Purpose:

Apoptotic dysregulation, redox adaptive mechanisms, and resilience to hypoxia are major causes of glioblastoma (GBM) resistance to therapy. Commonly known as crucial factors in energy metabolism, OCTN2 (SLC22A5) and its substrate L-carnitine (LC) are increasingly recognized as actors in cytoprotection. This study provides a comprehensive expression and survival analysis of the OCTN2/LC system in GBM and clarifies the system's impact on GBM progression.

Experimental Design:

OCTN2 expression and LC content were measured in 121 resected human GBM specimens and 10 healthy brain samples and analyzed for prognostic significance. Depending on LC administration, the effects of hypoxic, metabolic, and cytotoxic stress on survival and migration of LN18 GBM cells were further studied in vitro. Finally, an orthotopic mouse model was employed to investigate inhibition of the OCTN2/LC system on in vivo GBM growth.

Results:

Compared with healthy brain, OCTN2 expression was increased in primary and even more so in recurrent GBM on mRNA and protein level. High OCTN2 expression was associated with a poor overall patient survival; the unadjusted HR for death was 2.7 (95% CI, 1.47–4.91; P < 0.001). LC administration to GBM cells increased their tolerance toward cytotoxicity, whereas siRNA-mediated OCTN2 silencing led to a loss of tumor cell viability. In line herewith, OCTN2/LC inhibition by meldonium resulted in reduced tumor growth in an orthotopic GBM mouse model.

Conclusions:

Our data indicate a potential role of the OCTN2/LC system in GBM progression and resistance to therapy, and suggests OCTN2 as a prognostic marker in patients with primary GBM.

Translational Relevance

Glioblastoma (GBM) is highly resistant to treatment, largely due to disease heterogeneity and resistance mechanisms. Understanding the mechanisms that generate resistance is essential for developing more effective treatment strategies. Recent studies provide evidence that OCTN2 and its substrate L-carnitine (LC) function as a cytoprotective system that strengthens cellular robustness. Here, we report on the expression and prognostic impact of the OCTN2/LC system in resected specimens of patients with newly diagnosed and recurrent GBM compared with healthy brain. We identified a high OCTN2 expression profile to correlate with a poor prognosis in patients with primary GBM, especially in those with a holistic therapeutical approach (total tumor resection, radiochemotherapy according to the Stupp protocol). The mechanistic studies indicate that inhibition of the OCTN2/LC system could reduce survival of GBM cells through enhanced sensitivity to exogenous influences such as hypoxic, metabolic, and cytotoxic stress. Furthermore, the OCTN2/LC inhibitor meldonium diminished in vivo tumor growth in an orthotopic GBM mouse model. In summary, our study stresses the role of OCTN2/LC as an actor in GBM cytoprotection, representing a potential target for clinical therapies aimed to slow the growth and progression of GBM.

Glioblastoma (GBM), classified as WHO grade IV glioma, represents the most frequent and most aggressive type of primary brain tumor in adults. Despite multimodal therapy including surgical resection followed by adjuvant radiation and chemotherapy, GBMs are characterized by rapid tumor recurrence resulting in a poor prognosis with a median survival of only 12–15 months (1). Although a number of genetic and epigenetic alterations were discovered in recent decades, many of the molecular mechanisms underlying the resistance of GBM cells remain largely elusive, leading to a lack of substantial progress in the therapeutic management of GBM. Several potential reasons exist as to how these tumors acquire treatment resistance, including enhanced expression of drug efflux transporters, alterations in drug metabolism, mutations of drug targets, and the activation of survival or inactivation of death signaling pathways (2, 3). Furthermore, studies argue for a potential role of altered redox homeostasis and energy metabolism in the development of antitumoral drug resistance (4, 5).

In recent years, evidence has emerged suggesting that the amino acid derivate L-carnitine (trimethylamine-β-hydroxybutyrate, LC), which is transported into the cells mainly by the Na+-dependent transporter OCTN2 (SLC22A5), contributes to cell protection by interacting with various targets inside the cell. Besides its key role in energy metabolism, several studies attest the OCTN2/LC system a broad spectrum of capabilities including scavenging free radicals, stabilizing membranes, enhancing antioxidative resources, and promoting antiapoptotic pathways (6–9). Therefore, the OCTN2/LC system has been widely discussed as a possible treatment adjunct in some neurodegenerative disorders such as Parkinson's and Alzheimer's disease to stabilize cellular integrity of compromised neuronal cells (10, 11). However, it has not yet been investigated whether GBM cells may also benefit from the OCTN2/LC system to increase their survivability, and whether OCTN2 and LC could be prognostic markers in GBM progression.

LC-mediated cytoprotection is closely linked to elevated levels of glutathione peroxidase, catalase and superoxide peroxidase (12–14), and to the induction of the NRF2 (nuclear factor erythroid 2-like) transcriptional network, one of the major pathways used by both normal and cancerous cells to counteract oxidative insults (15). Given that GBM has a high proliferation rate combined with an elevated basal metabolic turnover and formation of reactive oxygen species (ROS) as a natural byproduct (16), cancer cells may upregulate their antioxidant resources and survival pathways to prevent irreversible cellular damage (17, 18). Depending on its pleiotropic effects, accumulation of LC may strengthen tumor cell resistance as anticancer treatments rely, in part, on the destructive effect of ROS in their target cells (19, 20).

The OCTN2/LC system exists in almost all tissues, including the brain, as it is involved in essential metabolic processes (21). Well-known as a key compound in the “carnitine shuttle,” LC facilitates the transport of middle- and long-chain acids into mitochondria to subsequently undergo β-oxidation, allowing cells to metabolize fatty acids as energy resource (22). Similar to other tumors, GBMs have long been thought to rely upon glycolysis for energy production, while current studies suggest that glucose accounts for only <50% of acetyl-CoA production in gliomas (23). However, it was shown that fatty acid metabolism serves as an alternative energy source to promote GBM progression, whereas inhibition of β-oxidation led to prolonged survival in a GBM mouse model (5).

