Glioblastoma stem cell (GSC) is the major cause of glioblastoma multiforme (GBM) chemotherapy failure. Hypoxia is one of the determinants of GSC. NF-κB plays a pivotal link between hypoxia and cancer stem cells (CSCs). Disulfiram, an antialcoholism drug, has very strong NF-κB–inhibiting and anti-CSC activity. In this study, the in vitro anti-GSC activity of disulfiram and in vivo anti-GBM efficacy of poly lactic–co-glycolic acid nanoparticle-encapsulated disulfiram (DS-PLGA) were examined. We attempt to elucidate the molecular network between hypoxia and GSCs and also examined the anti-GSC activity of disulfiram in vitro and in vivo. The influence of GSCs and hypoxia on GBM chemoresistance and invasiveness was studied in hypoxic and spheroid cultures. The molecular regulatory roles of NF-κB, hypoxia-inducible factor-1α (HIF1α), and HIF2α were investigated using stably transfected U373MG cell lines. The hypoxia in neurospheres determines the cancer stem cell characteristics of the sphere-cultured GBM cell lines (U87MG, U251MG, U373MG). NF-κB is located at a higher hierarchical position than HIF1α/HIF2α in hypoxic regulatory network and plays a key role in hypoxia-induced GSC characters. DS inhibits NF-κB activity and targets hypoxia-induced GSCs. It showed selective toxicity to GBM cells, eradicates GSCs, and blocks migration and invasion at very low concentrations. DS-PLGA efficaciously inhibits orthotopic and subcutaneous U87MG xenograft in mouse models with no toxicity to vital organs.

This article is featured in Highlights of This Issue, p. 1259

Glioblastoma multiforme (GBM) is one of the most lethal and aggressive forms of the malignant adult brain tumor. Despite multimodal treatment including surgical resection with concurrent and adjuvant radiotherapy and chemotherapy, the prognosis of GBM is poor with the median survival time of only 14.6 months and less than 5% of patients with GBM surviving for more than 2 years (1).

GBM manifests high intratumoural heterogeneity with a very small population of cells expressing cancer stem cell (CSC) markers, for example, aldehyde dehydrogenase (ALDH), CD44, CD133, Sox2, Oct4, and Nanog, with epithelial-to-mesenchymal transition (EMT) trend (2, 3). This population of cells has been clarified as glioblastoma stem cells (GSCs), which are quiescent, chemoresistant, and metastatic (4, 5). Studies indicate that GSCs, lodged in hypoxic niches, are in a microenvironment-inducible transient and reversible state rather than a permanent entity (6). As a heterogeneous tumor with an extensively hypoxic environment, GBM provides hypoxic niches for fostering and maintaining GCSs. Hypoxia-inducible factors (HIFs) are the master transcriptional regulators induced by hypoxia (7). NF-κB is a bridge linking inflammation, hypoxia, and cancer (8, 9). NF-κB is an important chemoresistance-related transcription factor upregulating multiple genes related to inflammation, hypoxic response, EMT, and antiapoptotic signaling (10). Many human cancer cells and drug-resistant cancer cell lines possess high constitutive NF-κB activity, which can be further induced by hypoxia (9). Ectopic overexpression of NF-κB has been shown to result in inducing genes related to cancer cell stemness, EMT, and antiapoptosis (11–14). The relationship between HIFs and NF-κB in coordinating the hypoxia-induced GSC characters remains obscure. Understanding the molecular mechanisms behind hypoxia-induced EMT and GSCs may facilitate development of new drugs for GBM therapeutics.

The demand of new drugs for GBM treatment is of clinical urgency, but new drug development is a highly time- and cash-consuming procedure. There is a global trend toward the repositioning of known drugs for new indications (15). Disulfiram, a long-established alcohol-reversal drug, possesses excellent anticancer activity against a wide range of cancers including GBM with low toxicity to normal cells (13, 16–19). Disulfiram also presents a synergistic activity and potentiates the cytotoxicity of conventional chemotherapy drugs and protects normal cells in kidney, gut, and bone marrow in vivo while increasing the therapeutic index (13, 19, 20).

Disulfiram specifically inhibits the activity of ALDH, a functional marker of CSCs with reactive oxygen species (ROS) scavenging activity (21). It effectively eliminates CSCs and reverses chemoresistance (20–22). The cytotoxicity of disulfiram depends on copper (II; Cu) and other divalent transition metal elements (23, 24). Disulfiram-metal complexes block the nuclear translocation of NF-κB by inhibiting proteasome activation and IkBα degradation (17). The functional component of disulfiram is diethyldithiocarbamate (DDC), which strongly chelates Cu to generate ROS and form the end product DDC-Cu. Both ROS and DDC-Cu induce cancer cell apoptosis (23, 24). Therefore, the intact sulfhydryl group is essential for the reaction between disulfiram and Cu. Only oral disulfiram is currently available in clinic. Oral disulfiram is quickly reduced to DDC in the gastrointestinal system. The sulfhydryl groups in DDC are instantly methylated and glucuronidated when disulfiram is enriched in the liver. The methylated and glucuronidated DDC loses its Cu chelating function and cytotoxicity (23, 24). The half-life of disulfiram is less than 1 minute in serum (16). This explains why anticancer activity of disulfiram has been known for more than 30 years without successful and reproducible cancer therapeutic outcomes. We have shown that using a nano-delivery system to protect the sulfhydryl group in disulfiram can improve the anticancer efficacy of disulfiram in breast, liver, and lung cancer models (16, 25).

Here, we demonstrate that NF-κB plays pivotal roles in hypoxia-induced GSC traits. Disulfiram inhibits the NF-κB pathway, targets GSCs, and reverses chemoresistance in vitro. A strong in vivo anti-GBM efficacy was achieved by intravenous administration of a long-half-life circulating poly lactic–co-glycolic acid (PLGA) encapsulated disulfiram (DS-PLGA) in both intracranial and subcutaneous mouse GBM models. Further study may translate DS-PLGA into GBM treatment.

