Purpose: Pediatric glioblastoma multiforme (pGBM) is a highly aggressive tumor in need of novel therapies. Our objective was to demonstrate the therapeutic efficacy of MLN8237 (alisertib), an orally available selective inhibitor of Aurora A kinase (AURKA), and to evaluate which in vitro model system (monolayer or neurosphere) can predict therapeutic efficacy in vivo.

Experimental Design: AURKA mRNA expressions were screened with qRT-PCR. In vitro antitumor effects were examined in three matching pairs of monolayer and neurosphere lines established from patient-derived orthotopic xenograft (PDOX) models of the untreated (IC-4687GBM), recurrent (IC-3752GBM), and terminal (IC-R0315GBM) tumors, and in vivo therapeutic efficacy through log rank analysis of survival times in two models (IC-4687GBM and IC-R0315GBM) following MLN8237 treatment (30 mg/kg/day, orally, 12 days). Drug concentrations in vivo and mechanism of action and resistance were also investigated.

Results: AURKA mRNA overexpression was detected in 14 pGBM tumors, 10 PDOX models, and 6 cultured pGBM lines as compared with 11 low-grade gliomas and normal brains. MLN8237 penetrated into pGBM xenografts in mouse brains. Significant extension of survival times were achieved in IC-4687GBM of which both neurosphere and monolayer were inhibited in vitro, but not in IC-R0315GBM of which only neurosphere cells responded (similar to IC-3752GBM). Apoptosis-mediated MLN8237 induced cell death, and the presence of AURKA-negative and CD133+ cells appears to have contributed to in vivo therapy resistance.

Conclusions: MLN8237 successfully targeted AURKA in a subset of pGBMs. Our data suggest that combination therapy should aim at AURKA-negative and/or CD133+ pGBM cells to prevent tumor recurrence. Clin Cancer Res; 24(9); 2159–70. ©2018 AACR.

Translational Relevance

Pediatric glioblastoma multiforme (pGBM) is a highly aggressive tumor with a poor prognosis despite multimodality therapy, thus new treatments are needed. We utilized a novel set of patient-derived orthotopic xenograft (PDOX) mouse models along with matching pairs of monolayer and neurospheres from different stages of clinical progression (diagnosis, recurrence, and terminal/autopsy). We detected Aurora A kinase (AURKA) overexpression in pGBM and identified MLN8237 (alisertib) as an effective agent against a treatment naïve pGBM. Also, our data involving three GBM models suggested that simultaneous inhibition of neurosphere and monolayer cells by MLN8237 correlated with prolonged animal survival. In addition, decreased AURKA expression (due to intratumoral heterogeneity) and the presence of CD133+ cells contributed to the recurrence or progression of pGBM xenografts following MLN8327 treatment. As MLN8237 is a reversible inhibitor of AURKA that is administered orally and can cross the blood–brain barrier, our data support the use of MLN8237 in pGBMs and highlight the need for combination therapies.

Glioblastoma multiforme (GBM) is one of the most malignant brain tumors that occurs in adults and children. Unlike adult GBM in which the standard treatment is radiotherapy and adjuvant temozolomide (1), a standard of care does not exist for the treatment of pediatric GBM (pGBM) (2). The five-year survival rate in patients with pGBM remains at 20% (3). Novel therapy for this disease is critically needed.

Recent studies suggest Aurora A serine/threonine kinases as potential oncologic targets (4, 5). Of the three related Aurora kinases (Aurora A, B, and C; ref. 6), Aurora A kinase (AURKA) regulates centrosome maturation, mitotic entry, centrosome separation, spindle formation, and cytokinesis (4). Amplification and/or overexpression of AURKA has been reported in multiple human tumors (7–14). In central nervous system tumors, AURKA overexpression correlates with disease progression and shorter survival times (15–17). Among the newly developed AURKA inhibitors, MLN8237 (alisertib) is an orally available reversible selective inhibitor with strong antitumor activity both in vitro and in vivo (12, 17–22). In addition, MLN8237 passes through the blood–brain barrier (BBB), and thus is an attractive agent to treat CNS malignancies (23). One of the key mechanisms of MLN8237-induced cell death is upregulation of p53 (11, 19). As p53 mutation is far less frequent in pGBM than in adult GBM (24, 25) and MLN8237 exhibited an acceptable safety profile in adult and pediatric phase I/II trials (26–30), the applicability of MLN8237 can potentially be greater and expedited in pGBM tumors.

As the incidence of pGBM is less than adult GBM and the number of available new candidate treatment agents is increasing, it is important to establish strong preclinical rationale to prioritize new agents for a clinical trial, and more importantly, to improve the chances of clinical success. For initial drug screening, it is desirable to develop an in vitro drug testing system that can predict in vivo efficacy in animal models. In addition to traditional monolayer cultures, new three-dimensional (3D) cultures, such as spheroids and organoids (31), have been developed. While neurospheres better represent 3D tumor architecture, microenvironment, and cellular heterogeneity of patient tumor and favor the growth of cancer stem cells (CSC), the lack of paired neurosphere and monolayer cultures derived from the same patient makes it difficult to determine which culture type better predicts in vivo treatment response or whether tumor cells in both cultures need to be targeted. For the subsequent in vivo evaluation of therapeutic efficacy, it is ideal to include model systems derived from tumors at different points of disease presentation. For example, therapies that are effective in treatment-naïve animal models frequently fail in the heavily pretreated patients with refractory tumors who are the subjects of most early-phase clinical trials. While conversely, testing new drugs in comparatively resistant tumor models jeopardizes discounting new therapies, which may prove effective in the context of upfront therapy.

We have optimized a surgical procedure that allows for the safe and rapid implantation of pediatric brain tumor cells into the matching locations in the brains of SCID mice (32–36). Our detailed characterization of these patient-derived orthotopic xenograft (PDOX) mouse models has confirmed their faithful replication of histopathologic features, invasive phenotypes, and major genetic abnormalities of the original patient tumors (32–36). From PDOX tumors of pGBM, we also established three matching pairs of cultured monolayer and neurospheres to facilitate the in vitro and in vivo evaluation of new therapies, such as MLN8237 in pGBMs.

In this report, we evaluated AURKA expression in pGBMs compared with pediatric low-grade gliomas, examined the in vitro antitumor effects of MLN8237 by treating paired monolayer and neurosphere cultures established from three pGBM models derived from untreated, recurrent, and terminal/lethal tumors, performed detailed analyses of in vivo therapeutic efficacy, and determined mechanisms of action of MLN8237 in two pGBM models. Our objectives were to examine (i) whether AURKA is a therapeutic target in pGBM, (ii) whether MLN8237 can effectively target this deadly disease, and (iii) whether effective targeting of both monolayer and neurosphere cells predicts prolonged animal survival time.

Pediatric glioma tumors

Fresh tumor tissue was collected from 11 patients with low-grade gliomas (LGG; WHO grade I/II) and 14 patients with pGBMs (WHO grade IV). Signed informed consent was obtained from the patient or legal guardian prior to sample acquisition in accordance with Institutional Review Board (IRB) policy. All studies were conducted in accordance with the ethical guideline of Declaration of Helsinki. Normal control human cerebellar RNAs from 5 adults as well as total RNAs from 2 fetal brains was procured from a commercial source (Clontech Laboratories, Inc. and Biochain; ref. 37).

PDOX mouse models

Orthotopic free-hand surgical transplantation of tumor cells into mouse cerebrum was performed as described previously (36) following an Institutional Animal Care and Use Committee–approved protocol. PDOX models of intracerebral (IC)-4687GBM, IC-3752GBM (38), and IC-R0315GBM were established by direct injection of surgical or autopsy specimens into mouse cerebra; maintenance of reproducible tumorigenicity was confirmed for 5 in vivo passages. These xenograft tumors replicated major histopathologic features of the original patient tumors (38), and all three models are highly invasive in mouse brains. Patient tumor 4687GBM was obtained at the time of initial tumor resection (therapy-naïve), while patients 3752GBM and R0315GBM were heavily treated prior to sample acquisition (Table 1). The NOD/SCID mice were maintained in a pathogen-free animal facility at Texas Children's Hospital (Baylor College of Medicine, Houston, TX). Mice of both sexes, ages 6–8 weeks, were anesthetized with sodium pentobarbital (50 mg/kg). Tumor cells (1 × 105), isolated from donor xenografts, were suspended in 2 μL of culture medium and injected into the cerebral hemisphere (1 mm to the right of the midline, 1.5 mm anterior to the lambdoid suture, and 3 mm deep) with a 10-μL 26-gauge Hamilton Gastight 1701 syringe needle.

Table 1.