With a broad spectrum of cytoprotective effects and of metabolic relevance, the OCTN2/LC system may be a potential actor in cell resistance, but its role in GBM biology has not yet been explored. Here, we report on the expression and prognostic impact of the OCTN2/LC system in resected tumor specimens of patients with newly diagnosed and recurrent GBM. We further aimed to investigate the influence of LC on GBM cell survival and migration in vitro to verify its underlying cytoprotective capabilities. To finally assess the OCTN2/LC system as a therapeutic target, we analyzed the influence of the OCTN2/LC inhibitor meldonium on in vivo GBM growth using an orthotopic mouse model.

Human samples

Following an institutional review board–approved protocol (in accordance with the ethical standards of the Helsinki Declaration of the World Medical Association), fresh human GBM tissues were obtained from patients undergoing surgical resection or biopsy within their therapeutic regimen at the Department of Neurosurgery, University Medicine Greifswald (Greifswald, Germany), in the period from April 2007 to August 2016. At our university hospital, approximately 2 patients with glioblastoma are undergoing surgery every month. Because written informed consent was mandatory for both cryopreservation of the specimens and obtainment of vital status from official population registry, a total of 121 patients finally served as cohort for this study. Baseline epidemiologic and clinical characteristics of all patients are shown in Table 1. On the basis of histologic confirmation according to the 2007 WHO Classification of Tumors of the CNS (24), resected specimens included 80 primary GBM and 41 recurrences of primary GBM. Furthermore, 24 tumor samples from patients with astrocytoma grade II or III were included in this study. Eight nonmalignant brain tissues (frontal and temporal lobes) were obtained by routine autopsies. The patients died of pneumonia, heart failure, sepsis or pancreatic cancer, but had no underlying brain disease. In addition, 2 further nonmalignant brain samples (1 frontal and 1 temporal) were obtained from the BioChain Institute Inc.. All resected tumor samples and control brain tissues were cut and frozen at −80°C.

Table 1.

Clinicopathologic characteristics of the analyzed patient cohort

Entire glioma patient cohort
Parameters   Patient cohort 
Glioma subtype   (n
 GBM WHO grade IV   121 
 Primary GBM   80 
 Relapses of primary GBM   41 
 Astrocytoma WHO grade II/III   24 
Parameters Patients with primary GBM Relapses of primary GBM Astrocytoma WHO grade II/III 
Age at diagnosis [years]   
 Median 65 59 37 
 Min.–Max. 25–83 25–79 1–59 
Age classes (n, %)   
 <50 years 7 (8.75) 9 (21.95) 21 (87.5) 
 50–59 years 24 (30) 11 (26.83) 3 (12.5) 
 60–70 years 22 (27.5) 15 (36.59) 0 (0) 
 >70 years 27 (33.75) 6 (14.63) 0 (0) 
Sex (n, %)   
 Male 52 (65) 32 (78) 20 (83.33) 
 Female 28 (35) 9 (22) 4 (16.67) 
Patients with primary glioblastoma 
Parameters  Median survival [days/months] Range of survival [days/months] 
Tumor resection status (n, %)   
 Biopsy 2 (2.5)   
 50% tumor resection 3 (3.75)   
 Total tumor resection (macroscopic) 42 (52.5)   
 Subtotal tumor resection 26 (32.5)   
 Unknown 7 (8.75)   
Therapeutic regimen (n, %)   
 RCTx according to Stupp and colleagues 49 (61.25)   
 Radiomonotherapy 26 (32.5)   
 No adjuvant therapy 3 (3.75)   
 Unknown 2 (2.5)   
Overall survival (n, %)   
 All patients with known vital status 76 (95) 275/9 33–1780/1.1–58.5 
 Patients with RCTx according to Stupp and colleagues 45 (56.25) 493/16.2 75–1780/2.5–58.5 
 Patients with radiomonotherapy 26 (32.5) 152.5/4.5 33–992/1.1–32.6 
 Patients with total tumor resection 37 (46.25) 373/12.3 75–1512/2.5–49.7 
 Patients with subtotal tumor resection 22 (27.5) 147/4.8 33–617/1.1–20.3 
Survival rates (n, %)   
 1-year survival 35 (43.75)   
 2-year survival 16 (20)   
Vital status at study end (08/2016) (n, %)   
 Dead 70 (87.5)   
 Alive 6 (7.5)   
 Unknown 4 (5)   
Entire glioma patient cohort
Parameters   Patient cohort 
Glioma subtype   (n
 GBM WHO grade IV   121 
 Primary GBM   80 
 Relapses of primary GBM   41 
 Astrocytoma WHO grade II/III   24 
Parameters Patients with primary GBM Relapses of primary GBM Astrocytoma WHO grade II/III 
Age at diagnosis [years]   
 Median 65 59 37 
 Min.–Max. 25–83 25–79 1–59 
Age classes (n, %)   
 <50 years 7 (8.75) 9 (21.95) 21 (87.5) 
 50–59 years 24 (30) 11 (26.83) 3 (12.5) 
 60–70 years 22 (27.5) 15 (36.59) 0 (0) 
 >70 years 27 (33.75) 6 (14.63) 0 (0) 
Sex (n, %)   
 Male 52 (65) 32 (78) 20 (83.33) 
 Female 28 (35) 9 (22) 4 (16.67) 
Patients with primary glioblastoma 
Parameters  Median survival [days/months] Range of survival [days/months] 
Tumor resection status (n, %)   
 Biopsy 2 (2.5)   
 50% tumor resection 3 (3.75)   
 Total tumor resection (macroscopic) 42 (52.5)   
 Subtotal tumor resection 26 (32.5)   
 Unknown 7 (8.75)   
Therapeutic regimen (n, %)   
 RCTx according to Stupp and colleagues 49 (61.25)   
 Radiomonotherapy 26 (32.5)   
 No adjuvant therapy 3 (3.75)   
 Unknown 2 (2.5)   
Overall survival (n, %)   
 All patients with known vital status 76 (95) 275/9 33–1780/1.1–58.5 
 Patients with RCTx according to Stupp and colleagues 45 (56.25) 493/16.2 75–1780/2.5–58.5 
 Patients with radiomonotherapy 26 (32.5) 152.5/4.5 33–992/1.1–32.6 
 Patients with total tumor resection 37 (46.25) 373/12.3 75–1512/2.5–49.7 
 Patients with subtotal tumor resection 22 (27.5) 147/4.8 33–617/1.1–20.3 
Survival rates (n, %)   
 1-year survival 35 (43.75)   
 2-year survival 16 (20)   
Vital status at study end (08/2016) (n, %)   
 Dead 70 (87.5)   
 Alive 6 (7.5)   
 Unknown 4 (5)   

