TRX-E-009–1 and SAHA is synergistic against DIPG. Combination of TRX-E-009–1 and SAHA is synergistic against HSJD-DIPG007 and SU-DIPGXVII neurospheres, resulting in decreased (A) cell survival and (B) fewer colonies formed upon combination treatment. Data are presented as mean values ± SEM. A, Single well examined over five independent experiments. B,n = 4 independent experiments performed in duplicate. Synergy scores were calculated by Calcusyn software.

Figure 3.

Combination of TRX-E-009–1 and SAHA is synergistic against DIPG. Combination of TRX-E-009–1 and SAHA is synergistic against HSJD-DIPG007 and SU-DIPGXVII neurospheres, resulting in decreased (A) cell survival and (B) fewer colonies formed upon combination treatment. Data are presented as mean values ± SEM. A, Single well examined over five independent experiments. B,n = 4 independent experiments performed in duplicate. Synergy scores were calculated by Calcusyn software.

Close modal

Combination of TRX-E-009–1 and SAHA is synergistic against DIPG neurospheres and restores H3K27 trimethylation and acetylation

Because TRX-E-009–1 affects H3K27 acetylation and trimethylation, we hypothesized that HDAC inhibitors would further potentiate the activity of TRX-E-009–1. We chose the class I HDAC inhibitor SAHA because it has activity against the HDAC6 isoform, which promotes hyperacetylation of α-tubulin (28, 29). Treatment of HSDJ-DIPG007 cells with SAHA (10 μmol/L) reduced HDAC6 expression by 50% and significantly increased hyperacetylation of α-tubulin (****, P < 0.0001; Supplementary Fig. S3). Consistent with our hypothesis, the combination of TRX-E-009–1 and SAHA synergistically inhibited cell proliferation and reduced clonogenicity against DIPG cultures, as indicated by combination indexes (CI; Fig. 3; Supplementary Fig. S4).

We next evaluated the effect of combining TRX-E-009–1 and SAHA on apoptosis and epigenetic markers. Combination treatment significantly increased the cleavage of both PARP and caspase-3 in HSJD-DIPG007, indicating induction of apoptosis (Fig. 4A). In addition, the combination also markedly increased H3K27 acetylation and H3K27 trimethylation, compared with single-agent treatment (Fig. 4B).

Figure 4.

Combination of TRX-E-009–1 and SAHA induces apoptosis and restores H3K27 trimethylation and acetylation in DIPG cells. Protein expression in HSJD-DIPG007 cells when treated with TRX-E-009–1 (1 μmol/L) and SAHA (5 μmol/L) of (A) c-PARP, c-caspase 3, γ-H2AX and (B) H3K27 trimethylation and acetylation. Representative blot from two independent experiments.

Figure 4.

Combination of TRX-E-009–1 and SAHA induces apoptosis and restores H3K27 trimethylation and acetylation in DIPG cells. Protein expression in HSJD-DIPG007 cells when treated with TRX-E-009–1 (1 μmol/L) and SAHA (5 μmol/L) of (A) c-PARP, c-caspase 3, γ-H2AX and (B) H3K27 trimethylation and acetylation. Representative blot from two independent experiments.

Close modal

TRX-E-009–1 and SAHA, in combination with irradiation, significantly reduce the clonogenicity of HSJD-DIPG007 cells and extend survival in mice models of DIPG

We next sought to determine if the addition of irradiation could further enhance the effect of the combination of TRX-E-009–1 and SAHA. Although colony formation of HSJD-DIPG007 cells in vitro was not affected by treatment with 0.005 μmol/L TRX-E-009–1 or 0.01 μmol/L SAHA alone, combining irradiation with TRX-E-009–1 or SAHA reduced colony numbers compared with control, with a CI value of 0.267. Furthermore, the triple combination of 2 Gy irradiation, TRX-E-009–1 and SAHA further inhibited colony formation, compared with TRX-E-009–1 and SAHA (*, P < 0.05; CI = 0.328; Fig. 5A).

Figure 5.