Patient clinical course

4687GBM3752GBMR0315GBM
Clinical timepoint Initial Recurrence Autopsy 
Age 7 y 4 y 9 y 
Gender Male Female Female 
Location Right hemisphere and thalamus Left frontal Left parietal 
Initial treatment Subtotal resection Gross total resection Subtotal resection 
 Sample obtained Radiotherapy Radiotherapy 
 Radiotherapy Temozolomide Temozolomide 
Recurrence  Recurrent/progressive left frontal GBM at 5 months from diagnosis Recurrent left parietal GBM at 8 months from diagnosis 
1st relapse treatment   Debulking and Cyberknife 
  Peptide vaccine pilot study Procarbazine, Lomustine, and Vincristine 
   Bevacizumab 
Progression Clinical tumor progression at 5 months from diagnosis Further progression of tumor at 7 months from diagnosis Further progression of tumor 14 months from diagnosis 
Subsequent relapse Comfort care Partial resection Comfort care 
treatment  Sample obtained  
  Phase I trial of vorinostat/bortezomib  
Autopsy   Sample obtained 
P53 mutation Not detected Not detected Not detected 
H3K27 mutation Not detected Not detected Not detected 
Sample Previously untreated sample obtained from initial surgery Sample obtained at repeat resection after primary and salvage treatment Therapy-resistant autopsy sample 
4687GBM3752GBMR0315GBM
Clinical timepoint Initial Recurrence Autopsy 
Age 7 y 4 y 9 y 
Gender Male Female Female 
Location Right hemisphere and thalamus Left frontal Left parietal 
Initial treatment Subtotal resection Gross total resection Subtotal resection 
 Sample obtained Radiotherapy Radiotherapy 
 Radiotherapy Temozolomide Temozolomide 
Recurrence  Recurrent/progressive left frontal GBM at 5 months from diagnosis Recurrent left parietal GBM at 8 months from diagnosis 
1st relapse treatment   Debulking and Cyberknife 
  Peptide vaccine pilot study Procarbazine, Lomustine, and Vincristine 
   Bevacizumab 
Progression Clinical tumor progression at 5 months from diagnosis Further progression of tumor at 7 months from diagnosis Further progression of tumor 14 months from diagnosis 
Subsequent relapse Comfort care Partial resection Comfort care 
treatment  Sample obtained  
  Phase I trial of vorinostat/bortezomib  
Autopsy   Sample obtained 
P53 mutation Not detected Not detected Not detected 
H3K27 mutation Not detected Not detected Not detected 
Sample Previously untreated sample obtained from initial surgery Sample obtained at repeat resection after primary and salvage treatment Therapy-resistant autopsy sample 

Growth of matched pairs of monolayer and 3D neurosphere cultures from PDOX pGBM cells

Three pairs of cultured pGBM cells were initiated from tumors harvested from PDOX models and established as both monolayer and neurosphere cultures (32, 39). Monolayer cells were cultured in DMEM supplemented with 10% FBS (Atlanta Biologicals, Inc.), 200 U/mL penicillin/streptomycin. Neurosphere cells, in which putative CSC populations are enriched, were cultured in serum-free cell growth medium consisting of neurobasal media, N2 and B27 supplements (Life Technologies, Grand Island, NY), recombinant human βFGF and EGF (50 ng/mL each; R&D Systems Inc.), and 200 U/mL penicillin/streptomycin (32, 39).

Quantitative real-time PCR

Total RNA was extracted with TRIzol reagent (Invitrogen) from snap frozen patient and xenograft tumor tissue and cultured cell lines. cDNA synthesis from 1 μg of total RNA was performed using High Capacity RNA-to-cDNA Kit (Applied Biosystems) following manufacturer's protocol. Gene-specific quantitative RT-PCR analysis of AURKA was performed with SYBR Select Master Mix (Applied Biosystems) on a StepOnePlus Real-Time PCR system and MicroAmp Fast optical 96-well reaction plate (Applied Biosystems) using synthesized 5 ng cDNA. The following primers were used: AURKA = forward, 5′-CTGCATTTCAGGACCTGTTAAGG-3′; reverse, 5′-AACGCGCTG GGAAGAATTT-3′ (40); and GAPDH = forward, 5′-AAGGTGAA GGTCGGAGTCAA-3′; reverse, 5′-AATGAAGGGGTCATTGATGG-3′. Relative mRNA expression levels against normal pediatric/adult cerebral tissues were determined by the 2−ΔΔCt method. All quantitative RT-PCR assays were performed in triplicate.

In vitro treatment with MLN8237

MLN8237, provided by Millennium Pharmaceuticals, Inc., was dissolved in DMSO for a stock solution of 10 mmol/L for in vitro experiments. To determine the time- and dose-dependent effects of MLN8237, paired pGBM monolayer and neurosphere cultures were seeded into 96-well plates and exposed to 7 doses of MLN8237 (ranging from 1 to 4,000 nmol/L) or vehicle control over a 2-week period. Cell viability was measured at day 1, 4, 7, 10, and 13 using the Cell Counting Kit-8 (CCK8; Dojindo Molecular Technologies; refs. 32, 35).

In vivo treatment of PDOX tumors with MLN8237

For in vivo experiments, MLN8237 was dissolved in 10% (2-Hydroxypropyl)-β-cyclodextrin (Sigma-Aldrich)/1% sodium bicarbonate to achieve a final concentration of 5 mg/mL. Two weeks after intracerebral tumor cell implantation, mice were orally administered MLN8237 (30 mg/kg/day; refs. 19, 23, 41, 42) once daily for 12 days. To determine any survival benefits from MLN8237 treatment, the mice were monitored daily until they developed signs of neurologic deficit or became moribund, at which time they were euthanized and their brains were removed for analysis. To test the biologic effects of MLN8237 at the end of treatment, mice were treated with MLN8237 (30 mg/kg/day), a dose frequently used before in animal experiments (19, 23, 41, 42) for 12 days once initial neurologic symptoms caused by GBM growth were observed; these mice were euthanized 1, 24, and 72 hours after the last MLN8237 dose, and their brains were removed for analysis. Plasma was collected from all euthanized mice.

Flow cytometry (FCM)

Flow cytometry was performed as we described previously (32, 35, 36, 38). To examine the changes in cell-cycle distribution, tumor cells were washed and incubated at 37°C for 45 minutes with 10 μg/mL Hoechst 33342 (Sigma-Aldrich) and Verapamil (ATP pump inhibitor, 50 μmol/L) in FCM buffer (Dulbecco's PBS, 0.5% BSA, and 2 mmol/L EDTA) for DNA staining following incubation with 0.5 μg/mL PyroninY (Sigma-Aldrich) for another 45 minutes to stain RNA. To exclude mouse cells from analysis, xenograft cells were stained with a cocktail of APC-conjugated antibodies specific to mouse cell surface antigen (CD90.1, CD133, CD140a, and CD24) together with matching APC-conjugated isotype control antibodies (Mouse IgG1 κ, Rat IgG2a κ, and Rat IgG2b κ; BioLegend, Inc.) to set the gate. To analyze the putative CSCs, tumor cells were washed and labeled with APC-conjugated anti-human CD133 (Miltenyi Biotec Inc) and FITC-conjugated anti-human CD15 (Miltenyi Biotec Inc) for 15 minutes on ice in 100 μL of FCM buffer containing FcR blocking reagent (32, 35, 36, 38). Anti-mouse IgG2b-APC and anti-mouse IgM-FITC antibodies (Miltenyi Biotec Inc) were also included as isotype controls. After washing, cells were resuspended in FCM buffer containing 2 μg/mL PI to gate out dead cells and analyzed with a LSR II flow cytometer and Kaluza Analysis Software Version 1.3 (Beckman Coulter, Inc.).

MLN8237 concentration in plasma, tumor, and normal brain

MLN8237 concentrations were determined using an Applied Biosystems API5000 mass spectrometer equipped with a Shimadzu Nexera UFLC System and a LEAP autosampler following an optimized method at Takeda Pharmaceuticals. A reverse-phase gradient method with flow rate of 0.4 mL/minute on a Phenomenex Synergi Polar-RP column was used for analyte separation. The mobile phases used were water and acetonitrile (supplemented with formic acid 0.1% volume-to-volume). MLN8237 was ionized under a positive ion spray mode and detected through the multiple-reaction monitoring of a mass transition pair at a mass-to-charge ratio of 519.1/328.1. Calibration curves of MLN8237 were established using standards, and the peak area ratios of the analyte against an isotopically labeled internal standard were used to quantify samples. Using a plasma volume of 50 μL, linearity was achieved in the MLN8237 concentration range of 1.93–3860 nmol/L. In addition, dilution linearity was also assessed to ensure that study samples above the upper limit of the standard curve could be diluted with blank matrix without affecting the final calculated concentration. This was performed by preparing dilution QCs at 19,300 nmol/L and diluting down to the curve range by 50-fold with blank mouse plasma. The triplicate dilution QCs have met the acceptance criteria of within 15% with respect to accuracy and precision. This approach validated the dilution cap of accurately quantifying samples that are AQL (above quantitation limit) up to 193,00 nmol/L. All data were acquired and processed using Analyst 1.6.2 software (Applied Biosystems).