Cell lines

The human GBM cell lines A-172, GaMG, HF66, U251MG, U373, LN18, U87MG (bought from the ATCC), the murine cell line GL261, and patient derived primary GBM cells were used for the qualitative protein assessment. LN18, a WHO grade IV GBM cell line, was further used as in vitro model for the mechanistic studies. Because of its similarity to human GBM growth characteristics (25), GL261 cells were selected for our mouse model of malignant glioma. Detailed information on all cell lines are provided in Supplementary Table S1. Only cells at low passages were used for the experiments. By employing a PCR-based assay, all cell lines were routinely monitored for potential Mycoplasma contamination.

Quantitative real-time PCR analysis

Total RNA was isolated using PeqGold RNAPure (PeqLab) and reverse transcripted using the High Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific). Quantitative real-time PCR was performed on a 7900HT Fast Real-Time PCR system (Thermo Fisher Scientific) using the following Gene Expression Assays on Demand (Thermo Fisher Scientific): OCTN2/SLC22A5, Hs00929869_m1; CPT1A, Hs00912671_m1; CPT1C, Hs00380581_m1; and eukaryotic 18S rRNA endogenous control, 4310893E. Target gene expression was normalized to 18S rRNA and presented as box plots with the median and the 5th/95th percentiles in relation to the average expression in the control samples using ΔΔCt method.

Western blot analysis

Preparation of primary GBM cells was done as described previously (26). OCTN2-rich placenta membrane vesicles were prepared as described elsewhere (27). Preparation of lysates from the investigated cell lines as well as the detailed steps of frozen tissue sample homogenization, protein measurement, and subsequent immunoblot analysis are described in the Supplementary Data.

Measurement of LC in GBM tissue

Free LC was measured in tissue samples using the L-carnitine Colorimetric/Fluorometric Assay Kit (BioVision) according to the manufacturer's protocol. Samples were analyzed on a TECAN Infinite M200 (Tecan) multimode reader (excitation: 535 nm, emission: 587 nm). LC concentrations were normalized to the protein content of each sample (measured in the homogenized tissue suspended in PBS) determined by the BCA method.

Generation of hypoxic conditions in cell culture

To achieve hypoxic conditions, LN18 GBM cells were cultivated in a hypoxia chamber (<0.5 Vol.-% O2; GENbox) for 24 to 96 hours using a hypoxia pad (GENbox anaer Generator, BioMerieux) at 37°C.

Cell viability assays

Cells were seeded in 96-well multiplates (5,000 cells/well) using 150-μL culture medium containing 0.05% FCS. After 24 hours, cells were incubated with LC alone or in combination with etomoxir, temozolomide, or H2O2 under either normoxic or hypoxic conditions as described above. Afterwards, medium was replaced by fresh medium containing 10% resazurine (PromoCell) and cells were incubated for 2 hours at 37°C, before cell viability was analyzed on multimode reader (TECAN Infinite M200, Tecan) by fluorescence measurement (excitation: 530 nm; emission: 590 nm). Viability data were calculated as percentage of solvent-treated cells.

OCTN2 silencing using RNAi

LN18 GBM cells were cultured in 12- and 96-well plates and transfected with OCTN2 or control siRNA (sc-42560/sc-37007, Santa Cruz Biotechnology, 5 pmol/well) using Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol. To enhance knockdown effectivity siRNA transfection was repeated after 24 hours.

L-[3H]-carnitine accumulation assay

The functional activity of OCTN2 in control and OCTN2-siRNA–transfected LN18 cells was analyzed by L-[N-methyl- 3H]-carnitine (3H-carnitine, 80Ci/mmol, American Radiolabeled Chemicals Inc.) uptake assay. Forty-eight hours after transfection cells were washed using prewarmed PBS and incubated with L-[3H]-carnitine in transport buffer (final concentration 1 μCi/mL, 12,5 nmol/L) for 30 minutes. LC uptake was stopped by washing thrice using ice-cold PBS before cells were lysed using 300-μL lysis buffer/well (0.2% SDS, 5 mmol/L EDTA). A total of 150 μL of the lysates were mixed with 2-mL scintillation liquid Rotiszint eco plus (Carl Roth) and analyzed on a β-Counter Tri-Carb 2810TR Low Activity Liquid Scintillation Analyzer (PerkinElmer Inc.). The uptake rates were normalized to the protein content of each sample lysate.

Caspase-3 activity assay

Cells were seeded on 12-well plates (0.1 × 106 cells/well). After preincubation with LC or aqua dest, in medium containing 0.05% FCS for 6 hours, cells were treated with temozolomide, carmustine, vincristine or H2O2 for 48 hours. Afterwards, apoptotic activity was studied by measuring caspase-3 activity using the Caspase-3 Fluorometric Assay (R&D Systems) according to manufacturer's instructions.