Cell lines and reagents

The following human GBM cell lines U87MG ATCC (RRID:CVCL_0022), U251MG (RRID:CVCL_0021), and U373MG ATCC (RRID:CVCL_2219) were purchased from ECACC. The U373MG ATCC cell line is a derivative of the U251MG cell line. All human cell lines have been authenticated using short tandem repeat (STR) profiling within the last 3 years, and all experiments were performed with Mycoplasma-free cells. Disulfiram, temozolomide, copper chloride (CuCl2), copper gluconate (CuGlu), crystal violet, dichloromethane, poly-2-hydroxyethyl methacrylate (poly-HEMA), and BSA were from Sigma-Aldrich. DMEM medium and FCS were supplied by Lonza. Antibodies were purchased from Cell Signaling Technology (Ki67 and BAX), Abcam (NF-κBp65, IκBa, Bax, Bcl2, MMP-2, BMP4, BMP9, Sox2, Oct4, Nanog, E-cadherin, N-cadherin, Vimentin, and ALDH1), BD Biosciences, or Miltenyi Biotec (CD44-FITC and CD133-FITC). The Hypoxyprobe-1 Plus kit was from Hypoxyprobe. ALDEFLUOR was from Stemcell Technologies. Matrigel and cell culture inserts were purchased from Fisher. The DS-PLGA was developed in our laboratory (16).

Normoxic and hypoxic cell culture

The GBM cell lines were cultured under normoxia (20% oxygen) or hypoxia (1% oxygen) and maintained in DMEM supplemented with 10% FCS, 2 mmol/L L-Glutamine, 50 units/mL penicillin, and 50 μg/mL streptomycin. Cells were cultured under normoxia or hypoxia in parallel for 5 days before further experiments. The hypoxic culture was performed in a Panasonic Hypoxia Incubator (Panasonic Biomedical).

In vitro spheroid culture

Cells were cultured in ultralow adherence poly-HEMA–coated T25 flasks at a cell density of 20,000 cells/mL. Neurosphere (NS) was grown in neural stem cell medium [serum-free DMEM-F12 supplemented with B27 and N2 serum replacement (Invitrogen), 0.3% glucose (Sigma-Aldrich), 10 ng/mL epidermal growth factor (Sigma-Aldrich), 10 ng/mL basic fibroblasts growth factor (R&D Systems), 20 mg/mL insulin (Sigma-Aldrich), 2 mg/mL heparin (Sigma-Aldrich)]. The group of suspension (SUS) culture was performed in normal DMEM supplemented with 10% serum. The cells were incubated at 37°C for 7 days, before further analysis, with necessary media replenishment every 3 days.

Detection of hypoxia in cell culture

The Hypoxyprobe Kit was used following the manufacturer's protocol. For immunocytochemistry assay, the attached cells (ATTs) were cultured at normoxic or hypoxic condition for 72 hours and cocultured with Hypoxyprobe reagent overnight. The NS and SUS cells were cocultured with Hypoxyprobe overnight and cytospined at 800 rpm for 3 minutes to spread the spheres onto Polylysine-coated slides (VWR). The cells were fixed and stained with FITC-conjugated anti-hypoxyprobe MAbs and imaged using a confocal microscope. For quantification of hypoxic cells, the cells were stained with Hypoxyprobe and subjected to flow cytometric analysis. The hypoxic population was detected using a FACS Calibur flow cytometer with a 488-nm blue laser and a standard FITC 530/30-nm bandpass filter.

Flow cytometric analysis of CSC markers

The trypsinized cells (2.5 × 105) were analyzed with corresponding CSC markers. ALDH activity was measured by staining with ALDH substrate. The CD133 and CD44 cell surface marker were detected by FITC-conjugated anti-CD133 and anti-CD44 antibodies. The cells were examined using BD FACS Calibur and Cell Quest Pro software.

In vitro cytotoxicity assay

The methodology of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay for the normoxia cultured cells has been described previously (22). To determine the effect of hypoxia on drug sensitivity, the cells were cultured in 1% oxygen at a cell density of 5 × 103 cells/well in 96-well plate for 4 days and exposed to anticancer drugs for another 72 hours before MTT assay with a parallel MTT assay in normoxia. The NS and SUS spheres were trypsinised and recultured in poly-HEMA–coated 96-well flat bottom plates in their corresponding culture medium at a cell density of 5 × 103 cells/well overnight. After exposure to drugs for 72 hours, the cells were subjected to a standard MTT assay. The NS and SUS cells were centrifuged at 800 rpm for 5 minutes before MTT assay.

Western blot analysis of proteins

The whole and nuclear protein was extracted from normoxia, hypoxia, NS, and SUS cultured GBM cells with RIPA buffer, quantified and separated by SDS PAGE. The proteins were blotted onto polyvinylidene difluoride (PVDF) membrane (Thermo Fisher Scientific), blocked with 5% nonfat milk, and incubated with appropriate primary and secondary antibodies of target proteins.

Stable transfection of U373MG cell line with NF-κBp65, HIF1α, and HIF2α

U373MG cells (2.5 × 105/well) were cultured in 35-mm dishes until 70% confluent, and Lipofectamine 2000 Superfect (Qiagen) was used to transfect pcDNA3.1(+; Invitrogen), pcDNA3.1/Hygro/NF-κBp65, pCMV6-Neo/HIF1α, pCMV6-Neo/HIF2α (Origene). The successfully transfected clones were selected in relevant antibiotics.

In vitro migration and invasion assay

The migration and invasion assays were performed using cell culture inserts (Thermo Fisher Scientific) coated with or without Matrigel (BD Biosciences). Cells (5 × 104) were resuspended in 200 μL of serum-free DMEM and placed in the upper chamber of the insert. The lower chamber was filled with medium containing 10% FCS. After 16 hours incubation at 37°C, migrated cells were fixed by methanol and stained in 0.5% crystal violet. Cells were imaged and counted under a microscope.

qRT-PCR

qRT-PCR was performed using Taqman assays. RNA was extracted using an RNA purification kit (Norgen) following the manufacturer's instructions and reverse transcribed into cDNA (Multiscribe Reverse Transcriptase, Applied Biosystems). A QuantStudio 6 Flex Real-Time PCR system (Applied Biosystems) was used for qPCR experiments. Gene expression levels were normalized to control genes (GAPDH and HPRT1) and calculated according to the ΔΔCT method.

Luciferase reporter gene assay

The cells (1 × 104/well) were cultured in 96-well plates overnight. The NF-κB (pNF-κBTal-Luc, BD Biosciences) and hypoxia response element (HRE) (pGL4.42-luc2P/HRE, Promega) luciferase reporter vectors and pGL3-Basic (Promega) were co-transfected with pSV40-Renilla (Promega) DNA. Forty-eight hours after transfection, the luciferase activity was determined using Dual Luciferase Assay reagents (Promega) according to the manufacturer's instructions. The relative luciferase activity was calculated (25).