Western hybridization

Proteins were extracted in M-PER Mammalian Protein Extraction Reagent (Life Technologies) including Protease Inhibitor Cocktail, separated with SDS-PAGE in a 4%–12% Bis-Tris gel, transferred to nitrocellulose membrane, and incubated with the primary antibodies: Anti-Aurora A (1:1,000, Cell Signaling Technology, catalog no. 4718), anti-PARP (recognizes both the full and the cleaved fragment, 1:500, Calbiochem, catalog no. AM30), anti-caspase-3 (recognizes both the full and the cleaved fragments, 1:1,000, Cell Signaling Technology, catalog no. 9662), and anti-β-actin (1:5,000, Sigma-Aldrich, catalog no. A2228). Following incubation with IRDye-conjugated secondary antibody (1:10,000, LI-COR Biosciences), proteins were detected and quantitated using an Odyssey Infrared Imaging System (LI-COR Biosciences) application Software version 3.0.

IHC staining

IHC staining was performed using a Vectastain Elite ABC kit Rabbit IgG (Vector Laboratories) or Mouse on Mouse (M.O.M.) Elite Peroxidase Kit (Vector Laboratories) on 5-μm paraffin sections of whole mouse brains as described previously (32, 35, 36, 43). After antigen retrieval with Target Retrieval Solution (Dako North America, Inc.) in a pressure cooker, sections were blocked with horse normal serum or M.O.M. Mouse IgG Blocking Reagent. Primary antibodies utilized were full PARP (1:100; Sigma-Aldrich), cleaved PARP (1:100), full caspase-3 (1:50), cleaved caspase-3 (1:100), AURKA (1:50; Cell Signaling Technology, Inc.), and Ki67 (1:50) (Abcam). After slides were incubated with primary antibodies, appropriate biotinylated secondary antibodies were applied, and the final signal was developed using 3, 3′-diaminobenzidine substrate kit for peroxidase. Negative control was performed by replacing primary antibodies with Dulbecco's PBS. IHC staining was assessed by combining intensity, scored as negative (−), low (+), medium (++), or strongly positive (+++), with extent of immunopositivity (percentage of positive cells). For AURKA, the IHC H-score was calculated as 3 × percentage of strong (+++) positive cells + 2 × medium (++) positive cells + 1 × low (+) positive cells (44).

Statistical analysis

Values were presented as mean ± SD. The effect on cell proliferation was analyzed with two-way ANOVA, and in vitro cell cycle analyzed with one-way ANOVA followed by a multiple comparison procedure (Holm–Sidak method). Animal survival times were compared through log-rank analysis. In vivo cell cycle and CSC populations were analyzed with pair-wise t test. All statistical analysis was performed by using SigmaStat 3.5 (Systat Software, Inc.). P < 0.05 was considered statistically significant.

AURKA was overexpressed in pGBM tumor, PDOX models, and cultured cells

To evaluate AURKA mRNA expression in pGBMs, qRT-PCR was performed on patient samples from 11 pediatric LGGs, 14 pGBMs, and 7 normal cerebral tissues. Consistent with previous observations (17), AURKA mRNA was significantly overexpressed in pGBM with a mean tumor/normal ratio of 12.50 ± 7.98 compared with LGG (1.67 ± 0.68) and normal cerebral tissue (1.60 ± 1.71; P < 0.001; Fig. 1A). Subsequent evaluation of 10 PDOX pGBM mouse models and 3 pairs of patient-matched monolayer and neurosphere cultures derived from PDOX models of pGBM confirmed the maintenance of AURKA mRNA overexpression in xenograft tumors (up to 5 in vivo passages; Fig. 1B) and in the cultured pGBM cells (Fig. 1C).

Figure 1.

AURKA is overexpressed in tumors from patients with pGBM and in pGBM xenografts and cell lines. A, Levels of AURKA mRNA expression in pediatric LGG and pGBM patient tumor (Pt) samples normalized to normal cerebra. B, AURKA mRNA expression in patient and PDOX tumor samples of pGBM at various in vivo passages. C, AURKA mRNA expression in xenograft-derived paired monolayer (Mono) and neurosphere (NS) lines quantified by real-time PCR. Relative levels of AURKA were calculated with the AURKA/GAPDH ratio observed in normal cerebral tissues as 1. †, P < 0.001 in pGBM as compared with normal cerebral tissue and LGG. *, P < 0.05; **, P < 0.01 as compared with patient (Pt). There were no significant differences between neurospheres and monolayer cells in the three models.

Figure 1.

AURKA is overexpressed in tumors from patients with pGBM and in pGBM xenografts and cell lines. A, Levels of AURKA mRNA expression in pediatric LGG and pGBM patient tumor (Pt) samples normalized to normal cerebra. B, AURKA mRNA expression in patient and PDOX tumor samples of pGBM at various in vivo passages. C, AURKA mRNA expression in xenograft-derived paired monolayer (Mono) and neurosphere (NS) lines quantified by real-time PCR. Relative levels of AURKA were calculated with the AURKA/GAPDH ratio observed in normal cerebral tissues as 1. †, P < 0.001 in pGBM as compared with normal cerebral tissue and LGG. *, P < 0.05; **, P < 0.01 as compared with patient (Pt). There were no significant differences between neurospheres and monolayer cells in the three models.

Close modal

MLN8237 inhibited cell proliferation in vitro in neurospheres and monolayer cells

To evaluate AURKA as a potential therapeutic target in pGBM, we examined in vitro effects of MLN8237 using cell cultures derived from 3 PDOX models (Table 1) representing different clinical timepoints of disease progression, that is, previously untreated (IC-4687GBM), recurrent following treatment (IC-3752GBM), and a progressive, terminal stage obtained at autopsy (IC-R0315GBM). Recognizing the role of CSCs in the progression and recurrence of GBM (45), stem cell–enriched neurosphere cell lines were treated together with the matching monolayer cells to MLN8237 (1–4,000 nmol/L).

In CSC-enriched neurospheres, MLN8237 showed significant inhibition of growth at nanomolar concentrations in all three lines in a time- and dose-dependent manner (Fig. 2A; Supplementary Fig. S1). Neurospheres from previously untreated patient tumor (IC-4687GBM) were the most sensitive to treatment with MLN8237, demonstrating significant suppression of cell proliferation after treatment with 15.6 nmol/L of MLN8237 from day 4 (28.2% ± 6.7%) through day 13 (>90%). In the recurrent model IC-3752GBM, the cells were more resistant to treatment as 62.5 nmol/L of MLN8237 led to 45.9% ± 6.2% growth suppression on day 7 and 75 ± 0.6% inhibition on day 13. In the autopsy/terminal model IC-R0315GBM, treatment with 62.5 nmol/L of MLN8237 resulted in >95% suppression of cell proliferation from day 7 to 13 (P < 0.05).

Figure 2.

Antiproliferative effects of MLN8237 through apoptosis in pGBM cell lines. A, Cells were exposed to varying concentrations of MLN8237 through day 13, and antitumor effects were examined by CCK8 assay in paired monolayer (Mono) and neurosphere (NS) in IC-4687GBM, IC-R0315GBM, and IC-3752GBM (2,000 cells/well). B and C, Paired monolayer (Mono) and neurosphere (NS) pGBM cell lines were treated with MLN8237 (62.5 nmol/L) for time-course and dose–response analyses at day 7. DNA was stained with Hoechst 33342 and analyzed with FCM. B, Representative DNA/RNA profiles in 3752GBM, time course at MLN8237 dose of 62.5 nmol/L. C, Percentage of sub-G1 phase. *, P < 0.01 compared with 0 nmol/L. D, Western hybridization of AURKA, PARP, and caspase-3 with β-actin as loading control for time-course treatment with 62.5 nmol/L of MLN8237.

Figure 2.

Antiproliferative effects of MLN8237 through apoptosis in pGBM cell lines. A, Cells were exposed to varying concentrations of MLN8237 through day 13, and antitumor effects were examined by CCK8 assay in paired monolayer (Mono) and neurosphere (NS) in IC-4687GBM, IC-R0315GBM, and IC-3752GBM (2,000 cells/well). B and C, Paired monolayer (Mono) and neurosphere (NS) pGBM cell lines were treated with MLN8237 (62.5 nmol/L) for time-course and dose–response analyses at day 7. DNA was stained with Hoechst 33342 and analyzed with FCM. B, Representative DNA/RNA profiles in 3752GBM, time course at MLN8237 dose of 62.5 nmol/L. C, Percentage of sub-G1 phase. *, P < 0.01 compared with 0 nmol/L. D, Western hybridization of AURKA, PARP, and caspase-3 with β-actin as loading control for time-course treatment with 62.5 nmol/L of MLN8237.

Close modal

In monolayer cell lines, only the IC-4687GBM cells (previously untreated) were sensitive to MLN8237, but it required a higher dose (250 nmol/L) and longer exposure (day 7) to achieve >90% inhibition. Monolayer cells derived from the recurrent and terminal GBM models were resistant to MLN8237, and <50% inhibition was achieved even with 4,000 nmol/L for 13 days. Worthy of note is that monolayer cells from IC-4687GBM proliferated much faster than the other two models, which may partially explain their early responses to MLN8237. Overall, these data identified the treatment naïve IC-4687GBM as the most responsive tumor model and revealed differential responses between neurosphere and monolayer cells in the recurrent and terminal pGBM models.