Cell migration analysis

Cells were seeded in 24-well plates. After reaching confluence, the cell layer was scratched with a 100-μL pipette tip and washed 3 times with PBS. From each scratch 3 randomly distributed images were taken with PALM RoboSoftware on a AxioObserver.Z1 microscope (Zeiss). Afterwards, cells were cultivated with DMEM supplemented with 0.05% FCS, and 5 mmol/L hydroxyurea to prevent cell proliferation. LC was added for 24 hours. The scratches were analyzed as described above after 24 hours at the same positions. Finally, the scratch area was calculated with ImageJ (NIH, Bethesda, MD).

Orthotopic GBM mouse model

To evaluate OCTN2 as a suitable therapeutic target, we used the murine GBM cell line GL261, which was stereotactically implanted into the brain of C57BL/6J mice. The animal study has been evaluated and approved according to animal welfare guidelines and German laws on animal welfare by the Landesveterinär- und Lebensmitteluntersuchungsamt Mecklenburg-Vorpommern. The detailed procedures and housing conditions are described in the Supplementary Data.

Statistical analysis

Statistical analyses were performed using SPSS 21.0 (IBM) and GraphPad Prism 6 (GraphPad Software) and considered significant at *, P < 0.05; **, P < 0.01; ***, P < 0.001 level. Data are shown as mean ± SD. The tests used for statistical comparison are indicated in the respective figure legends. Overall survival was calculated from the time of diagnosis until death at the last follow-up. Information on vital status and date of death were obtained from official population registry.

OCTN2 expression is enhanced in GBM on mRNA and protein level

Compared with nonmalignant brain (NMB), a stepwise 2.3- to 4.4-fold increase of OCTN2 mRNA expression was measured in astrocytoma grade III (AC-III) and GBM specimens, respectively (Fig. 1A), while its expression was not elevated in astrocytoma grade II (AC-II) samples. Subdivision into primary and relapsed GBM revealed no significant differences (4.3-fold vs. 4.5-fold, Fig. 1B). In comparison with NMB, OCTN2 mRNA expression was similar in U87MG cells, while LN18 cells had 3-fold higher mRNA levels (Fig. 1B). Immunoblot analysis also revealed a significant upregulation of OCTN2 on protein level in primary GBM (34.5-fold) and relapsed tumor samples (37.2-fold; Fig. 1C and E). LN18 and U87MG had 3.5-fold and about 2-fold higher OCTN2 protein levels compared with NMB, respectively. Expression of OCTN2 was detectable in various human glioma cell lines (A-172, GaMG, HF66, LN18, U251MG, U373, U87MG), in a patient-derived culture of primary GBM cells and in the murine GL261 cell line. OCTN2 expression was validated by a positive control of high OCTN2–expressing placenta vesicles (Fig. 1D). More detailed information on the used cell lines are provided in Supplementary Table S1.

Figure 1.

mRNA and protein expression of OCTN2 in glioblastoma in comparison with nonmalignant brain. A, qRT-PCR analyses demonstrating OCTN2 mRNA expression levels in frontal/temporal lobes of nonmalignant brain (NMB, control), astrocytoma grade II (AC-II), astrocytoma grade III (AC-III), and glioblastoma (GBM) specimens. B, OCTN2 mRNA expression in NMB, primary GBM (pr. GBM), relapsed GBM (rel. GBM), and in the GBM cell lines LN18 and U87MG. C, Representative immunoblots of total protein lysates from NMB, GBM, and LN18 cell line analyzed for OCTN2 protein expression, each well representing an individual GBM sample numbered with a 3-digit code (anonymized sample identification), NMB: F, frontal lobe; T, temporal lobe. D, Qualitative assessment of OCTN2 protein expression in different GBM cell lines (A-172, GaMG, GL261, HF66, LN18, U251MG, U373, U87MG), patient derived primary (pr.) GBM cells and high OCTN2–expressing placenta vesicles (positive control) determined by immunoblot analysis. Detection of β-actin was used as loading control. E, Densitometric analyses of protein expression determined by Western blot (WB) in GBM in comparison with NMB. F, Uptake of 3H-labeled LC (12.5 nmol/L, 30 minutes) in LN18 cells in sodium-containing and sodium-free buffer (sodium replaced by choline) as well as after treatment with the OCTN2 inhibitor meldonium (100 μmol/L) in sodium-containing buffer, n = 3 (each in triplicates). G and H, Correlation between OCTN2 mRNA and protein expression in primary GBM (G) and relapsed GBM (H; Spearman correlation analysis). I, LC concentration in NMB as well as primary and relapsed GBM specimen normalized to the protein content of each individual sample. Box plots of patient sample data are shown with median and lower/upper quartile, whiskers from 5th to 95th percentile. One-way ANOVA (*, P < 0.05; **, P < 0.01; ***, P < 0.001; arb., arbitrary).

Figure 1.

mRNA and protein expression of OCTN2 in glioblastoma in comparison with nonmalignant brain. A, qRT-PCR analyses demonstrating OCTN2 mRNA expression levels in frontal/temporal lobes of nonmalignant brain (NMB, control), astrocytoma grade II (AC-II), astrocytoma grade III (AC-III), and glioblastoma (GBM) specimens. B, OCTN2 mRNA expression in NMB, primary GBM (pr. GBM), relapsed GBM (rel. GBM), and in the GBM cell lines LN18 and U87MG. C, Representative immunoblots of total protein lysates from NMB, GBM, and LN18 cell line analyzed for OCTN2 protein expression, each well representing an individual GBM sample numbered with a 3-digit code (anonymized sample identification), NMB: F, frontal lobe; T, temporal lobe. D, Qualitative assessment of OCTN2 protein expression in different GBM cell lines (A-172, GaMG, GL261, HF66, LN18, U251MG, U373, U87MG), patient derived primary (pr.) GBM cells and high OCTN2–expressing placenta vesicles (positive control) determined by immunoblot analysis. Detection of β-actin was used as loading control. E, Densitometric analyses of protein expression determined by Western blot (WB) in GBM in comparison with NMB. F, Uptake of 3H-labeled LC (12.5 nmol/L, 30 minutes) in LN18 cells in sodium-containing and sodium-free buffer (sodium replaced by choline) as well as after treatment with the OCTN2 inhibitor meldonium (100 μmol/L) in sodium-containing buffer, n = 3 (each in triplicates). G and H, Correlation between OCTN2 mRNA and protein expression in primary GBM (G) and relapsed GBM (H; Spearman correlation analysis). I, LC concentration in NMB as well as primary and relapsed GBM specimen normalized to the protein content of each individual sample. Box plots of patient sample data are shown with median and lower/upper quartile, whiskers from 5th to 95th percentile. One-way ANOVA (*, P < 0.05; **, P < 0.01; ***, P < 0.001; arb., arbitrary).