TRX-E-009–1 synergizes with SAHA and irradiation in orthotopic models of DIPG. A, Triple combination treatment of TRX-E-009–1 (0.005 μmol/L), SAHA (0.01 μmol/L), and irradiation (2Gy) significantly reduced the clonogenic activity of HSJD-DIPG007 cells. Data are presented as mean values ±SEM. n = 4 independent experiments performed in duplicate. Synergy scores were calculated by Calcusyn software (*, P < 0.05; ****, P < 0.0001). B, Mice were intracranially injected with DIPG cells and 4 weeks post-injection, treatments commenced. Mice were humanely euthanized when they displayed severe neurological decline and/or weight loss or reached maximum holding time (MHT). Survival curve of HSJD-DIPG007 with TRX-E-009–1/SAHA/Irradiation treatment. Median survival of cohorts is indicated in brackets. Statistical analysis has been performed using the log-rank (Mantel–Cox) test.

Figure 5.

TRX-E-009–1 synergizes with SAHA and irradiation in orthotopic models of DIPG. A, Triple combination treatment of TRX-E-009–1 (0.005 μmol/L), SAHA (0.01 μmol/L), and irradiation (2Gy) significantly reduced the clonogenic activity of HSJD-DIPG007 cells. Data are presented as mean values ±SEM. n = 4 independent experiments performed in duplicate. Synergy scores were calculated by Calcusyn software (*, P < 0.05; ****, P < 0.0001). B, Mice were intracranially injected with DIPG cells and 4 weeks post-injection, treatments commenced. Mice were humanely euthanized when they displayed severe neurological decline and/or weight loss or reached maximum holding time (MHT). Survival curve of HSJD-DIPG007 with TRX-E-009–1/SAHA/Irradiation treatment. Median survival of cohorts is indicated in brackets. Statistical analysis has been performed using the log-rank (Mantel–Cox) test.

Close modal

The triple combination was then assessed in vivo using an orthotopic mouse model of patient-derived HSJD-DIPG007 cells. Compared with the survival of vehicle-treated animals (61 days), irradiation alone had no significant effect (63 days). Although TRX-E-009–1 (67 days) and SAHA (64 days) as single agents prolonged survival (P = 0.0007 and 0.0212, respectively), the antitumor efficacy of both TRX-E-009–1 (68 days) and SAHA (69 days) was increased when combined with irradiation (P = 0.0518 and 0.0106, respectively). Consistent with the in vitro data, the triple combination of TRX-E-009–1, SAHA and irradiation further prolonged survival (91 days; Fig. 5B). Furthermore, histologic analysis revealed a significant reduction in the proliferation of tumor cells in the treatment groups and an increase in H3K27ac, H3K27me3, and γ-H2AX in the triple combination group compared with the other cohorts (Fig. 6; Supplementary Table S1).

Figure 6.

Triple combination of TRX-E-009–1, SAHA, and irradiation inhibits tumour cell proliferation, induces DNA damage response and restores H3K27 trimethylation and acetylation in orthotopic tumors. Ki67, γ-H2AX. H3K27ac and H3K27me3 staining of DIPG tumors after 4 weeks of treatment in HSJD-DIPG007 model. Three images were taken from brain samples collected from two mice in each cohort. Statistical analysis was calculated using one-way ANOVA between cohorts and for treated and untreated samples (**, P < 0.01; ***, P < 0.001; ****, P < 0.0001). Specific P values are listed in Supplementary Table S1.

Figure 6.

Triple combination of TRX-E-009–1, SAHA, and irradiation inhibits tumour cell proliferation, induces DNA damage response and restores H3K27 trimethylation and acetylation in orthotopic tumors. Ki67, γ-H2AX. H3K27ac and H3K27me3 staining of DIPG tumors after 4 weeks of treatment in HSJD-DIPG007 model. Three images were taken from brain samples collected from two mice in each cohort. Statistical analysis was calculated using one-way ANOVA between cohorts and for treated and untreated samples (**, P < 0.01; ***, P < 0.001; ****, P < 0.0001). Specific P values are listed in Supplementary Table S1.

Close modal

This study has demonstrated that targeting microtubule polymerization with a BBB permeable compound TRX-E-009–1 is a potential novel therapeutic strategy for DIPG. Our results show for the first time that this anti-microtubule agent inhibits cell proliferation and colony formation in DIPG. Anti-microtubule agents can result in either tubulin stabilization or destabilization (12). We found potent tubulin depolymerization in DIPG by TRX-E-009–1 alongside induction of apoptosis. These data clearly identify tubulin as a valid molecular target in DIPG and suggest that the use of TRX-E-009–1 represents a potentially valuable therapeutic approach in this disease.