IHC and hematoxylin and eosin staining

The sections from paraffin-embedded tumor and normal tissues were stained with primary antibodies [Ki67, Bax, NF-κBp65, and ALDH1 (1:200)] then biotinylated secondary antibody and followed by incubation in ABC reagent (DAKO Labs). For hematoxylin and eosin (H&E) staining, the paraffin-embedded sample slides were stained with H&E, and the slides were mounted with coverslips using Permount (Thermo Fisher Scientific).

Terminal deoxyribonucleotidyl transferase–mediated dUTP nick- end labeling assay

Tumor tissues were paraffin-embedded and stained according to the manufacturer's instructions (Roche). Briefly, the slides were incubated with TdT Enzyme, Stop/Wash Buffer, antidigoxigenenin, and then stained with peroxidase substrate and incubated in ABC reagent. Finally, the slide was mounted with 3,3′-diaminobenzidine and visualized under a light microscope.

Pharmacokinetic study and in vivo anti-GBM xenograft experiments

The animal experiments were reviewed and approved by the Ethical Committee of Third Military Medical University (Chongqing, China) and University of Wolverhampton (Wolverhampton, UK). Rats (five per group) were injected with disulfiram or DS-PLGA (250 mg/kg) through a tail vein. Blood samples were collected at 0.25, 0.5, 1, 2, 4, and 8 minutes for the disulfiram group and at 5, 10, 20, 40, 60, 120, and 180 minutes for the DS-PLGA group. The plasma (500 μL) was separated and mixed with methanol (250 μL). After vigorously shaking and centrifuging (10,000 rpm, 15 minutes), the disulfiram concentration in the supernatant was determined by high-performance liquid chromatography (HPLC) as described elsewhere (16). The intracranial GBM model was developed as previously described (26). Briefly, the 5-week-old female BALB/c Nu/Nu mice were anesthetized. The U87MG-luciferase-GFP cells (2 × 105/5 μL PBS) were injected into the brain using a 25-μL microsyringe. After 10 days, the animals were treated with empty PLGA or DS-PLGA (10 mg/kg i.v.) plus CuGlu (6 mg/kg orally) three times per week for 4 weeks. For the subcutaneous GBM model, U87MG cells (5 × 106) were injected into the front flank of BABL/c Nu/Nu mice. When the tumor volume reached approximately 200 mm3, the tumor-bearing mice were subjected to the above treatment. The xenograft size was recorded twice per week. The tumor volume was calculated by the following formula: V = (L × W2) × 0.5, where L is the length and W is the width of the tumor. After 4 weeks, the animals were sacrificed. The tumors were removed, photographed, and subjected to further analysis.

Statistical analysis

Student t test and one-way ANOVA were used in this study. P < 0.05 and <0.01 were considered statistically significant and highly significant, respectively.

Data availability statement

The data generated in this study are available within the article and its supplementary data files.

Hypoxia, stemness, NF-κB activation, and temozolomide resistance were detected in both NS and SUS cultured GBM cells

Characteristic spheres were formed in both serum-free NS and serum-rich SUS culturing conditions (Fig. 1A). All the sphere cells expressed high stem cell (Fig. 1B) and embryonic stem cell markers (Fig. 1C). Overexpression of N-cadherin, Vimentin, and downregulation of E-cadherin were detected in NS and SUS cells indicating the presence of EMT in the sphere cells (Fig. 1D). Significant temozolomide resistance was detected when the sphere cells were exposed to temozolomide for 72 hours and subjected to sphere reformation and MTT cytotoxicity assay (Fig. 1A; Supplementary Tables S1 and S2).

Figure 1.

Hypoxia, EMT/stemness, temozolomide resistance and overexpression of NF-κBp65 were detected in spheroid cells. A, Morphology of temozolomide-treated ATT, NS, and SUS-cultured GBM cells. B, Flow cytometric analysis of the expression of CD133, CD44, and ALDH activity (mean ± SD; n = 3). C and D, Western blot analysis of embryonic stem- and EMT-related proteins. E, Hypoxic cells were detected by Hypoxyprobe and FITC-conjugated anti-Hypoxyprobe MAb staining (green; cytoplasm). The nuclei were counterstained by propidium iodide (PI; red). F, Flow cytometric analysis of Hypoxyprobe-stained cells (mean ± SD; n = 3). Western blot of HIF1α and HIF2α (G; nuclear protein) and NF-κBp65 (H) protein expression. β-actin and Nucleolin were used as housekeeping control. **, P < 0.01. ve, negative; Nor, normoxia.

Figure 1.

Hypoxia, EMT/stemness, temozolomide resistance and overexpression of NF-κBp65 were detected in spheroid cells. A, Morphology of temozolomide-treated ATT, NS, and SUS-cultured GBM cells. B, Flow cytometric analysis of the expression of CD133, CD44, and ALDH activity (mean ± SD; n = 3). C and D, Western blot analysis of embryonic stem- and EMT-related proteins. E, Hypoxic cells were detected by Hypoxyprobe and FITC-conjugated anti-Hypoxyprobe MAb staining (green; cytoplasm). The nuclei were counterstained by propidium iodide (PI; red). F, Flow cytometric analysis of Hypoxyprobe-stained cells (mean ± SD; n = 3). Western blot of HIF1α and HIF2α (G; nuclear protein) and NF-κBp65 (H) protein expression. β-actin and Nucleolin were used as housekeeping control. **, P < 0.01. ve, negative; Nor, normoxia.

Close modal

Hypoxyprobe staining showed hypoxic population in the core region of both NS and SUS (Fig. 1E). A high proportion of hypoxic cells were detected in NS and SUS cells using flow cytometry (Fig. 1F). Further evidence of the hypoxic condition in NS and SUS cells was shown by the overexpression of HIF1α and HIF2α proteins (Fig. 1G). Overexpression of NF-κBp65 was detected in the sphere cells (Fig. 1H).

Hypoxia induces stemness, NF-κB activation, EMT, and temozolomide resistance in GBM cells

To confirm the link between hypoxia and GSC phenotypes, we cultured monolayer GBM cells under hypoxia. The cellular hypoxia was confirmed by Hypoxyprobe staining (Fig. 2A and B) and overexpression of HIF1α and HIF2α proteins (Fig. 2C). The hypoxia culture induces high levels of GSC and embryonic stem cell markers (Fig. 2D and E). The hypoxic GBM cells are significantly resistant to temozolomide (Fig. 2F). The overexpression of Bcl2 and downregulation of Bax indicate that hypoxia activates antiapoptotic pathways (Fig. 2G). Hypoxia-induced EMT was confirmed by the overexpression of CD44, Vimentin, N-cadherin, downregulation of E-cadherin (Fig. 2D and H), and overexpression of mRNA from EMT- and metastasis-related genes (Supplementary Fig. S1). Increased migration and invasion abilities were manifested in the hypoxia-cultured GBM cells (Fig. 2I and J; Supplementary Table S3). Together, the above findings indicate that hypoxia induces stemness, EMT, and chemoresistance in GBM cells. Similar to spheroid cultures, the high expression of NF-κBp65 protein and its nuclear translocation were also detected in the hypoxia-cultured cells (Fig. 2K).