Close modal

The uptake of LC in cells by OCTN2 is known to depend on sodium (27). To analyze the extent to which LC-uptake into LN18 cells is mediated by OCTN2, uptake of tritium-labeled LC was investigated both in sodium-containing and sodium-free medium as well as in the presence of the OCTN2 inhibitor meldonium (28). The results demonstrate a sodium-dependent LC uptake via OCTN2, which is sensitive to meldonium (Fig. 1F).

Subsequent correlation analyses revealed a positive association between OCTN2 mRNA and protein expression in both primary GBM samples (Spearman r = 0.5327, P < 0.001; Fig. 1G) and relapsed tumor specimens (Spearman r = 0.6108, P < 0.001; Fig. 1H). Furthermore, the tissue content of the OCTN2 substrate LC was analyzed. LC concentration was elevated in both primary (4-fold) and relapsed GBM (5-fold) compared with NMB (P < 0.001 and P = 0.001, respectively; Fig. 1I), but did not correlate with the OCTN2 expression (Fig. 2E and F).

Figure 2.

Survival and correlation analyses of OCTN2 and LC in primary GBM. Kaplan–Meier estimates for patients with primary GBM based on their OCTN2 mRNA (A) and protein (B) expression. Patients were divided into 2 subgroups depending on median mRNA or protein expression, log-rank test, **, P < 0.01 and ***, P < 0.001. C, Summary of the survival analysis from A and B showing the median survival time and the 1- and 2-year survival rate (1-YS, 2-YS) for both OCTN2 mRNA and protein expression at a glance. D, Kaplan–Meier estimates for patients with GBM based on their intratumoral LC levels. Patients were divided into 2 subgroups depending on median LC content, log-rank test. E, Correlation between OCTN2 mRNA and intratumoral LC level in primary GBM (Spearman correlation analysis). F, Correlation between OCTN2 protein expression and intratumoral LC level in primary GBM (Spearman correlation analysis).

Figure 2.

Survival and correlation analyses of OCTN2 and LC in primary GBM. Kaplan–Meier estimates for patients with primary GBM based on their OCTN2 mRNA (A) and protein (B) expression. Patients were divided into 2 subgroups depending on median mRNA or protein expression, log-rank test, **, P < 0.01 and ***, P < 0.001. C, Summary of the survival analysis from A and B showing the median survival time and the 1- and 2-year survival rate (1-YS, 2-YS) for both OCTN2 mRNA and protein expression at a glance. D, Kaplan–Meier estimates for patients with GBM based on their intratumoral LC levels. Patients were divided into 2 subgroups depending on median LC content, log-rank test. E, Correlation between OCTN2 mRNA and intratumoral LC level in primary GBM (Spearman correlation analysis). F, Correlation between OCTN2 protein expression and intratumoral LC level in primary GBM (Spearman correlation analysis).

Close modal

OCTN2 overexpression correlates with poor patient survival

On the basis of the median OCTN2 mRNA/protein expression, the GBM patient cohort was divided into a high and a low expression group. As demonstrated in Fig. 2A and B, a high OCTN2 mRNA or protein expression was associated with a significantly worse survival time. On mRNA level, the median survival time for the low OCTN2–expressing group were 528 days (17.4 months) compared with 268 days (8.8 months) of the high OCTN2–expressing group (Fig. 2A). The unadjusted HR for death was 2.16 (95% CI, 1.25–3.73, P = 0.005) for a high OCTN2 mRNA expression. For OCTN2 protein expression, the survival values were 254 days (8.4 months) for the high OCTN2–expressing group and 579 days (19 months) for the low OCTN2–expressing group [unadjusted HR: 2.69 (95% CI, 1.47–4.91, P < 0.001; Fig. 2B)]. Later, the HR for death was adjusted by fitting the Cox proportional-hazard models. In addition to the stratification factors (extent of surgery, therapeutic regimen), other possible confounding factors such as age and sex were included. The adjusted HR for death in the high OCTN2 protein– expressing group was 2.94 (95% CI, 1.29–6.67, P = 0.01), whereas the adjusted HR for the mRNA dataset did not reach statistical significance (P = 0.06). As a result, the 1-year and 2-year survival rates of patients with GBM also varied markedly according to the OCTN2 mRNA and protein expression levels (Fig. 2C). On the basis of the protein dataset, 1 year after diagnosis, 26% of GBM patients with a high OCTN2 tumor expression were alive compared with 64% with a low intratumoral OCTN2 content. A similar observation was also made 2 years after diagnosis with survival rates of 6% (high OCTN2 expression) versus 30% in the low OCTN2–expressing group.

The LC concentration in primary GBM was not associated with the survival time (Fig. 2D). The median survival time for patients with GBM having a high intratumoral LC level (>median) accounted for 381 days versus 408 days for patients with a lower LC content [<median; adjusted HR for death: 1.01 (95% CI, 0.5–2.04, P = 0.982)].