MLN8237 induced apoptosis in pGBM cells in vitro

To examine the drug's mechanism of action, first we examined apoptosis in vitro through FCM. To examine time–course responses, pGBM cells were treated with MLN8237 at 62.5 nmol/L, a dose active in neurospheres from all three models, for 1–7 days. In neurospheres, significantly elevated apoptosis (% of cells in sub-G1; P < 0.05) was observed on day 2 (20%) in IC-4687GBM, day 4 (40%) in IC-3752GBM, and day 1 (18%) in IC-R0315GBM. The levels of apoptosis increased over time and peaked on day 7 reaching 65%–75% (Fig. 2C). In monolayer cells, only moderate apoptosis was noted in IC-3752GBM (18% on day 1, 25% on day 7). In agreement with these data, Western hybridization demonstrated similar results with time-dependent cleavage of PARP detected in all neurosphere cultures but minimal effects observed in the monolayer cells, including a minor effect on caspase-3 (Fig. 2D). To examine dose responses, cells were treated with MLN8237 (4, 62.5, and 1,000 nmol/L; Fig. 2C) and analyzed by FCM on day 7 of treatment, a point at which growth inhibition was observed in the CCK8 assays. In neurospheres, a significant increase in the sub-G1 population was observed on day 7 with 4 nmol/L of MLN8237 in IC-4687GBM (15%), and treatment with 62.5 nmol/L and higher resulted in more significant increases (60%–90%) in apoptosis in all three models. However, in monolayer cells, higher drug doses (e.g., 1,000 nmol/L in IC-4687GBM) were required to induce even lower levels of apoptosis (e.g., 60% in IC-4687GBM; Fig. 2C). Overall, MLN8237 induced significant apoptosis in pGBM cells, particularly in the neurospheres.

Next, we analyzed changes in cell-cycle distribution in viable tumor cells in vitro by excluding the sub-G1 population (Fig. 2B). In neurospheres, a time-course treatment with 62.5 nmol/L of MLN8237 revealed early (day 1–2) reduction of G0–G1 phase cells accompanied by accumulation of G2–M phase cells in all three models (Supplementary Fig. S2A, right). Prolonged treatment to day 7 resulted in release of G2–M blockage and an increase in G0–G1 phase cells. Analysis of dose responses on day 7 failed to detect dose-dependent G2–M block (Supplementary Fig. S2A, left). In monolayer cells, time-course analysis detected G0–G1 reduction and G2–M block in IC-3752GBM (recurrent) and IC-R0315GBM (autopsy) cells, but not in IC-4687GBM (treatment naïve; Supplementary Fig. S2A, right). Also, IC-3752GBM was the only model that displayed a dose-dependent G0–G1 decrease (from 80% to 38%) and G2–M blockage (from 10% to 60%) at 1,000 nmol/L (Supplementary Fig. S2A, left). These data indicated that MLN8237 was able to induce short-term (1–2 days) G2–M blockage in pGBM neurospheres but failed to trigger major responses in monolayer cells from the three models.

MLN8237 effectively crosses the BBB in PDOX models of pGBM

One major hurdle in the treatment of CNS tumors is the necessity for drugs to cross the BBB. Before we launch a large-scale in vivo efficacy examination, we examined the CNS penetration capability of MLN8237 in a responsive pGBM by treating IC-4687GBM with MLN8237 for 12 days (30 mg/kg/day; Fig. 3A). Drug levels in xenograft tumors were compared with levels in the neighboring “normal” mouse brains and in the serum (Fig. 3B). In plasma, MLN8237 was detected at a concentration of 6,636 ± 1,687 nmol/L (n = 6) 1 hour following the completion of treatment and decreased to 1,126 ± 546 nmol/L at 24 hours (n = 4) and to undetectable levels (<1 nmol/L) at 72 hours (Fig. 3B). Compared with levels of MLN8237 in the normal mouse brain tissues (397 ± 100 nmol/L; n = 2) at 1 hour posttreatment and (45 ± 4 nmol/L) (n = 2) at 24 hours posttreatment, MLN8237 accumulated in tumor tissue to 615 nmol/L (n = 1) at 1 hour and remained elevated at 90 nmol/L (n = 1) at 24 hours posttreatment (Fig. 3B). Worthy of note is that this tumor is highly invasive. Therefore, the normal tissues harvested from the tumor-bearing mice may contain small amount of invasive tumor cells. The significantly elevated drug concentration in the tumor mass indicated that MLN8237 was able to penetrate the BBB and accumulate in IC-4687GBM. Compared with serum concentrations achieved in adults in a phase I trial (approximately 5,000 nmol/L ± 1,800 nmol/L, 7 days at 100 mg/kg, twice daily), the serum concentration in our animals are slightly higher (27).

Figure 3.

MLN8237 treatment significantly prolongs animal survival in a therapy-naïve IC-4687GBM model but not in a therapy-resistant model IC-R0315GBM. A, Treatment schema showing the grouping and timing of MLN8237 treatment. For the survival group, MLN8237 treatment (30 mg/kg/day, oral gavage × 12 days) was started two weeks posttumor implantation. When mice showed signs of neurologic deficit (or became moribund), their brains were harvested and tumor tissues were collected (referred to as the recurrent tumor). For the biology group, the MLN8237 treatment (30 mg/kg/day, oral gavage × 12 days) was withheld for four weeks (to allow the xenograft tumors to grow bigger to meet the need of biological assays), and the mouse brains harvested at the end of treatment (referred to as the End of Treatment sample). B,In vivo examination of MLN8237 concentration in mice bearing IC-4687GBM. Plasma, tumor, and normal brain were obtained from the biology group 1–72 hours post the last oral administration of MLN8237 and analyzed for MLN8237 concentration. C, Log-rank analysis of animal survival times. Mice (n = 10–15/group) bearing IC-4687GBM or IC-R0315GBM were treated with MLN8237 (MLN8237) and compared with those administered with vehicle (Vehicle). D, Representative IHC images of Ki-67, cleaved PARP, cleaved caspase-3, and H&E in IC-4687GBM (magnification, ×40). E, MLN8237-induced apoptosis was detected as sub-G1 in IC-4687GBM. DNA were stained with Hoechst 33342 followed by mouse antibody cocktail staining to gate out mouse cells. F, MLN8237 treatment decreases cell density in IC-4687GBM. Cell number/field was counted under ×40 magnification. *, P < 0.001 compared with vehicle. †, P < 0.001 compared with end of treatment. G, Representative IHC images of AURKA showing resistant tumor cells expressed low levels of AURKA in the recurrent tumor of IC-4687GBM (magnification, ×40).

Figure 3.

MLN8237 treatment significantly prolongs animal survival in a therapy-naïve IC-4687GBM model but not in a therapy-resistant model IC-R0315GBM. A, Treatment schema showing the grouping and timing of MLN8237 treatment. For the survival group, MLN8237 treatment (30 mg/kg/day, oral gavage × 12 days) was started two weeks posttumor implantation. When mice showed signs of neurologic deficit (or became moribund), their brains were harvested and tumor tissues were collected (referred to as the recurrent tumor). For the biology group, the MLN8237 treatment (30 mg/kg/day, oral gavage × 12 days) was withheld for four weeks (to allow the xenograft tumors to grow bigger to meet the need of biological assays), and the mouse brains harvested at the end of treatment (referred to as the End of Treatment sample). B,In vivo examination of MLN8237 concentration in mice bearing IC-4687GBM. Plasma, tumor, and normal brain were obtained from the biology group 1–72 hours post the last oral administration of MLN8237 and analyzed for MLN8237 concentration. C, Log-rank analysis of animal survival times. Mice (n = 10–15/group) bearing IC-4687GBM or IC-R0315GBM were treated with MLN8237 (MLN8237) and compared with those administered with vehicle (Vehicle). D, Representative IHC images of Ki-67, cleaved PARP, cleaved caspase-3, and H&E in IC-4687GBM (magnification, ×40). E, MLN8237-induced apoptosis was detected as sub-G1 in IC-4687GBM. DNA were stained with Hoechst 33342 followed by mouse antibody cocktail staining to gate out mouse cells. F, MLN8237 treatment decreases cell density in IC-4687GBM. Cell number/field was counted under ×40 magnification. *, P < 0.001 compared with vehicle. †, P < 0.001 compared with end of treatment. G, Representative IHC images of AURKA showing resistant tumor cells expressed low levels of AURKA in the recurrent tumor of IC-4687GBM (magnification, ×40).