Figure 2.

EMT/stemness, temozolomide resistance, invasiveness, and NF-κB status detected in hypoxia-cultured GBM cells. A, Images of Hypoxyprobe-stained cells. Hypoxic cells were detected by Hypoxyprobe and FITC-conjugated anti-Hypoxyprobe MAb staining (green; cytoplasm). The nuclei were counterstained by PI (red). B, Flow cytometric analysis of Hypoxyprobe-stained cells (mean ± SD; n = 3). C, Western blot of HIF1α and HIF2α (nuclear protein). D, Flow cytometric analysis of CD133, CD44, and ALDH status in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). E, Western blot of embryonic proteins in normoxia- and hypoxia-cultured cells. F, IC50 of temozolomide in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). G, Western blot of Bcl2 and Bax. H, Western blot of EMT-related factors. Typical migration (I) and invasion (J) images of normoxia- and hypoxia-cultured cells. K, Western blot of NF-κBp65. **, P < 0.01. Nor, normoxia; Hyp, hypoxia; TMZ, temozolomide.

Figure 2.

EMT/stemness, temozolomide resistance, invasiveness, and NF-κB status detected in hypoxia-cultured GBM cells. A, Images of Hypoxyprobe-stained cells. Hypoxic cells were detected by Hypoxyprobe and FITC-conjugated anti-Hypoxyprobe MAb staining (green; cytoplasm). The nuclei were counterstained by PI (red). B, Flow cytometric analysis of Hypoxyprobe-stained cells (mean ± SD; n = 3). C, Western blot of HIF1α and HIF2α (nuclear protein). D, Flow cytometric analysis of CD133, CD44, and ALDH status in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). E, Western blot of embryonic proteins in normoxia- and hypoxia-cultured cells. F, IC50 of temozolomide in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). G, Western blot of Bcl2 and Bax. H, Western blot of EMT-related factors. Typical migration (I) and invasion (J) images of normoxia- and hypoxia-cultured cells. K, Western blot of NF-κBp65. **, P < 0.01. Nor, normoxia; Hyp, hypoxia; TMZ, temozolomide.

Close modal

The regulatory relationship of NF-κB, HIF1α, and HIF2α in GBM cells

HIFs and NF-κB are hypoxia-induced genes responsible for EMT and CSC traits (14, 27). The regulatory relationship between HIFs and NF-κB is still not clear. To establish which one of these three factors is on the central regulatory position in hypoxia response, we stably transfected U373MG cells with NF-κBp65, HIF1α, and HIF2α. The successful transfected clones were confirmed by Western blot and luciferase reporter gene assay (Fig. 3AF). The NF-κBp65–transfected clones express high levels of HIF1α and HIF2α proteins (Fig. 3G) and mRNAs (Supplementary Fig. S2) and possess high hypoxia response element (HRE) transcriptional activity (Fig. 3H). In contrast, HIF1α and HIF2α transfected clones did not show significant NF-κB upregulation at mRNA (Supplementary Fig. S2), protein (Fig. 3I and K), and transcription (Fig. 3J and L) levels. The expression of HIF2α mRNA was upregulated by HIF1α transfection but not vice versa (Supplementary Fig. S2). Therefore, NF-κB may be located at a higher hierarchical position than HIF1α and HIF2α in hypoxia response gene network.

Figure 3.

The regulatory relationship among NF-κB, HIF1α, and HIF2α were analyzed in stably transfected GBM clones. NF-κBp65 expression (A) and κB transcriptional activity (B) respectively in mock and NF-κBp65 transfected clones. HIF1α expression (C) and HRE transcriptional activity (D) respectively in mock and HIF1α transfected clones. HIF2α expression (E) and HRE activity (F) respectively in mock and HIF2α transfected clones. HIF1α, HIF2α protein expression (G), and HRE activity (H) in mock and NF-κBp65 transfected clones. NF-κBp65 protein expression (I) and κB activity (J) in mock and HIF1α transfected clones. NF-κBp65 protein expression (K) and κB activity (M) in mock and HIF2α transfected clones. Clone C4 (red framed) was not used in this study. Reporter gene assay: mean ± SD; n = 3. **, P < 0.01. Nu, nuclear protein expression.

Figure 3.

The regulatory relationship among NF-κB, HIF1α, and HIF2α were analyzed in stably transfected GBM clones. NF-κBp65 expression (A) and κB transcriptional activity (B) respectively in mock and NF-κBp65 transfected clones. HIF1α expression (C) and HRE transcriptional activity (D) respectively in mock and HIF1α transfected clones. HIF2α expression (E) and HRE activity (F) respectively in mock and HIF2α transfected clones. HIF1α, HIF2α protein expression (G), and HRE activity (H) in mock and NF-κBp65 transfected clones. NF-κBp65 protein expression (I) and κB activity (J) in mock and HIF1α transfected clones. NF-κBp65 protein expression (K) and κB activity (M) in mock and HIF2α transfected clones. Clone C4 (red framed) was not used in this study. Reporter gene assay: mean ± SD; n = 3. **, P < 0.01. Nu, nuclear protein expression.

Close modal

The effect of NF-κB and HIFs on stemness, EMT, temozolomide resistance and invasiveness in GBM cells

Next, we examined the effect of NF-κB and HIFs on EMT and stemness in GBM cells. The NFκBp65, HIF1α, and HIF2α transfected clones expressed higher levels of GSC (Fig. 4A) and embryonic stem cell (Fig. 4B) markers. The mRNA and protein of EMT-related genes were detected in the transfected clones (Fig. 4C and D). The NF-κBp65 and HIF2α but not HIF1α transfected clones are significantly resistant to temozolomide (Fig. 4E). We also examined the migration and invasion abilities, one of the most common features of EMT (28), in the transfected cell lines. All of the NF-κBp65, HIF1α, and HIF2α transfected cell lines possess significantly higher migration and invasion activity (Fig. 4FH; Supplementary Table S4). Some metastasis-related genes are overexpressed in these cell lines (Fig. 4D). These data indicate that all of these three genes promote the invasiveness of GBM, but only NF-κB and HIF2α are responsible for temozolomide resistance.