Impact of OCTN2 expression on patient survival fitted on potential confounders

In addition to the Cox proportional-hazards models, ANOVA II analyses were performed to determine the extent to which OCTN2 expression is influenced by the therapeutic regimen, resection status, age, and sex. For these analyses, our patient cohort was subdivided into 4 age classes (<50 years, 50–59 years, 60–70 years, and > 70 years at diagnosis) with nearly the same group size. The ANOVA II analyses revealed no significant interactions between the OCTN2 mRNA or protein expression and age at diagnosis (Supplementary Fig. S1E and S1F). However, OCTN2 mRNA and protein expression tended to be lower in female patients. In line with the results given in the Cox regression, ANOVA II analysis revealed a significant influence of the OCTN2 expression level on survival time of patients with GBM in each of the 4 age groups [P = 0.04 (mRNA) and P = 0.01 (protein), Fig. 3A and B].

Figure 3.

ANOVA II analysis of OCTN2 mRNA and protein expression on patient survival with regard to potential confounders. ANOVA II analysis of patient survival depending on the OCTN2 mRNA (A) or protein (B) expression in the 4 age classes. OCTN2 expression was divided according to the median. Influence of OCTN2 mRNA (C) and protein (E) expression on GBM patient survival depending on the therapeutic regimen (RCTx treatment vs. radiomonotherapy). Influence of OCTN2 mRNA (D) or protein (F) expression on patient survival depending on the resection status of the patients (total vs. subtotal tumor resection). Tukey-HSD post hoc test (*, P < 0.05; **, P < 0.01).

Figure 3.

ANOVA II analysis of OCTN2 mRNA and protein expression on patient survival with regard to potential confounders. ANOVA II analysis of patient survival depending on the OCTN2 mRNA (A) or protein (B) expression in the 4 age classes. OCTN2 expression was divided according to the median. Influence of OCTN2 mRNA (C) and protein (E) expression on GBM patient survival depending on the therapeutic regimen (RCTx treatment vs. radiomonotherapy). Influence of OCTN2 mRNA (D) or protein (F) expression on patient survival depending on the resection status of the patients (total vs. subtotal tumor resection). Tukey-HSD post hoc test (*, P < 0.05; **, P < 0.01).

Close modal

Furthermore, the patient survival time for both the RCTx and radiomonotherapy group is significantly shorter at high OCTN2 levels in the resected GBM samples [vs. low OCTN2 expression level, Fig. 3C and E, P = 0.04 (mRNA) and P = 0.001 (protein)]. A similar effect was observed for the resection status [Fig. 3D and F, P = 0.04 (mRNA) and P = 0.008 (protein)]. It is noteworthy that in patients with a total tumor resection, the effect of OCTN2 expression is somewhat more pronounced in comparison with the subtotal resection group. Regarding this aspect, specific subanalysis via Cox regression revealed an adjusted HR of 4.6 for death in the high OCTN2 protein–expressing group with a total tumor resection (95% CI, 1.63–13.12, P = 0.004), and an adjusted HR of 3.3 for patients having received RCTx (95% CI, 1.31–8.37, P = 0.01).

Influence of LC and OCTN2 on survival of LN18 GBM cells in vitro

To investigate the impact of OCTN2 and its substrate LC in GBM cells, in vitro experiments using the LN18 GBM cell line were performed. Cells treated with LC had only a slightly increased viability after 48-hour serum deprivation (116%), which, however, was significantly elevated in a 72-hour period of starvation (142%, Fig. 4A). Furthermore, administration of temozolomide, the chemotherapeutic agent used in standard of care treatment of GBM, caused a decreased viability of LN18 cells cultured in serum-starved medium (78%). This effect was reversed in the presence of 50 μmol/L LC (109%).

Figure 4.

Influence of LC and OCTN2 on survival of LN18 GBM cells in vitro. A, Analysis of LN18 cell viability after treatment with LC using the resazurine assay. Cells were treated for 48 or 72 hours with 50 μmol/L LC or solvent (aqua dest.) in serum-starved medium (SSM, 0.05% FCS). Cell viability was normalized to untreated control cells (100%; Mann–Whitney U test; ***, P < 0.001 vs. solvent). B, Influence of LC on temozolomide (TMZ)-induced loss of LN18 cell viability. Cells were treated either with temozolomide (100 μmol/L) alone or in combination with 50 μmol/L LC for 72 hours in SSM. Cell viability was normalized to solvent (DMSO + aqua dest.) treated control cells (100%; 1-way ANOVA; *, P < 0.05; ***, P < 0.001 vs. solvent, #, P < 0.01 TMZ + LC vs. 100 μmol/L TMZ alone). C, Influence of LC on H2O2-induced loss of LN18 cell viability. Cells were preincubated for 24 hours with 50 μmol/L LC followed by application of H2O2 (10, 50, or 100 μmol/L) either alone or in combination with 50 μmol/L LC for 30 minutes in SSM. After further 24 hours, the cell viability was determined, 1-way ANOVA (*, P < 0.05; ***, P < 0.001 vs. solvent (aqua dest.); #, P < 0.05; ##, P < 0.01 H2O2 + LC vs. H2O2 alone). Impact of etomoxir (ETO) on LN18 cell viability under normoxia (D, 21 Vol.-% O2) and hypoxia (E, 0.5 Vol.-% O2). Cells were pretreated for 24 hours with 50 μmol/L LC followed by application of 100 μmol/L ETO either alone or in combination with 50 μmol/L LC. After 48, 72, or 96 hours, cell viability was measured [1-way ANOVA; ***, P < 0.001 vs. aqua dest. (solvent); ##, P < 0.01; ###, P < 0.001 ETO + LC vs. ETO alone]. F–H, siRNA-mediated knockdown of OCTN2 expression in LN18 cells. F, OCTN2 mRNA expression 48 and 72 hours after transfection with either scrambled control siRNA or OCTN2-specific siRNA (unpaired t test, ***, P < 0.001 vs. control siRNA). G, LC uptake into LN18 cells transfected with either scrambled control siRNA or OCTN2-specific siRNA. Cells were incubated with 100 nmol/L 3H-labeled LC at 37°C for 30 minutes (unpaired t test **, P < 0.01 vs. control siRNA). H, Determination of cell viability using resazurine assay of LC-treated (50 μmol/L) LN18 cells 48 or 72 hours after transfection with either scrambled control siRNA or OCTN2-specific siRNA under normoxia (21 Vol.-% O2) and hypoxia (0.5 Vol.-% O2). Cell viability was normalized to cells transfected with scrambled control siRNA (100%; 1-way ANOVA *, P < 0.05; ***, P < 0.001 vs. control siRNA; #, P < 0.05 normoxia vs. hypoxia). All data are shown as mean ± SD (n = 3; each in triplicates; arb., arbitrary).