Close modal

MLN8237 prolonged animal survival times through induction of apoptosis

We next examined the activity of MLN8237 in vivo and assessed whether in vitro activity predicted the in vivo therapeutic efficacy. Specifically, we examined whether effective targeting of CSC-enriched neurospheres (38, 46–49) was sufficient to predict significant extension of animal survival times or whether simultaneous targeting of both neurospheres and monolayers was required. Mice bearing IC-4687GBM (previously untreated pGBM with monolayer and neurosphere MLN8237 sensitivity) and IC-R0315GBM (terminal pGBM with only neurosphere MLN8237 sensitivity) were orally treated with MLN8237 (30 mg/kg/day for 12 days) 2 weeks after tumor implantation. Animal survival times (median) in IC-4687GBM were significantly prolonged from 53 days in the vehicle group to 78 days in the treatment group (P = 0.022; Fig. 3C). However, in the autopsy model IC-R0315GBM, the median survival time was 41 days in vehicle group and 42 days in the MLN8237-treated group (P = 0.859). While these data suggested that simultaneous suppression of both neurospheres and monolayer cells correlates with improved therapeutic efficacy, in vitro suppression of neurospheres alone was insufficient to extend animal survival times, additional studies are needed in more tumor models to validate the power of paired monolayer and neurosphere cultures in predicting in vivo success of drug treatment.

As our in vitro data showed that MLN8237 induced apoptosis, we validated the role of apoptosis in vivo in mice 1 hour post-drug treatment (acute response) and in the recurrent tumors in the survival group (long-term/delayed effects). In the responsive model IC-4687GBM, MLN8237 (30 mg/kg/day for 12 days by gavage) resulted in significant increases in apoptosis (1 hour after completion of treatment) as evidenced by increased cleaved PARP (from + in ≤25% cells to +++ in nearly 75% of cells), cleaved caspase-3 (from ++ in ≤25% cells to +++ in 50%–75% cells; Fig. 3D; Table 2), and accumulation of cells in the sub-G1 phase from pretreatment (3.6% ± 0.4%) to 1 hour after completion of treatment (10.4% ± 1.9%; Fig. 3E). As anticipated, in the cells from recurrent tumor (harvested in the survival group), cleaved PARP and cleaved caspase-3 decreased to low levels similar to baseline (+ in ≤25% cells and sub-G1 to 3.1% ± 1.5%). Parallel to the increased apoptosis, changes in histologic features, such as decreased cell density (Fig. 3F) and increase of cell size (Fig. 3D, H&E staining), were also noted. In the autopsy model IC-R0315GBM, elevated cleavage of PARP and caspase-3 were only found in <25% cells. In all the tumor cells in both models, a high level (+++) expression of full PARP and caspase-3 was maintained, suggesting that surviving cells had functional apoptotic machinery. Cell proliferation (Ki-67) was not significantly altered at any timepoints in either model.

Table 2.

Summary of IHC characteristics of tumors from xenografts treated with MLN8237

IC-4687GBMIC-R0315GBM
MoleculeTargetVehicleAt the end of treatmentRecurrentVehicleRecurrent
Ki-67 Cell proliferation 50% 60% 20%–50% 35%–50% 40%–50% 
Full PARP Apoptosis ++ 4 +++ 4 +++ 4 +++ 4 +++ 4 
Cleaved PARP Apoptosis + 1 +++ 3 + 1 + 1 +++ 1 
Full caspase-3 Apoptosis +++ 4 +++ 4 +++ 4 +++ 4 +++ 4 
Cleaved caspase-3 Apoptosis ++ 1 +++ 3 + 1 ++ 1 +++ 1 
IC-4687GBMIC-R0315GBM
MoleculeTargetVehicleAt the end of treatmentRecurrentVehicleRecurrent
Ki-67 Cell proliferation 50% 60% 20%–50% 35%–50% 40%–50% 
Full PARP Apoptosis ++ 4 +++ 4 +++ 4 +++ 4 +++ 4 
Cleaved PARP Apoptosis + 1 +++ 3 + 1 + 1 +++ 1 
Full caspase-3 Apoptosis +++ 4 +++ 4 +++ 4 +++ 4 +++ 4 
Cleaved caspase-3 Apoptosis ++ 1 +++ 3 + 1 ++ 1 +++ 1 

NOTE: Scored intensity as negative (−), low (+), medium (++), and strongly positive (+++) and extent of immunopositivity as 0 = negative; 1 = 1%–25%; 2 = 26%–50%; 3 = 51%–75%; 4 = >75% positive cells.

Tumor cells lacking AURKA expression survived MLN8237 treatment in vivo

Despite significant prolongation of survival times following MLN8237 treatment in IC-4687GBM mice, all animals in our study ultimately succumbed to their tumors. To understand the mechanisms of therapy resistance, we examined changes of AURKA (molecular target of MLN8237) through IHC and evaluated as H-score, calculated as 3× percentage of strong (+++) positive cells + 2× medium (++) positive cells + 1× low (+) positive cells (ref. 44; Supplementary Fig. S3A). Compared with vehicle-treated IC-4687GBM tumors with H-score 19.8 ± 4.8 for AURKA, it decreased to 8.8 ± 4.4 (P < 0.001) with many areas lacking AURKA expression in recurrent tumors. There was no change in the H-score at the end of treatment tumor (22.4 ± 9.4) compared with vehicle-treated IC-4687GBM. In the resistant model IC-R0315GBM, cells with strong (+++) or medium (++) positivity decreased while low (+) positive cells slightly increased in the recurrent tumor (Supplementary Fig. S3B). These data suggest that pGBM tumor cells with low or no AURKA expression escaped MLN8237-induced cell killing and contributed to progression and recurrence of xenograft tumors in these two pGBM models, although data from additional models are needed to draw a definitive conclusion.

In vivo tumor progression was associated with persistence of CD133+ cells

Next, we examined the role of CSCs using FCM analysis of dual-stained CD133+ and CD15+ cells, the two most commonly utilized cell surface markers for brain tumor stem cells in both neurospheres and xenografts. In vitro, CD133+CD15 cells represented the major population of neurosphere cells derived from both IC-4687GBM (36.1%) and IC-R0315GBM (57.2%; Fig. 4A). Treatment with MLN8237 (4–62.5 nmol/L) for 7 days resulted in a minor shift (≤7%) of the mono (CD133+CD15 and CD133CD15+) and dual (CD133+CD15+) subpopulations, suggesting that all subpopulations were similarly, if not equally, sensitive to MLN8237 treatment. In vivo in the MLN8237-responsive model (IC-4687GBM), we detected an increase of mono-CD133+ (CD133+CD15) cells from 9.5% ± 5.9% in vehicle-treated tumors to 28.2 ± 22.2% (P > 0.05) in tumors harvested 1 hour after MLN8237 treatment (30 mg/kg × 12 days), and we observed a further increase to 52.3% ± 12.8% (P < 0.05) in the recurrent tumors (Fig. 4B). These changes were accompanied by a steady decrease of CD15-mono positive (CD133CD15+) cells (P = 0.055), whereas changes of dual-positive (CD133+CD15+) cells were not significant (Fig. 4B). Progressive tumors from the MLN8237-resistant model (IC-R0315GBM) showed similar enrichment of CD133+ cells (69.8% ± 8.6% in vehicle and 80.8% ± 0.0% in MLN8237-treated group; P < 0.05). However, the majority of the xenograft CD133+ cells in IC-R0315GBM coexpressed CD15 (59.4% ± 7.6% in vehicle and 76.2% ± 0.0% in MLN8237-treated group; Fig. 4B, bottom), which was significantly different from the neurospheres that were predominantly CD133-mono–positive (CD133+CD15; Fig. 4A, bottom). In summary, these data suggest that the putative CSC population in vitro in cultured neurosphere cells did not have identical responsiveness toward MLN8237 as the CSCs in xenograft tumors, and CD133+ cell were the cause of therapy resistance because nearly all the CD15+ cells were dual positive with CD133.

Figure 4.

Effects of MLN8237 on the profile of putative CSCs in vitro and in vivo. A,In vitro dose response of CD133+ and/or CD15+ cells in IC-4687GBM (4687) and IC-R0315GBM (R0315) treated with MLN8237 (4–62.5 nmol/L) for 7 days. B, Representative FCM profiles (left) and graphs of quantitative analysis (right) of in vivo changes of CD133+ and/or CD15+ cells. The vehicle-treated xenografts (Vehicle) of IC-4687GBM (4687) and IC-R0315GBM (R0315) were compared with the recurrent tumors (Recurrent) that were harvested when the animals in the survival group were euthanized. (*, P = 0.013).

Figure 4.

Effects of MLN8237 on the profile of putative CSCs in vitro and in vivo. A,In vitro dose response of CD133+ and/or CD15+ cells in IC-4687GBM (4687) and IC-R0315GBM (R0315) treated with MLN8237 (4–62.5 nmol/L) for 7 days. B, Representative FCM profiles (left) and graphs of quantitative analysis (right) of in vivo changes of CD133+ and/or CD15+ cells. The vehicle-treated xenografts (Vehicle) of IC-4687GBM (4687) and IC-R0315GBM (R0315) were compared with the recurrent tumors (Recurrent) that were harvested when the animals in the survival group were euthanized. (*, P = 0.013).