Figure 4.

Analysis of EMT/stemness, cytotoxicity of temozolomide, and migration/invasion status in NF-κBp65, HIF1α, and HIF2α transfected clones. A, Flow cytometric analysis of CD44, CD133 expression, and ALDH activity (mean ± SD; n = 3). Western blot analysis of embryonic stem cell (B) and EMT-related (C) proteins. D, Real-time RT-PCR analysis of EMT and invasion-related genes. E, Cytotoxicity of temozolomide (mean ± SD; n = 3). Representative migration/invasion images of HIF1α (F), HIF2α (G), and NF-κBp65 (H) transfected clones. *, P < 0.05; **, P < 0.01.

Figure 4.

Analysis of EMT/stemness, cytotoxicity of temozolomide, and migration/invasion status in NF-κBp65, HIF1α, and HIF2α transfected clones. A, Flow cytometric analysis of CD44, CD133 expression, and ALDH activity (mean ± SD; n = 3). Western blot analysis of embryonic stem cell (B) and EMT-related (C) proteins. D, Real-time RT-PCR analysis of EMT and invasion-related genes. E, Cytotoxicity of temozolomide (mean ± SD; n = 3). Representative migration/invasion images of HIF1α (F), HIF2α (G), and NF-κBp65 (H) transfected clones. *, P < 0.05; **, P < 0.01.

Close modal

Disulfiram inhibits hypoxia-induced GSC and EMT phenotypes in vitro

Hypoxic cells remain sensitive to disulfiram (Fig. 5A), although they are significantly resistant to temozolomide. Disulfiram and Cu block the sphere reformation in NS and SUS cultures. The disulfiram or Cu alone had no effect on sphere reformation indicating the essential requirement of Cu in the function of disulfiram against GSCs (Fig. 5B and C; Supplementary Table S2). The GSC markers induced by spheroid and hypoxia culture were blocked by disulfiram/Cu (Fig. 5D and E; Supplementary Figs. S3 and S4). Disulfiram/Cu inhibited the migration and invasion of GBM cell lines at an extremely low concentration (25 nmol/L; Fig. 5F and G; Supplementary Table S3). NF-κB is a key player in GSC and EMT. The expression of NF-κBp65 protein in hypoxia and sphere cultured GBM cells was inhibited by disulfiram/Cu (Fig. 5H). Disulfiram/Cu treatment enhanced and inhibited the expression of Bax and Bcl2, respectively (Fig. 5H).

Figure 5.

Analysis of the effect of disulfiram/Cu on hypoxia-induced chemoresistance, EMT/stemness, invasiveness, and NF-κBp65 expression. A, IC50s of disulfiram/Cu in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). The effect of disulfiram, Cu, and disulfiram/Cu on the sphere-reformation ability of NS (B) and SUS (C) cultured cells. Representative flow cytometric dot plots of the effect of disulfiram/Cu on the ALDH activity (D) and CD133 expression (E). Effect of disulfiram/Cu on the migration (F) and invasion (G) of the hypoxia-cultured GBM cell lines. H, Western blot analysis of the effect of disulfiram/Cu on ATT normoxia, hypoxia, NS, and SUS culture induced NF-κBp65 protein expression. DS, disulfiram; NOR, normoxia; HYP, hypoxia.

Figure 5.

Analysis of the effect of disulfiram/Cu on hypoxia-induced chemoresistance, EMT/stemness, invasiveness, and NF-κBp65 expression. A, IC50s of disulfiram/Cu in normoxia- and hypoxia-cultured cells (mean ± SD; n = 3). The effect of disulfiram, Cu, and disulfiram/Cu on the sphere-reformation ability of NS (B) and SUS (C) cultured cells. Representative flow cytometric dot plots of the effect of disulfiram/Cu on the ALDH activity (D) and CD133 expression (E). Effect of disulfiram/Cu on the migration (F) and invasion (G) of the hypoxia-cultured GBM cell lines. H, Western blot analysis of the effect of disulfiram/Cu on ATT normoxia, hypoxia, NS, and SUS culture induced NF-κBp65 protein expression. DS, disulfiram; NOR, normoxia; HYP, hypoxia.

Close modal

DS-PLGA extends the half-life of disulfiram in vivo and suppresses GBM in orthotopic and subcutaneous GBM mouse models

To overcome the very short half-life of disulfiram in the bloodstream (16, 29), we used a DS-PLGA formulation (16) to deliver disulfiram intravenously in a mouse model. The half-life of DS-PLGA in rat was 13.4 minutes (Supplementary Table S5) while the free disulfiram was immediately undetectable after injection.

The in vivo efficacy of DS-PLGA was assessed in both intracranial and subcutaneous models. After 4 weeks of treatment, DS-PLGA/Cu significantly reduced the sizes of the intracranial and subcutaneous tumors and the tumor weight in subcutaneous injected mice (Fig. 6AC). These results indicate that the PLGA encapsulation delivers the intact disulfiram to GBM tissues. The TUNEL staining indicated apoptosis induced by DS-PLGA/Cu in vivo (Fig. 6D). DS-PLGA/Cu treatment inhibited the expression of NF-κBp65, ALDH, and Ki67 expression in the GBM xenografts (Fig. 6E). No toxic effect on vital organs was observed and mouse body weight was maintained (Fig. 6F and G).

Figure 6.

Anti-GBM efficacy and selectivity of DS-PLGA/Cu in mouse GBM xenograft models. A, The fluorescent and pathological images of anti-GBM efficacy of DS-PLGA/Cu on intracranial GBM model. B, The macrographic images of the effect of DS-PLGA/Cu on subcutaneous xenografts. C, The subcutaneous xenograft weight (mean ± SD; n = 7; **, P < 0.01). D, TUNEL staining subcutaneous xenograft. E, IHC analysis of NF-κBp65, Ki67, and ALDH expression in subcutaneous xenografts. F, The alteration of mouse body weight. G, The histologic images of mouse organs.

Figure 6.

Anti-GBM efficacy and selectivity of DS-PLGA/Cu in mouse GBM xenograft models. A, The fluorescent and pathological images of anti-GBM efficacy of DS-PLGA/Cu on intracranial GBM model. B, The macrographic images of the effect of DS-PLGA/Cu on subcutaneous xenografts. C, The subcutaneous xenograft weight (mean ± SD; n = 7; **, P < 0.01). D, TUNEL staining subcutaneous xenograft. E, IHC analysis of NF-κBp65, Ki67, and ALDH expression in subcutaneous xenografts. F, The alteration of mouse body weight. G, The histologic images of mouse organs.