Previous work with TRX-E-009–1 and other similar microtubule polymerization inhibiting compounds have shown links to additional mechanisms of action, including a significant increase in mitochondrial reactive oxygen species (ROS) production (30, 31). Targeting mitochondria has been a focus of interest in DIPG to promote cytotoxic ROS in tumor cells selectively (2). Furthermore, studies have shown that these compounds can also affect cell-cycle regulation and apoptosis, as well as modulate the activity of other signaling pathways (32, 33). These findings suggest that these compounds may have potential as multitargeted therapies, with the ability to simultaneously target multiple pathways involved in cancer progression. Further research is needed to fully understand the mechanisms of action of these compounds and to determine their potential as effective treatments for cancer.

The current standard therapy for DIPG is irradiation. Adding irradiation to the TRX/SAHA combination greatly extended survival in this aggressive tumor model, from 72 to 91 days.

The in vivo efficacy exhibited in this study shows, for the first time, that the combined use of TRX-E-009–1 and SAHA significantly enhances survival in an aggressive orthotopic model of DIPG. We have demonstrated enhanced efficacy of epigenetic modifying agents with irradiation, however, given that DIPG is an extremely aggressive brain tumor, additional combinatorial studies are warranted to enhance further effectiveness that ultimately may lead to greater benefit for the patients.

In conclusion, our findings provide the first evidence that combining a tubulin polymerization inhibitor with a HDAC inhibitor represents a novel therapeutic approach for DIPG. The cytotoxic effects are mediated through G2–M arrest, depolarized tubulin and subsequent apoptosis, and the dual targeting approach was found to combine effectively with irradiation in vitro and in vivo. TRX-E-009–1 is not currently in clinical development, however these results provide the proof of principle for this class of compounds—that is, targeting microtubule polymerization with a BBB permeable agent represents a new potential treatment strategy. Both anti-microtubule agents (NCT01729260) and HDAC inhibitors have been tested in clinical trials for DIPG (34). Although limited efficacy has been seen to date with single agents, our data suggest that the combination of these therapies may represent a novel and promising combinatorial therapeutic strategy for DIPG.

D.S. Ziegler reports personal fees from Bayer, Astra Zeneca, Novartis, Day One, FivePhusion, Amgen, Alexion, and Norgine; grants and personal fees from Accendatech outside the submitted work. No disclosures were reported by the other authors.

A. Ehteda: Formal analysis, investigation, methodology, writing–original draft. A. Khan: Formal analysis, investigation, writing–original draft, writing–review and editing. G. Rajakumar: Investigation, methodology. A.S. Vanniasinghe: Investigation. A. Gopalakrishnan: Investigation. J. Liu: Investigation. M. Tsoli: Resources, supervision, writing–original draft, writing–review and editing. D.S. Ziegler: Resources, supervision, writing–original draft, writing–review and editing.

We would like to thank A/Prof Monje and Dr Angel Montero Carcaboso for generously supplying the SU-DIPG and HSJD-DIPG cells respectively, and David Brown from Novogen for providing TRX-E-009–1. We also thank Han Shen, Laura Franshaw, Swapna Joshi, Cecilia Chang, Eleanor Ager, along with the Scientific Services Group and Flow Cytometry at UNSW for their technical support and advice. This research was supported by the Kids Cancer Project, a Cancer Institute NSW Translational Program Grant (2019/TPG2037), a NHMRC Investigator grant (GNT2017898), and a NHMRC project grant (2014/GNT1085411).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