Close modal

In this report, we identified AURKA as a potential therapeutic target in a subset of pGBM by validating its overexpression in patient tumors, PDOX models, and matching pairs of cultured neurosphere and monolayer cultures of pGBM. We demonstrated strong antitumor activity of the AURKA inhibitor MLN8237 in vitro and in vivo and identified variable responses in pGBM cells derived from different stage of clinical progression (treatment naïve, recurrence, and terminal/autopsy) and between matching pairs of neurospheres and monolayer cultures. While our data suggested that successful inhibition in both neurosphere and monolayer cells correlates with extension of animal survival times, additional studies involving more tumor models are needed to further test the “adherent versus spheroid” hypothesis and to correlate with the frequent molecular changes. Mechanistically, we confirmed apoptosis as the primary cause of MLN8237-induced cell death and suggested the lack of AURKA expression and the expression of CSC marker CD133 as causes of therapy resistance. These findings also support a secondary transplantation of the treated (resistant) tumor cells followed by another round of AURKA inhibitor treatment to confirm the sustained resistance of the AURKA-negative and/or CD133+ cells toward AURKA inhibition in pGBMs.

Overexpression of AURKA and preclinical antitumor activity of AURKA inhibitors have been reported extensively in various human cancers. AURKA inhibitor MLN8237 (alisertib) is orally available and has exhibited strong antitumor activity in a wide range of human cancers and has a well-established clinical safety profile (26–30, 50, 51). The advantage of preclinical testing of therapeutic efficacy in PDOX models of pGBM, a disease that desperately needs new therapies, is that in vivo findings can potentially be rapidly translated into clinical trials. In an effort to facilitate the initiation of clinical trials, we address two important issues. First, we examined the treatment effects in samples from different stages of clinical progression and how in vitro models predict in vivo efficacy. Using our unique panel of PDOX models of pGBM, we examined the impact of clinical timepoint of the original patient tumor on the overall response to treatment with MLN8237. Although these models were not derived from the same patients (which is highly desired but very difficult to accomplish), testing new therapies in models representing untreated, recurrent, and terminal/autopsy pGBMs can provide important clues for future selection of patients. Our findings that previously treated recurrent and autopsy pGBM models responded differently to MLN8237 compared with the treatment-naïve tumor model suggests a need for a broader range of tumor models in future preclinical drug testing.

Second, as in vivo evaluation of therapeutic efficacy can be both time consuming and costly, in vitro prescreening assays that can better predict in vivo success are highly desired. Unlike traditional monolayer cells, neurospheres are enriched with CSCs, making them frequently favored over monolayer cells for in vitro preclinical drug testing (22, 52). Using AURKA-overexpressing neurosphere and monolayer pGBM lines, we found that MLN8237 was effective in reducing growth in all three neurosphere cultures enriched with putative glioma stem (CD133+ and CD15+) cells; these results are similar to preclinical findings in adult GBMs (21, 52). Unexpectedly, monolayer cultures were less responsive to MLN8237 treatment despite expressing similarly high levels of AURKA. Our data demonstrate that tumor cells maintained in these two growth conditions did not always respond identically to the same treatment, and that when MLN8237 effectively suppressed both neurosphere and monolayer cells in vitro, the xenograft models demonstrated the most significant in vivo drug responses. These data support the inclusion and testing of both monolayer and neurosphere cells in future drug development to validate the predictability of this strategy.

Similar to many previous studies on the molecular mechanisms of action of MLN8237 (11, 19, 22, 53–56), we demonstrated that apoptosis and cell-cycle arrest in G2–M phase mediated the antitumor activity in pGBM both in vitro and in vivo. Despite the significantly prolonged animal survival time in IC-4687GBM, all the animals in the treatment group died of disease. To further understand the mechanisms of therapy resistance, we analyzed the protein expression of AURKA, the target of MLN8237, in the remnant/recurrent xenograft tumors through IHC. Not surprisingly, the number of AURKA-positive cells was dramatically decreased in the remnant/recurrent tumors, suggesting that 12 days of treatment with MLN8237 successfully eliminates AURKA-positive tumor cells, and implying that the AURKA-negative tumor cells that escaped MLN8237 treatment caused tumor recurrence/progression. This finding supports additional studies to establish the role of AURKA levels in determine drug responsiveness, for example, by showing that knockdown of AURKA levels in vitro results in a shift of drug potency. As high expression of full PARP and caspase-3 was detected in the remnant/recurrent tumors, these residual tumor cells may have functional apoptotic machinery that can be targeted in the future development of combination therapy with MLN8237 (22, 57–61).

In addition, our study of the mechanisms of resistance provided new insights on the role of CSCs in MLN8237 resistance. In IC-4687GBM, although CD133 and CD15 (both mono-and dual-positive) cells were killed by MLN8237 in neurospheres in vitro, the fraction of CD133-mono–positive cells increased in the remnant/recurrent xenografts in vivo. In IC-R0315GBM, while CSCs in neurospheres were predominately (>50%) composed of CD133-mono–positive (CD133+CD15) cells, the xenografts were dual positive (CD133+CD15+) cells (59.4% before treatment and 76.2% in recurrent tumor). Such cellular differences, likely caused by changes in microenvironment, may have contributed to the differential responses observed in this terminal-stage pGBM model. This finding suggests that serum-free cell culture may favor the growth of a subpopulation of stem cells, which may not accurately reflect in vivo behavior. Our data shows that some pGBM cells expressing CD133 (alone or with CD15) were resistant to MLN8237 in vivo, and new strategies capable of effectively targeting CSCs are needed to further enhance the in vivo efficacy of MLN8237.

In conclusion, we identified AURKA as a novel therapeutic target for pGBM and demonstrated the strong antitumor activity of MLN8237 in vitro in neurospheres and in vivo in a treatment-naïve PDOX model. Also, this study suggested that simultaneous targeting of neurosphere and monolayer cell lines correlates with prolonged animal survival in vivo. Our data also support additional in vivo studies to confirm the loss of molecular target AURKA and increased expression of CD133 by tumor cells as the mechanisms of MLN8237 resistance in pGBM cells. Our data suggests that combination therapy is required to target AURKA-negative fractions and CD133+ cells following treatment with AURKA inhibition to further improve therapeutic efficacy.

No potential conflicts of interest were disclosed.

Conception and design:M. Kogiso, S. Zhao, H. Lindsay, P.A. Baxter, J. Muscal, X.-N. Li

Development of methodology:M. Kogiso, F.K. Braun, P.A. Baxter, X.-N. Li

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.):M. Kogiso, F.K. Braun, L. Zhang, F.Y. Lin, S. Zhao, A.M. Adesina, D. Liao, M.G. Qian, X.-N. Li

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis):M. Kogiso, L. Qi, H. Lindsay, J. Muscal, X.-N. Li

Writing, review, and/or revision of the manuscript:M. Kogiso, S.G. Injac, H. Zhang, F.Y. Lin, S. Zhao, H. Lindsay, J.M. Su, A.M. Adesina, S.L. Berg, J. Muscal, X.-N. Li

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases):M. Kogiso, L. Qi, F.K. Braun, L. Zhang, Y. Du, H. Zhang, F.Y. Lin