Close modal

Investigation and unveiling of molecular mechanisms of GSC-induced chemoresistance and invasiveness (4, 5) will likely facilitate the development of novel and efficacious therapies for patients with GBM. Since Brent A. Reynolds and Samuel Weiss successfully isolated normal neural stem cells using sphere-forming culture in growth factor-supplemented serum-free medium (30), this system has been widely adapted for in vitro culture of GSCs. It has been generally believed that serum-free and growth factor-rich medium is essential for maintenance of anchorage-independent proliferation and stemness. Our study manifested that GBM cells form NSs in both serum-rich and serum-free media, which express comparable levels of GSC markers (Fig. 1B and C) and are significantly resistant to temozolomide (Fig. 1A; Supplementary Table S1). Similar findings were reported previously (25, 31, 32). EMT and CSCs are inextricably linked microenvironment-determined transient epigenetic phenomena (33), which were detected in NS and SUS cells (Fig. 1B and D). Hypoxia is a central determinant of the stem cell niche (34), which is relevant to GBM, as these tumors have significantly lower intratumoral oxygen concentration than normal brain tissues (35). In line with our previous findings in breast cancer (25), the core regions of NSs contain a high proportion of hypoxic cells (Fig. 1E and F), which express high levels of HIF1α, HIF2α, and NF-κBp65 (Fig. 1G and H), the key factors induced by inflammation and hypoxia (7, 8). The GBM cells cultured in hypoxia produce epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF); ref. 36. Our results suggest that the GSC traits may be determined by the hypoxic microstructure of the spheroid culture rather than the medium, and the selective medium may not be essential for the in vitro GSC culture. In contrast, the conventional serum-containing medium may better mimic the in vivo pathophysiologic conditions and may be more suitable for in vitro GSC study.

The interconversion of CSCs and non-CSCs is a common phenomenon driven by environmental stimuli (2, 3). As such, the GSC and non-GSC cells are mutable cellular traits determined by oxygen levels in the microenvironment (34). Hypoxia-cultured cells grown as a monolayer expressed high levels of GSC and embryonic stem cell markers and were markedly resistant to temozolomide (Fig. 2DF). A high proportion of both sphere- and hypoxia-cultured cells overexpressed CD44, N-cadherin, Vimentin, and some key EMT markers, whereas E-cadherin was downregulated (Figs. 1B, D, and 2H; Supplementary Fig. S1). These results indicate hypoxia induces EMT in these cells. EMT is responsible for cancer cell migration, invasion, and metastasis (28). The hypoxia-cultured cells showed higher migration and invasion abilities (Fig. 2I and J; Supplementary Table S3).

Overexpression of HIF1α, HIF2α, and NF-κBp65 was detected in both sphere- and hypoxia-cultured GBM cells (Figs. 1G and H, 2C and K). These transcription factors are related to dedifferentiation and maintenance of GSCs (37). The stably transfected U373MG models were used to elucidate the hierarchical relationship among these three genes. NF-κBp65 transfection induced HIF1α and HIF2α protein, mRNA expression and HRE activity, but not vice versa (Fig. 3GM; Supplementary Fig. S2). NF-κB-, HIF1α-, and HIF2α-transfected cells express some key EMT transcription factors (Fig. 4C and D). NF-κB may manipulate the hypoxia-induced activation of HIF1α and HIF2α via binding to the κB sites on their promoter regions (38). HIF1α-transfected clones overexpressed HIF2α but not vice versa (Supplementary Fig. S2). Although all of the HIF1α-, HIF2α-, and NF-κBp65–transfected clones overexpress GSC and EMT markers, only NF-κBp65 and HIF2α-transfected cells are resistant to temozolomide (Fig. 4E). This is consistent with previous reports that HIF1α mainly regulates acute hypoxia-induced cell responses. In contrast, HIF2α is responsible for chronic hypoxia, which induces GSC and EMT traits (39, 40). HIF1α may counterbalance the drug resistant effect of HIF2α (41). All of these three genes induce the expression of key EMT- and metastasis-related genes and promote GBM cell migration and invasion (Fig. 4C, D, F–H; Supplementary Table S4).

In line with our previous studies (16, 22, 24, 25), disulfiram targets NF-κB and reverses hypoxia-induced cancer metastasis/chemoresistance in a Cu-dependent manner (Fig. 5AC, F, and G). Disulfiram/Cu abolished GSC markers and completely blocked NS reformation ability (Fig. 5BE). Intracranial metastasis is the major cause of GBM recurrence (42, 43). Disulfiram blocked GBM cell migration and invasion at an extremely low concentration (25 nmol/L). In combination with radiotherapy, disulfiram may effectively improve the current therapeutic outcomes of GBM.

The very short half-life of disulfiram in the bloodstream is the key limit for its efficacy as cancer therapeutics. The DS-PLGA has a significantly longer half-life (Supplementary Table S5; ref. 16) and can successfully deliver intact disulfiram to cancer tissues. The low molecular weight (296) and high lipophilicity facilitate disulfiram to penetrate GBM tissues. The DS-PLGA/Cu showed very strong in vivo anti-GBM efficacy in both intracranial and subcutaneous mouse models. It inhibited NF-κB and ALDH expression in GBM tissues. No systemic toxic effect of DS-PLGA/Cu on vital organs (kidney, liver, lung, spleen, and brain) and no mouse body weight loss was observed. The intracranial metastasis is the major cause of postoperative relapse of GBM, although local drug delivery using 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU)/wafer marginally improved the survival time, the vulnerability of brain tissue to BCNU hinders its success in GBM treatment (44). Disulfiram showed selective toxicity in cancer cells (25). The migration/invasion inhibiting concentration (25 nmol/L) is significantly lower than its toxic concentration. DS-PLGA may potentially be locally delivered using convection-enhanced delivery (45) or wafer formulation in GBM treatment. Nevertheless, our results suggest that repurposing disulfiram through a novel type of delivery system that prolongs its plasma half-life offers new possible therapeutic options for the treatment of GBM.

V. Kannappan reports personal fees from Disulfican Ltd. during the conduct of the study and personal fees from Disulfican Ltd. outside the submitted work. W. Wang reports grants from Innovate UK, European Regional Developmetal Fund, and Smart Concept Fund during the conduct of the study; nonfinancial support from Disulfican Ltd. outside the submitted work; and has a patent for US10695299B2 issued. No disclosures were reported by the other authors.