Figure 4.

Influence of LC and OCTN2 on survival of LN18 GBM cells in vitro. A, Analysis of LN18 cell viability after treatment with LC using the resazurine assay. Cells were treated for 48 or 72 hours with 50 μmol/L LC or solvent (aqua dest.) in serum-starved medium (SSM, 0.05% FCS). Cell viability was normalized to untreated control cells (100%; Mann–Whitney U test; ***, P < 0.001 vs. solvent). B, Influence of LC on temozolomide (TMZ)-induced loss of LN18 cell viability. Cells were treated either with temozolomide (100 μmol/L) alone or in combination with 50 μmol/L LC for 72 hours in SSM. Cell viability was normalized to solvent (DMSO + aqua dest.) treated control cells (100%; 1-way ANOVA; *, P < 0.05; ***, P < 0.001 vs. solvent, #, P < 0.01 TMZ + LC vs. 100 μmol/L TMZ alone). C, Influence of LC on H2O2-induced loss of LN18 cell viability. Cells were preincubated for 24 hours with 50 μmol/L LC followed by application of H2O2 (10, 50, or 100 μmol/L) either alone or in combination with 50 μmol/L LC for 30 minutes in SSM. After further 24 hours, the cell viability was determined, 1-way ANOVA (*, P < 0.05; ***, P < 0.001 vs. solvent (aqua dest.); #, P < 0.05; ##, P < 0.01 H2O2 + LC vs. H2O2 alone). Impact of etomoxir (ETO) on LN18 cell viability under normoxia (D, 21 Vol.-% O2) and hypoxia (E, 0.5 Vol.-% O2). Cells were pretreated for 24 hours with 50 μmol/L LC followed by application of 100 μmol/L ETO either alone or in combination with 50 μmol/L LC. After 48, 72, or 96 hours, cell viability was measured [1-way ANOVA; ***, P < 0.001 vs. aqua dest. (solvent); ##, P < 0.01; ###, P < 0.001 ETO + LC vs. ETO alone]. F–H, siRNA-mediated knockdown of OCTN2 expression in LN18 cells. F, OCTN2 mRNA expression 48 and 72 hours after transfection with either scrambled control siRNA or OCTN2-specific siRNA (unpaired t test, ***, P < 0.001 vs. control siRNA). G, LC uptake into LN18 cells transfected with either scrambled control siRNA or OCTN2-specific siRNA. Cells were incubated with 100 nmol/L 3H-labeled LC at 37°C for 30 minutes (unpaired t test **, P < 0.01 vs. control siRNA). H, Determination of cell viability using resazurine assay of LC-treated (50 μmol/L) LN18 cells 48 or 72 hours after transfection with either scrambled control siRNA or OCTN2-specific siRNA under normoxia (21 Vol.-% O2) and hypoxia (0.5 Vol.-% O2). Cell viability was normalized to cells transfected with scrambled control siRNA (100%; 1-way ANOVA *, P < 0.05; ***, P < 0.001 vs. control siRNA; #, P < 0.05 normoxia vs. hypoxia). All data are shown as mean ± SD (n = 3; each in triplicates; arb., arbitrary).

Close modal

Later, the influence of LC on H2O2-induced oxidative stress in LN18 cells was evaluated. Coadministration of LC overcompensated (up to 108%) the viability loss of H2O2-treated LN18 cells (89%) compared with the respective control cells (Fig. 4C). To subsequently assess whether the cytoprotective effect of LC depends on its function as a mitochondrial fatty acids shuttle and thus on metabolic effects based on β-oxidation, the carnitine palmitoyltransferase-1 (CPT-1) inhibitor etomoxir (ETO) (29) was used. Furthermore, the experiments were carried out under hypoxic conditions in parallel to examine additional hypoxic stress. As shown in Fig. 4D and E, ETO reduced the viability of LN18 cells significantly. The effect of ETO was more pronounced under hypoxic conditions. Under both normoxic and hypoxic conditions, the cell viability loss was partially reversed by administration of 50 μmol/L LC.

In addition, the impact of OCTN2 on GBM cell growth was studied after siRNA-mediated OCTN2 knockdown. siRNA treatment resulted in significantly reduced OCTN2 expression and function as shown by mRNA analysis and 3H-labeled LC uptake experiments (Fig. 4F and G). Subsequently, we determined the influence of OCTN2 knockdown on viability of LC-pretreated (50 μmol/L) LN18 cells (Fig. 4H). Under both normoxic and hypoxic conditions knockdown of OCTN2 caused a significant loss of viability of LN18 cells which was, again, more pronounced in cells confronted with hypoxic stress.