Study supervision:X.-N. Li

This project is funded by NIH/NCIRO1 CA185402 (to X.-N. Li), Cancer Prevention and Research Institute of Texas (CPRIT) RP150032 (to X.-N. Li), St Baldrick's Foundation (to J.M. Su), and by the Cytometry and Cell Sorting Core at The Baylor College of Medicine with funding from the NIH (P30 AI036211, P30 CA125123, and S10 RR024574), Texas Children's Hospital Pediatric Pilot Award (to J. Muscal), Kappa Alpha Theta Foundation Research Scholar (J. Muscal), and Alex's Lemonade Stand Center of Excellence (to S.L. Berg).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Stupp
R
,
Hegi
ME
,
Gilbert
MR
,
Chakravarti
A
. 
Chemoradiotherapy in malignant glioma: standard of care and future directions
.
J Clan Uncool
2007
;
25
:
4127
36
.
2.
Fangusaro
J
,
Warren
KE
. 
Unclear standard of care for pediatric high grade glioma patients
.
J Neurooncol
2013
;
113
:
341
2
.
3.
Dolecek
TA
,
Propp
JM
,
Stroup
NE
,
Kruchko
C
. 
CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005–2009
.
Neuro Oncol
2012
;
14
Suppl 5
:
v1
49
.
4.
Marumoto
T
,
Zhang
D
,
Saya
H
. 
Aurora-A - a guardian of poles
.
Nat Rev Cancer
2005
;
5
:
42
50
.
5.
D'Assoro
AB
,
Haddad
T
,
Galanis
E
. 
Aurora-A kinase as a promising therapeutic target in cancer
.
Front Oncolgy
2015
;
5
:
295
.
6.
Carvajal
RD
,
Tse
A
,
Schwartz
GK
. 
Aurora kinases: new targets for cancer therapy
.
Clin Cancer Res
2006
;
12
:
6869
75
.
7.
Ye
D
,
Garcia-Manero
G
,
Kantarjian
HM
,
Xiao
L
,
Vadhan-Raj
S
,
Fernandez
MH
, et al
Analysis of Aurora kinase A expression in CD34(+) blast cells isolated from patients with myelodysplastic syndromes and acute myeloid leukemia
.
J Hematop
2009
;
2
:
2
8
.
8.
Humme
D
,
Haider
A
,
Mobs
M
,
Mitsui
H
,
Suarez-Farinas
M
,
Ohmatsu
H
, et al
Aurora kinase A is upregulated in cutaneous T-cell lymphoma and represents a potential therapeutic target
.
J Invest Dermatol
2015
;
135
:
2292
300
.
9.
Ferchichi
I
,
Sassi Hannachi
S
,
Baccar
A
,
Marrakchi Triki
R
,
Cremet
JY
,
Ben Romdhane
K
, et al
Assessment of Aurora A kinase expression in breast cancer: a tool for early diagnosis?
Dis Markers
2013
;
34
:
63
9
.
10.
Siggelkow
W
,
Boehm
D
,
Gebhard
S
,
Battista
M
,
Sicking
I
,
Lebrecht
A
, et al
Expression of aurora kinase A is associated with metastasis-free survival in node-negative breast cancer patients
.
BMC Cancer
2012
;
12
:
562
.
11.
Zhou
N
,
Singh
K
,
Mir
MC
,
Parker
Y
,
Lindner
D
,
Dreicer
R
, et al
The investigational Aurora kinase A inhibitor MLN8237 induces defects in cell viability and cell-cycle progression in malignant bladder cancer cells in vitro and in vivo
.
Clin Cancer Res
2013
;
19
:
1717
28
.
12.
Sehdev
V
,
Peng
D
,
Soutto
M
,
Washington
MK
,
Revetta
F
,
Ecsedy
J
, et al
The aurora kinase A inhibitor MLN8237 enhances cisplatin-induced cell death in esophageal adenocarcinoma cells
.
Mol Cancer Ther
2012
;
11
:
763
74
.
13.
Belt
EJ
,
Brosens
RP
,
Delis-van Diemen
PM
,
Bril
H
,
Tijssen
M
,
van Essen
DF
, et al
Cell cycle proteins predict recurrence in stage II and III colon cancer
.
Ann Surg Oncol
2012
;
19
Suppl 3
:
S682
92
.
14.
Gritsko
TM
,
Coppola
D
,
Paciga
JE
,
Yang
L
,
Sun
M
,
Shelley
SA
, et al
Activation and overexpression of centrosome kinase BTAK/Aurora-A in human ovarian cancer
.
Clin Cancer Res
2003
;
9
:
1420
6
.
15.
Neben
K
,
Korshunov
A
,
Benner
A
,
Wrobel
G
,
Hahn
M
,
Kokocinski
F
, et al
Microarray-based screening for molecular markers in medulloblastoma revealed STK15 as independent predictor for survival
.
Cancer Res
2004
;
64
:
3103
11
.
16.
Barton
VN
,
Foreman
NK
,
Donson
AM
,
Birks
DK
,
Handler
MH
,
Vibhakar
R
. 
Aurora kinase A as a rational target for therapy in glioblastoma
.
J Neurosurg Pediatr
2010
;
6
:
98
105
.
17.
Lehman
NL
,
O'Donnell
JP
,
Whiteley
LJ
,
Stapp
RT
,
Lehman
TD
,
Roszka
KM
, et al
Aurora A is differentially expressed in gliomas, is associated with patient survival in glioblastoma and is a potential chemotherapeutic target in gliomas
.
Cell Cycle
2012
;
11
:
489
502
.
18.
Maris
JM
,
Morton
CL
,
Gorlick
R
,
Kolb
EA
,
Lock
R
,
Carol
H
, et al
Initial testing of the aurora kinase A inhibitor MLN8237 by the Pediatric Preclinical Testing Program (PPTP)
.
Pediatr Blood Cancer
2010
;
55
:
26
34
.
19.
Gorgun
G
,
Calabrese
E
,
Hideshima
T
,
Ecsedy
J
,
Perrone
G
,
Mani
M
, et al
A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma
.
Blood
2010
;
115
:
5202
13
.
20.
Muscal
JA
,
Scorsone
KA
,
Zhang
L
,
Ecsedy
JA
,
Berg
SL
. 
Additive effects of vorinostat and MLN8237 in pediatric leukemia, medulloblastoma, and neuroblastoma cell lines
.
Invest New Drugs
2013
;
31
:
39
45
.
21.
Mannino
M
,
Gomez-Roman
N
,
Hochegger
H
,
Chalmers
AJ
. 
Differential sensitivity of Glioma stem cells to Aurora kinase A inhibitors: implications for stem cell mitosis and centrosome dynamics
.
Stem Cell Res
2014
;
13
:
135
43
.
22.
Hong
X
,
O'Donnell
JP
,
Salazar
CR
,
Van Brocklyn
JR
,
Barnett
KD
,
Pearl
DK
, et al
The selective Aurora-A kinase inhibitor MLN8237 (alisertib) potently inhibits proliferation of glioblastoma neurosphere tumor stem-like cells and potentiates the effects of temozolomide and ionizing radiation
.
Cancer Chemother Pharmacol
2014
;
73
:
983
90
.
23.
Hill
RM
,
Kuijper
S
,
Lindsey
JC
,
Petrie
K
,
Schwalbe
EC
,
Barker
K
, et al
Combined MYC and P53 defects emerge at medulloblastoma relapse and define rapidly progressive, therapeutically targetable disease
.
Cancer Cell
2015
;
27
:
72
84
.
24.
Paugh
BS
,
Qu
C
,
Jones
C
,
Liu
Z
,
Adamowicz-Brice
M
,
Zhang
J
, et al
Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease
.
J Clin Oncol
2010
;
28
:
3061
8
.
25.
Jones
C
,
Perryman
L
,
Hargrave
D
. 
Paediatric and adult malignant glioma: close relatives or distant cousins?
Nat Rev Clin Oncol
2012
;
9
:
400
13
.
26.
Dees
EC
,
Cohen
RB
,
von Mehren
M
,
Stinchcombe
TE
,
Liu
H
,
Venkatakrishnan
K
, et al
Phase I study of Aurora A kinase inhibitor MLN8237 in advanced solid tumors: safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations
.
Clin Cancer Res
2012
;
18
:
4775
84
.
27.
Cervantes
A
,
Elez
E
,
Roda
D
,
Ecsedy
J
,
Macarulla
T
,
Venkatakrishnan
K
, et al
Phase I pharmacokinetic/pharmacodynamic study of MLN8237, an investigational, oral, selective Aurora a kinase inhibitor, in patients with advanced solid tumors
.
Clin Cancer Res
2012
;
18
:
4764
74
.
28.
Matulonis
UA
,
Sharma
S
,
Ghamande
S
,
Gordon
MS
,
Del Prete
SA
,
Ray-Coquard
I
, et al
Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma
.
Gynecol Oncol
2012
;
127
:
63
9
.
29.
Mosse
YP
,
Lipsitz
E
,
Fox
E
,
Teachey
DT
,
Maris
JM
,
Weigel
B
, et al
Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of Aurora kinase A: a Children's Oncology Group Phase I Consortium study
.
Clin Cancer Res
2012
;
18
:
6058
64
.
30.
DuBois
SG
,
Marachelian
A
,
Fox
E
,
Kudgus
RA
,
Reid
JM
,
Groshen
S
, et al
Phase I study of the Aurora A kinase inhibitor alisertib in combination with irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial
.
J Clin Oncol
2016
;
34
:
1368
75
.
31.
Hubert
CG
,
Rivera
M
,
Spangler
LC
,
Wu
Q
,
Mack
SC
,
Prager
BC
, et al
A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo
.
Cancer Res
2016
;
76
:
2465
77
.
32.
Shu
Q
,
Wong
KK
,
Su
JM
,
Adesina
AM
,
Yu
LT
,
Tsang
YT
, et al