V. Kannappan: Data curation, formal analysis, investigation, methodology, writing–original draft. Y. Liu: Data curation, formal analysis, validation, investigation, methodology. Z. Wang: Data curation, formal analysis, validation, investigation, methodology. K. Azar: Data curation, formal analysis, validation, investigation, methodology. S. Kurusamy: Data curation, formal analysis, validation, investigation. R.S. Kilari: Investigation, methodology. A.L. Armesilla: Supervision. M.R. Morris: Supervision, writing–review and editing. M. Najlah: Investigation, methodology. P. Liu: Investigation, methodology. X.-W. Bian: Conceptualization, resources, supervision, investigation, project administration. W. Wang: Conceptualization, resources, supervision, funding acquisition, writing–original draft, project administration.

We would like to express our great thanks to Prof. Andrew Pollard (Chairman/Director, Disulfican Ltd.) for his significant contribution to Innovate UK- Jiangsu grant application and project management. We are also immensely grateful to Prof. Patrick Ball and Dr. Hana Morrissey for careful reading and very constructive comments on an earlier version of this manuscript.

This study was supported by Research Institute in Healthcare Science/University of Wolverhampton PhD studentship and Innovate UK Jiangsu- UK Industrial Challenge Programme (104022). Some key equipment was jointly funded by European Regional Developmental Fund, Smart Concept Fund, University of Wolverhampton, and Disulfican Ltd.