1.
Lin
GL
,
Wilson
KM
,
Ceribelli
M
,
Stanton
BZ
,
Woo
PJ
,
Kreimer
S
, et al
.
Therapeutic strategies for diffuse midline glioma from high-throughput combination drug screening
.
Sci Transl Med
2019
;
11
:
eaaw0064
.
2.
Donaldson
SS
,
Laningham
F
,
Fisher
PG
.
Advances toward an understanding of brainstem gliomas
.
J Clin Oncol
2006
;
24
:
1266
72
.
3.
Johung
TB
,
Monje
M
.
Diffuse intrinsic pontine glioma: new pathophysiological insights and emerging therapeutic targets
.
Curr Neuropharmacol
2017
;
15
:
88
97
.
4.
Jansen
MHA
,
van Vuurden
DG
,
Vandertop
WP
,
Kaspers
GJL
.
Diffuse intrinsic pontine gliomas: a systematic update on clinical trials and biology
.
Cancer Treat Rev
2012
;
38
:
27
35
.
5.
Upton
DH
,
Ung
C
,
George
SM
,
Tsoli
M
,
Kavallaris
M
,
Ziegler
DS
.
Challenges and opportunities to penetrate the blood-brain barrier for brain cancer therapy
.
Theranostics
2022
;
12
:
4734
52
.
6.
Tsoli
M
,
Shen
H
,
Mayoh
C
,
Franshaw
L
,
Ehteda
A
,
Upton
D
, et al
.
International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma
.
J Neurooncol
2019
;
141
:
253
63
.
7.
Damodharan
S
,
Lara-Velazquez
M
,
Williamsen
BC
,
Helgager
J
,
Dey
M
.
Diffuse intrinsic pontine glioma: molecular landscape, evolving treatment strategies and emerging clinical trials
.
J Pers Med
2022
;
12
:
840
.
8.
Louis
DN
,
Giannini
C
,
Capper
D
,
Paulus
W
,
Figarella-Branger
D
,
Lopes
MB
, et al
.
cIMPACT-NOW update 2: diagnostic clarifications for diffuse midline glioma, H3 K27M-mutant and diffuse astrocytoma/anaplastic astrocytoma, IDH-mutant
.
Acta Neuropathol
2018
;
135
:
639
42
.
9.
Grasso
CS
,
Tang
Y
,
Truffaux
N
,
Berlow
NE
,
Liu
L
,
Debily
M-A
, et al
.
Functionally defined therapeutic targets in diffuse intrinsic pontine glioma
.
Nat Med
2015
;
21
:
827
.
10.
Mount
CW
,
Majzner
RG
,
Sundaresh
S
,
Arnold
EP
,
Kadapakkam
M
,
Haile
S
, et al
.
Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M(+) diffuse midline gliomas
.
Nat Med
2018
;
24
:
572
9
.
11.
Cockle
JV
,
Picton
S
,
Levesley
J
,
Ilett
E
,
Carcaboso
AM
,
Short
S
, et al
.
Cell migration in paediatric glioma; characterisation and potential therapeutic targeting
.
Br J Cancer
2015
;
112
:
693
703
.
12.
Dumontet
C
,
Jordan
MA
.
Microtubule-binding agents: a dynamic field of cancer therapeutics
.
Nat Rev Drug Discov
2010
;
9
:
790
803
.
13.
Calinescu
AA
,
Castro
MG
.
Microtubule targeting agents in glioma
.
Transl Cancer Res
2016
;
5
(
Suppl 1
):
S54
60
.
14.
Colin
DJ
,
Hain
KO
,
Allan
LA
,
Clarke
PR
.
Cellular responses to a prolonged delay in mitosis are determined by a DNA damage response controlled by Bcl-2 family proteins
.
Open Biol
2015
;
5
:
140156
.
15.
Kraus
GA
,
Mengwasser
J
,
Maury
W
,
Oh
C
.
Synthesis of chroman aldehydes that inhibit HIV
.
Bioorg Med Chem Lett
2011
;
21
:
1399
401
.
16.
Rawat
P
,
Verma
SM
.
Design and synthesis of chroman derivatives with dual anti-breast cancer and antiepileptic activities
.
Drug Des Devel Ther
2016
;
10
:
2779
88
.
17.
Patil
SA
,
Patil
SA
,
Fariyike
T
,
Marichev
KO
,
Heras Martinez
HM
,
Bugarin
A
, et al
.
Medicinal applications of coumarins bearing azetidinone and thiazolidinone moieties
.
Future Med Chem
2021
;
13
:
1907
34
.
18.
Selepe
MA
,
Kunyane
P
,
Seboletswe
P
,
Nair
S
,
Cele
N
,
Engelbrecht
M
, et al
.
Synthesis and evaluation of benzoylbenzofurans and isoflavone derivatives as sirtuin 1 inhibitors with antiproliferative effects on cancer cells
.
Bioorg Chem
2022
;
128
:
106101