Direct orthotopic transplantation of fresh surgical specimen preserves CD133+ tumor cells in clinically relevant mouse models of medulloblastoma and glioma
.
Stem Cells
2008
;
26
:
1414
24
.
33.
Yu
L
,
Baxter
PA
,
Zhao
X
,
Liu
Z
,
Wadhwa
L
,
Zhang
Y
, et al
A single intravenous injection of oncolytic picornavirus SVV-001 eliminates medulloblastomas in primary tumor-based orthotopic xenograft mouse models
.
Neuro Oncol
2011
;
13
:
14
27
.
34.
Lindsay
H
,
Huang
Y
,
Du
Y
,
Braun
FK
,
Teo
WY
,
Kogiso
M
, et al
Preservation of KIT genotype in a novel pair of patient-derived orthotopic xenograft mouse models of metastatic pediatric CNS germinoma
.
J Neurooncol
2016
;
128
:
47
56
.
35.
Yu
L
,
Baxter
PA
,
Voicu
H
,
Gurusiddappa
S
,
Zhao
Y
,
Adesina
A
, et al
A clinically relevant orthotopic xenograft model of ependymoma that maintains the genomic signature of the primary tumor and preserves cancer stem cells in vivo
.
Neuro Oncol
2010
;
12
:
580
94
.
36.
Liu
Z
,
Zhao
X
,
Wang
Y
,
Mao
H
,
Huang
Y
,
Kogiso
M
, et al
A patient tumor-derived orthotopic xenograft mouse model replicating the group 3 supratentorial primitive neuroectodermal tumor in children
.
Neuro Oncol
2014
;
16
:
787
99
.
37.
Zhao
X
,
Liu
Z
,
Yu
L
,
Zhang
Y
,
Baxter
P
,
Voicu
H
, et al
Global gene expression profiling confirms the molecular fidelity of primary tumor-based orthotopic xenograft mouse models of medulloblastoma
.
Neuro Oncol
2012
;
14
:
574
83
.
38.
Zhao
X
,
Zhao
YJ
,
Lin
Q
,
Yu
L
,
Liu
Z
,
Lindsay
H
, et al
Cytogenetic landscape of paired neurospheres and traditional monolayer cultures in pediatric malignant brain tumors
.
Neuro Oncol
2015
;
17
:
965
77
.
39.
Liu
Z
,
Zhao
X
,
Mao
H
,
Baxter
PA
,
Huang
Y
,
Yu
L
, et al
Intravenous injection of oncolytic picornavirus SVV-001 prolongs animal survival in a panel of primary tumor-based orthotopic xenograft mouse models of pediatric glioma
.
Neuro Oncol
2013
;
15
:
1173
85
.
40.
Ulisse
S
,
Delcros
JG
,
Baldini
E
,
Toller
M
,
Curcio
F
,
Giacomelli
L
, et al
Expression of Aurora kinases in human thyroid carcinoma cell lines and tissues
.
Int J Cancer
2006
;
119
:
275
82
.
41.
Manfredi
MG
,
Ecsedy
JA
,
Chakravarty
A
,
Silverman
L
,
Zhang
M
,
Hoar
KM
, et al
Characterization of alisertib (MLN8237), an investigational small-molecule inhibitor of Aurora A kinase using novel in vivo pharmacodynamic assays
.
Clin Cancer Res
2011
;
17
:
7614
24
.
42.
Ahmad
Z
,
Jasnos
L
,
Gil
V
,
Howell
L
,
Hallsworth
A
,
Petrie
K
, et al
Molecular and in vivo characterization of cancer-propagating cells derived from MYCN-dependent medulloblastoma
.
PLoS One
2015
;
10
:
e0119834
.
43.
Shu
Q
,
Antalffy
B
,
Su
JM
,
Adesina
A
,
Ou
CN
,
Pietsch
T
, et al
Valproic acid prolongs survival time of severe combined immunodeficient mice bearing intracerebellar orthotopic medulloblastoma xenografts
.
Clin Cancer Res
2006
;
12
:
4687
94
.
44.
Detre
S
,
Saclani Jotti
G
,
Dowsett
M
. 
A "quickscore" method for immunohistochemical semiquantitation: validation for oestrogen receptor in breast carcinomas
.
J Clin Pathol
1995
;
48
:
876
8
.
45.
Bao
S
,
Wu
Q
,
McLendon
RE
,
Hao
Y
,
Shi
Q
,
Hjelmeland
AB
, et al
Glioma stem cells promote radioresistance by preferential activation of the DNA damage response
.
Nature
2006
;
444
:
756
60
.
46.
Pavon
LF
,
Marti
LC
,
Sibov
TT
,
Malheiros
SM
,
Brandt
RA
,
Cavalheiro
S
, et al
In vitro analysis of neurospheres derived from glioblastoma primary culture: a novel methodology paradigm
.
Front Neurol
2014
;
4
:
214
.
47.
Wan
F
,
Zhang
S
,
Xie
R
,
Gao
B
,
Campos
B
,
Herold-Mende
C
, et al
The utility and limitations of neurosphere assay, CD133 immunophenotyping and side population assay in glioma stem cell research
.
Brain Pathol
2010
;
20
:
877
89
.
48.
Laks
DR
,
Masterman-Smith
M
,
Visnyei
K
,
Angenieux
B
,
Orozco
NM
,
Foran
I
, et al
Neurosphere formation is an independent predictor of clinical outcome in malignant glioma
.
Stem Cells
2009
;
27
:
980
7
.
49.
Chaichana
K
,
Zamora-Berridi
G
,
Camara-Quintana
J
,
Quinones-Hinojosa
A
. 
Neurosphere assays: growth factors and hormone differences in tumor and nontumor studies
.
Stem Cells
2006
;
24
:
2851
7
.
50.
Venkatakrishnan
K
,
Kim
TM
,
Lin
CC
,
Thye
LS
,
Chng
WJ
,
Ma
B
, et al
Phase 1 study of the investigational Aurora A kinase inhibitor alisertib (MLN8237) in East Asian cancer patients: pharmacokinetics and recommended phase 2 dose
.
Invest New Drugs
2015
;
33
:
942
53
.
51.
Kelly
KR
,
Shea
TC
,
Goy
A
,
Berdeja
JG
,
Reeder
CB
,
McDonagh
KT
, et al
Phase I study of MLN8237–investigational Aurora A kinase inhibitor–in relapsed/refractory multiple myeloma, non-Hodgkin lymphoma and chronic lymphocytic leukemia
.
Invest New Drugs
2014
;
32
:
489
99
.
52.
Van Brocklyn
JR
,
Wojton
J
,
Meisen
WH
,
Kellough
DA
,
Ecsedy
JA
,
Kaur
B
, et al
Aurora-A inhibition offers a novel therapy effective against intracranial glioblastoma
.
Cancer Res
2014
;
74
:
5364
70
.
53.
Qi
L
,
Zhang
Y
. 
Alisertib (MLN8237), a selective Aurora-A kinase inhibitor, induces apoptosis in human tongue squamous cell carcinoma cell both in vitro and in vivo
.
Tumour Biol
2015
;
36
:
1797
802
.
54.
Asteriti
IA
,
Di Cesare
E
,
De Mattia
F
,
Hilsenstein
V
,
Neumann
B
,
Cundari
E
, et al
The Aurora-A inhibitor MLN8237 affects multiple mitotic processes and induces dose-dependent mitotic abnormalities and aneuploidy
.
Oncotarget
2014
;
5
:
6229
42
.
55.
Qi
W
,
Spier
C
,
Liu
X
,
Agarwal
A
,
Cooke
LS
,
Persky
DO
, et al
Alisertib (MLN8237) an investigational agent suppresses Aurora A and B activity, inhibits proliferation, promotes endo-reduplication and induces apoptosis in T-NHL cell lines supporting its importance in PTCL treatment
.
Leuk Res
2013
;
37
:
434
9
.
56.
Qi
W
,
Cooke
LS
,
Liu
X
,
Rimsza
L
,
Roe
DJ
,
Manziolli
A
, et al
Aurora inhibitor MLN8237 in combination with docetaxel enhances apoptosis and anti-tumor activity in mantle cell lymphoma
.
Biochem Pharmacol
2011
;
81
:
881
90
.
57.
Mahadevan
D
,
Stejskal
A
,
Cooke
LS
,
Manziello
A
,
Morales
C
,
Persky
DO
, et al
Aurora A inhibitor (MLN8237) plus vincristine plus rituximab is synthetic lethal and a potential curative therapy in aggressive B-cell non-Hodgkin lymphoma
.
Clin Cancer Res
2012
;
18
:
2210
9
.
58.
Kozyreva
VK
,
Kiseleva
AA
,
Ice
RJ
,
Jones
BC
,
Loskutov
YV
,
Matalkah
F
, et al
Combination of eribulin and Aurora A inhibitor MLN8237 prevents metastatic colonization and induces cytotoxic autophagy in breast cancer
.
Mol Cancer Ther
2016
;
15
:
1809
22
.
59.
Jensen
JS
,
Omarsdottir
S
,
Thorsteinsdottir
JB
,
Ogmundsdottir
HM
,
Olafsdottir
ES
. 
Synergistic cytotoxic effect of the microtubule inhibitor marchantin A from Marchantia polymorpha and the Aurora kinase inhibitor MLN8237 on breast cancer cells in vitro
.
Planta Med
2012
;
78
:
448
54
.
60.
Kelly
KR
,
Ecsedy
J
,
Medina
E
,
Mahalingam
D
,
Padmanabhan
S
,
Nawrocki
ST
, et al
The novel Aurora A kinase inhibitor MLN8237 is active in resistant chronic myeloid leukaemia and significantly increases the efficacy of nilotinib
.
J Cell Mol Med
2011
;
15
:
2057
70
.
61.
Fiskus
W
,
Hembruff
SL
,
Rao
R
,
Sharma
P
,
Balusu
R
,
Venkannagari
S
, et al
Co-treatment with vorinostat synergistically enhances activity of Aurora kinase inhibitor against human breast cancer cells
.
Breast Cancer Res Treat
2012
;
135
:
433
44
.

Supplementary data