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.
Darefsky
AS
,
King
JT
Jr
,
Dubrow
R
.
Adult glioblastoma multiforme survival in the temozolomide era: a population-based analysis of Surveillance, Epidemiology, and End Results registries
.
Cancer
2012
;
118
:
2163
72
.
2.
Mani
SA
,
Guo
W
,
Liao
MJ
,
Eaton
EN
,
Ayyanan
A
,
Zhou
AY
, et al
.
The epithelial-mesenchymal transition generates cells with properties of stem cells
.
Cell
2008
;
133
:
704
15
.
3.
Batlle
E
,
Clevers
H
.
Cancer stem cells revisited
.
Nat Med
2017
;
23
:
1124
34
.
4.
Singh
SK
,
Hawkins
C
,
Clarke
ID
,
Squire
JA
,
Bayani
J
,
Hide
T
, et al
.
Identification of human brain tumour initiating cells
.
Nature
2004
;
432
:
396
401
.
5.
Cheray
M
,
Begaud
G
,
Deluche
E
,
Nivet
A
,
Battu
S
,
Lalloue
F
, et al
.
Cancer stem-like cells in glioblastoma
. In:
De Vleeschouwer
S
,
editor
.
Brisbane (Australia)
:
Glioblastoma
;
2017
.
6.
Zipori
D
.
The nature of stem cells: state rather than entity
.
Nat Rev Genet
2004
;
5
:
873
8
.
7.
Keith
B
,
Johnson
RS
,
Simon
MC
.
HIF1alpha and HIF2alpha: sibling rivalry in hypoxic tumour growth and progression
.
Nat Rev Cancer
2011
;
12
:
9
22
.
8.
Clevers
H
.
At the crossroads of inflammation and cancer
.
Cell
2004
;
118
:
671
4
.
9.
D'Ignazio
L
,
Rocha
S
.
Hypoxia induced NF-kappaB
.
Cells
2016
;
5
:
10
.
10.
Soubannier
V
,
Stifani
S
.
NF-kappaB signalling in glioblastoma
.
Biomedicines
2017
;
5
:
29
.
11.
Wang
W
,
Cassidy
J
.
Constitutive nuclear factor-kappa B mRNA, protein overexpression and enhanced DNA-binding activity in thymidylate synthase inhibitor-resistant tumour cells
.
Br J Cancer
2003
;
88
:
624
9
.
12.
Wang
W
,
Cassidy
J
,
O'Brien
V
,
Ryan
KM
,
Collie-Duguid
E
.
Mechanistic and predictive profiling of 5-fluorouracil resistance in human cancer cells
.
Cancer Res
2004
;
64
:
8167
76
.
13.
Guo
X
,
Xu
B
,
Pandey
S
,
Goessl
E
,
Brown
J
,
Armesilla
AL
, et al
.
Disulfiram/copper complex inhibiting NFkappaB activity and potentiating cytotoxic effect of gemcitabine on colon and breast cancer cell lines
.
Cancer Lett
2010
;
291
:
104
13
.
14.
Prasad
S
,
Ravindran
J
,
Aggarwal
BB
.
NF-kappaB and cancer: how intimate is this relationship
.
Mol Cell Biochem
2010
;
336
:
25
37
.
15.
Pushpakom
S
,
Iorio
F
,
Eyers
PA
,
Escott
KJ
,
Hopper
S
,
Wells
A
, et al
.
Drug repurposing: progress, challenges and recommendations
.
Nat Rev Drug Discov
2019
;
18
:
41
58
.
16.
Wang
Z
,
Tan
J
,
McConville
C
,
Kannappan
V
,
Tawari
PE
,
Brown
J
, et al
.
Poly lactic-co-glycolic acid controlled delivery of disulfiram to target liver cancer stem-like cells
.
Nanomedicine
2017
;
13
:
641
57
.
17.
Chen
D
,
Cui
QC
,
Yang
H
,
Dou
QP
.
Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity
.
Cancer Res
2006
;
66
:
10425
33
.
18.
Brar
SS
,
Grigg
C
,
Wilson
KS
,
Holder
WD
Jr
,
Dreau
D
,
Austin
C
, et al
.
Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease
.
Mol Cancer Ther
2004
;
3
:
1049
60
.
19.
Lun
X
,
Wells
JC
,
Grinshtein
N
,
King
JC
,
Hao
X
,
Dang
NH
, et al
.
Disulfiram when combined with copper enhances the therapeutic effects of temozolomide for the treatment of glioblastoma
.
Clin Cancer Res
2016
;
22
:
3860
75
.
20.
Liu
P
,
Kumar
IS
,
Brown
S
,
Kannappan
V
,
Tawari
PE
,
Tang
JZ
, et al
.
Disulfiram targets cancer stem-like cells and reverses resistance and cross-resistance in acquired paclitaxel-resistant triple-negative breast cancer cells
.
Br J Cancer
2013
;
109
:
1876
85
.
21.
Triscott
J
,
Pambid
MR
,
Dunn
SE
.
Bullseye: targeting cancer stem cells to improve the treatment of gliomas by repurposing Disulfiram
.
Stem Cells
2015
;
33
:
1042
6
.
22.
Liu
P
,
Brown
S
,
Goktug
T
,
Channathodiyil
P
,
Kannappan
V
,
Hugnot
JP
, et al
.
Cytotoxic effect of disulfiram/copper on human glioblastoma cell lines and ALDH-positive cancer-stem-like cells
.
Br J Cancer
2012
;
107
:
1488
97
.
23.
Tawari
PE
,
Wang
Z
,
Najlah
M
,
Tsang
CW
,
Kannappan
V
,
Liu
P
, et al
.
The cytotoxic mechanisms of disulfiram and copper(ii) in cancer cells
.
Toxicol Res
2015
;
4
:
1439
42
.
24.
Butcher
K
,
Kannappan
V
,
Kilari
RS
,
Morris
MR
,
McConville
C
,
Armesilla
AL
, et al
.
Investigation of the key chemical structures involved in the anticancer activity of disulfiram in A549 non-small cell lung cancer cell line
.
BMC Cancer
2018
;
18
:
753
.
25.
Liu
P
,
Wang
Z
,
Brown
S
,
Kannappan
V
,
Tawari
PE
,
Jiang
J
, et al
.
Liposome encapsulated Disulfiram inhibits NFκB pathway and targets breast cancer stem cells in vitro and in vivo
.
Oncotarget
2014
;
5
:
7471
85
.
26.
Ping
YF
,
Yao
XH
,
Chen
JH
,
Liu
H
,
Chen
DL
,
Zhou
XD
, et al
.
The anti-cancer compound Nordy inhibits CXCR4-mediated production of IL-8 and VEGF by malignant human glioma cells
.
J Neurooncol
2007
;
84
:
21
9
.
27.
Mannello
F
,
Medda
V
,
Tonti
GA
.
Hypoxia and neural stem cells: from invertebrates to brain cancer stem cells
.
Int J Dev Biol
2011
;
55
:
569
81
.
28.
Chaffer
CL
,
San Juan
BP
,
Lim
E
,
Weinberg
RA
.
EMT, cell plasticity and metastasis
.
Cancer Metastasis Rev
2016
;
35
:
645
54
.
29.
Johansson
B
.
A review of the pharmacokinetics and pharmacodynamics of disulfiram and its metabolites
.
Acta Psychiatr Scand Suppl
1992
;
369
:
15
26
.
30.
Reynolds
BA
,
Weiss
S
.
Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system
.
Science
1992
;
255
:
1707
10
.
31.
Hong
X
,
Chedid
K
,
Kalkanis
SN
.
Glioblastoma cell line-derived spheres in serumcontaining medium versus serum-free medium: a comparison of cancer stem cell properties
.
Int J Oncol
2012
;
41
:
1693
700
.
32.
Min
SO
,
Lee
SW
,
Bak
SY
,
Kim
KS
.
Ideal sphere-forming culture conditions to maintain pluripotency in a hepatocellular carcinoma cell lines
.
Cancer Cell Int
2015
;
15
:
95
.
33.
Shibue
T
,
Weinberg
RA
.
EMT, CSCs, and drug resistance: the mechanistic link and clinical implications
.
Nat Rev Clin Oncol
2017
;
14
:
611
29
.
34.
Colwell
N
,
Larion
M
,
Giles
AJ
,
Seldomridge
AN
,
Sizdahkhani
S
,
Gilbert
MR
, et al
.
Hypoxia in the glioblastoma microenvironment: shaping the phenotype of cancer stem-like cells
.
Neuro Oncol
2017
;
19
:
887
96
.
35.
Beppu
T
,
Kamada
K
,
Yoshida
Y
,
Arai
H
,
Ogasawara
K
,
Ogawa
A
.
Change of oxygen pressure in glioblastoma tissue under various conditions
.
J Neurooncol
2002
;
58
:
47
52
.
36.
Li
G
,
Chen
Z
,
Hu
YD
,
Wei
H
,
Li
D
,
Ji
H
, et al
.
Autocrine factors sustain glioblastoma stem cell self-renewal
.
Oncol Rep
2009
;
21
:
419
24
.
37.
Schwitalla
S
,
Fingerle
AA
,
Cammareri
P
,
Nebelsiek
T
,
Goktuna
SI
,
Ziegler
PK
, et al
.
Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties
.
Cell
2013
;
152
:
25
38
.
38.
Bhat
KPL
,
Balasubramaniyan
V
,
Vaillant
B
,
Ezhilarasan
R
,
Hummelink
K
,
Hollingsworth
F
, et al
.
Mesenchymal differentiation mediated by NF-kappaB promotes radiation resistance in glioblastoma
.
Cancer Cell
2013
;
24
:
331
46
.
39.
Holmquist-Mengelbier
L
,
Fredlund
E
,
Lofstedt
T
,
Noguera
R
,
Navarro
S
,
Nilsson
H
, et al
.
Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype
.
Cancer Cell
2006
;
10
:
413
23
.
40.
Bar
EE
.
Glioblastoma, cancer stem cells and hypoxia
.
Brain Pathol
2011
;
21
:
119
29
.
41.
Bordji
K
,
Grandval
A
,
Cuhna-Alves
L
,
Lechapt-Zalcman
E
,
Bernaudin
M
.
Hypoxia-inducible factor-2alpha (HIF-2alpha), but not HIF-1alpha, is essential for hypoxic induction of class III beta-tubulin expression in human glioblastoma cells
.
FEBS J
2014
;
281
:
5220
36
.
42.
Ostrom
QT
,
Bauchet
L
,
Davis
FG
,
Deltour
I
,
Fisher
JL
,
Langer
CE
, et al
.
The epidemiology of glioma in adults: a “state of the science” review
.
Neuro Oncol
2014
;
16
:
896
913
.
43.
Lim
M
,
Xia
Y
,
Bettegowda
C
,
Weller
M
.
Current state of immunotherapy for glioblastoma
.
Nat Rev Clin Oncol
2018
;
15
:
422
42
.
44.
Bregy
A
,
Shah
AH
,
Diaz
MV
,
Pierce
HE
,
Ames
PL
,
Diaz
D
, et al
.
The role of Gliadel wafers in the treatment of high-grade gliomas
.
Expert Rev Anticancer Ther
2013
;
13
:
1453
61
.
45.
Kunwar
S
,
Chang
S
,
Westphal
M
,
Vogelbaum
M
,
Sampson
J
,
Barnett
G
, et al
.
Phase III randomized trial of CED of IL13-PE38QQR vs Gliadel wafers for recurrent glioblastoma
.
Neuro Oncol
2010
;
12
:
871
81
.

Supplementary data