.
19.
Stevenson
AJ
,
Ager
EI
,
Proctor
MA
,
Škalamera
D
,
Heaton
A
,
Brown
D
, et al
.
Mechanism of action of the third generation benzopyrans and evaluation of their broad anti-cancer activity in vitro and in vivo
.
Sci Rep
2018
;
8
:
5144
.
20.
Pfister
SX
,
Ashworth
A
.
Marked for death: targeting epigenetic changes in cancer
.
Nat Rev Drug Discov
2017
;
16
:
241
63
.
21.
Hashizume
R
.
Epigenetic targeted therapy for diffuse intrinsic pontine glioma
.
Neurol Med Chir (Tokyo)
2017
;
57
:
331
42
.
22.
Zhang
Y
,
Li
N
,
Caron
C
,
Matthias
G
,
Hess
D
,
Khochbin
S
, et al
.
HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo
.
EMBO J
2003
;
22
:
1168
79
.
23.
Giannakakou
P
,
Villalba
L
,
Li
H
,
Poruchynsky
M
,
Fojo
T
.
Combinations of paclitaxel and vinblastine and their effects on tubulin polymerization and cellular cytotoxicity: characterization of a synergistic schedule
.
Int J Cancer
1998
;
75
:
57
63
.
24.
Tsoli
M
,
Liu
J
,
Franshaw
L
,
Shen
H
,
Cheng
C
,
Jung
M
, et al
.
Dual targeting of mitochondrial function and mTOR pathway as a therapeutic strategy for diffuse intrinsic pontine glioma
.
Oncotarget
2018
;
9
:
7541
56
.
25.
Izzo
S
,
Naponelli
V
,
Bettuzzi
S
.
Flavonoids as epigenetic modulators for prostate cancer prevention
.
Nutrients
2020
;
12
:
1010
.
26.
Gilbert
ER
,
Liu
D
.
Flavonoids influence epigenetic-modifying enzyme activity: structure–function relationships and the therapeutic potential for cancer
.
Curr Med Chem
2010
;
17
:
1756
68
.
27.
Jiang
W
,
Xia
T
,
Liu
C
,
Li
J
,
Zhang
W
,
Sun
C
.
Remodeling the epigenetic landscape of cancer-application potential of flavonoids in the prevention and treatment of cancer
.
Front Oncol
2021
;
11
:
705903
.
28.
Li
D
,
Marchenko
ND
,
Moll
UM
.
SAHA shows preferential cytotoxicity in mutant p53 cancer cells by destabilizing mutant p53 through inhibition of the HDAC6-Hsp90 chaperone axis
.
Cell Death Differ
2011
;
18
:
1904
13
.
29.
Lee
JY-C
,
Kuo
C-W
,
Tsai
S-L
,
Cheng
SM
,
Chen
S-H
,
Chan
H-H
, et al
.
Inhibition of HDAC3- and HDAC6-promoted survivin expression plays an important role in SAHA-induced autophagy and viability reduction in breast cancer cells
.
Front Pharmacol
2016
;
7
:
81
.
30.
Jeyaraju
DV
,
Hurren
R
,
Wang
X
,
MacLean
N
,
Gronda
M
,
Shamas-Din
A
, et al
.
A novel isoflavone, ME-344, targets the cytoskeleton in acute myeloid leukemia
.
Oncotarget
2016
;
7
:
49777
85
.
31.
Manevich
Y
,
Reyes
L
,
Britten
CD
,
Townsend
DM
,
Tew
KD
.
Redox signaling and bioenergetics influence lung cancer cell line sensitivity to the isoflavone ME-344
.
J Pharmacol Exp Ther
2016
;
358
:
199
208
.
32.
Alvero
AB
,
Heaton
A
,
Lima
E
,
Pitruzzello
M
,
Sumi
N
,
Yang-Hartwich
Y
, et al
.
TRX-E-002–1 induces c-Jun-dependent apoptosis in ovarian cancer stem cells and prevents recurrence in vivo
.
Mol Cancer Ther
2016
;
15
:
1279
90
.
33.
Wang
T-H
,
Wang
H-S
,
Ichijo
H
,
Giannakakou
P
,
Foster
JS
,
Fojo
T
, et al
.
Microtubule-interfering agents activate c-Jun N-terminal kinase/stress-activated protein kinase through both Ras and apoptosis signal-regulating kinase pathways
.
J Biol Chem
1998
;
273
:
4928
36
.
34.
Su
JM
,
Kilburn
LB
,
Mansur
DB
,
Krailo
M
,
Buxton
A
,
Adekunle
A
, et al
.
Phase I/II trial of vorinostat and radiation and maintenance vorinostat in children with diffuse intrinsic pontine glioma: a children's oncology group report
.
Neuro Oncol
2022
;
24
:
655
64
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.