Influence of LC on apoptosis and migration of LN18 GBM cells

To analyze whether LC protects GBM cells from apoptosis caused either by cytostatics or oxidative stress, caspase-3 activity was used as a marker for apoptotic processes. LN18 cells treated with LC showed no significant alterations in caspase-3 activity (Fig. 5A). In contrast, doxorubicin (7-fold) and carmustine (BCNU, 1.8-fold) led to a significant increase in caspase-3 activity after 48 hours. Coadministration of LC reduced this effect in BCNU-treated cells [1.8-fold (BCNU alone) to 1.2-fold (BCNU + LC)], but not for doxorubicin. Temozolomide and H2O2 had no effect on caspase-3 activity. Furthermore, cell migration was analyzed using a wound closure assay since migration of GBM cells into healthy brain is a main feature of this brain tumor. Treatment with BCNU and temozolomide did not alter cell migration, whereas administration of ETO, an inhibitor of β-oxidation, significantly reduced migration of LN18 cells to about 50%. Coadministration of LC did not cause any changes in migration (Fig. 5B). After the detection of ETO-induced inhibition of LN18 cell migration, we assessed expression of 2 key enzymes of the mitochondrial β-oxidation, CPT1A (carnitine palmitoyltransferase 1A) and CPT1C (carnitine palmitoyltransferase 1C), in GBM tissue. CPT1A and CPT1C showed a similar expression pattern in NMB compared with GBM specimens (Supplementary Figs. S2A and S2B). Kaplan–Meier analyses revealed no significant influence of CPT1A or CPT1C expression on patient survival indicating a limited prognostic value in GBM (Supplementary Figs. S2C and S2D).

Figure 5.

Influence of LC on caspase-3 activity and migration of LN18 GBM cells as well as analysis of OCTN2 as a therapeutic target in an orthotopic in vivo mouse model. A, Analysis of caspase-3 activity in LN18 cells after treatment with doxorubicin (Doxo, 1 μmol/L), temozolomide (TMZ, 100 μmol/L), carmustine (BCNU, 100 μmol/L), H2O2 (200 μmol/L), or solvent (DMSO for doxorubicin, TMZ, and BCNU; H2O for H2O2) either alone or in combination with LC (50 μmol/L). Cells were treated with the indicated substances for 48 hours in serum-starved medium (SSM, 0.05% FCS). Caspase-3 activity was normalized to solvent-treated control cells, 2-way ANOVA with Bonferroni post hoc test (**, P < 0.01 vs. control cells; ##, P < 0.05 for with vs. without LC (arb., arbitrary). B, Wound closure assay for determination of the migratory capacity of LN18 cells after stimulation with etomoxir (ETO, 100 μmol/L), BCNU (250 μmol/L), temozolomide (250 μmol/L), and DMSO (control) either alone or in combination with LC (50, 100, and 500 μmol/L). The migratory capacity is depicted as wound closure in percent after an incubation time of 24 hours [2-way ANOVA with Bonferroni post hoc test; ***, P < 0.001 vs. control cells (without any drugs), all data are shown as mean ± SD]. C and D, Influence of the OCTN2 inhibitor meldonium on in vivo GBM growth in an orthotopic mouse model using murine GL261 GBM cells. Mice were intraperitoneally treated from day 12 to 26 postinjection every day with either 0.9% NaCl (control animals, n = 10) or 250 mg/kg meldonium (n = 9) as OCTN2 inhibitor. Tumor volume was calculated with Horos software in coronal and axial gadolinium-enhanced T1-weighted sections 12 days and 26 days postinjection; data were log-transformed. C shows individual animals while D shows the mean ± SD, unpaired t test, *, P < 0.05. E, Comparison of tumor growth rates in control group versus meldonium-treated group. Growth rate was calculated as difference between the log-transformed tumor volumes on day 26 (V2) and day 12 (V1). Positive values indicate growth while negative values indicate downsizing, unpaired t test, *, P < 0.05. F, Representative gadolinium-enhanced T1-weighted images (coronal) for animals with low, mid, and high tumor growth from each group (control: 0.9% NaCl; meldonium: 250 mg/kg) at the first day of pharmacologic intervention (day 12 postinjection) and at the end of the study (day 26 postinjection).

Figure 5.

Influence of LC on caspase-3 activity and migration of LN18 GBM cells as well as analysis of OCTN2 as a therapeutic target in an orthotopic in vivo mouse model. A, Analysis of caspase-3 activity in LN18 cells after treatment with doxorubicin (Doxo, 1 μmol/L), temozolomide (TMZ, 100 μmol/L), carmustine (BCNU, 100 μmol/L), H2O2 (200 μmol/L), or solvent (DMSO for doxorubicin, TMZ, and BCNU; H2O for H2O2) either alone or in combination with LC (50 μmol/L). Cells were treated with the indicated substances for 48 hours in serum-starved medium (SSM, 0.05% FCS). Caspase-3 activity was normalized to solvent-treated control cells, 2-way ANOVA with Bonferroni post hoc test (**, P < 0.01 vs. control cells; ##, P < 0.05 for with vs. without LC (arb., arbitrary). B, Wound closure assay for determination of the migratory capacity of LN18 cells after stimulation with etomoxir (ETO, 100 μmol/L), BCNU (250 μmol/L), temozolomide (250 μmol/L), and DMSO (control) either alone or in combination with LC (50, 100, and 500 μmol/L). The migratory capacity is depicted as wound closure in percent after an incubation time of 24 hours [2-way ANOVA with Bonferroni post hoc test; ***, P < 0.001 vs. control cells (without any drugs), all data are shown as mean ± SD]. C and D, Influence of the OCTN2 inhibitor meldonium on in vivo GBM growth in an orthotopic mouse model using murine GL261 GBM cells. Mice were intraperitoneally treated from day 12 to 26 postinjection every day with either 0.9% NaCl (control animals, n = 10) or 250 mg/kg meldonium (n = 9) as OCTN2 inhibitor. Tumor volume was calculated with Horos software in coronal and axial gadolinium-enhanced T1-weighted sections 12 days and 26 days postinjection; data were log-transformed. C shows individual animals while D shows the mean ± SD, unpaired t test, *, P < 0.05. E, Comparison of tumor growth rates in control group versus meldonium-treated group. Growth rate was calculated as difference between the log-transformed tumor volumes on day 26 (V2) and day 12 (V1). Positive values indicate growth while negative values indicate downsizing, unpaired t test, *, P < 0.05. F, Representative gadolinium-enhanced T1-weighted images (coronal) for animals with low, mid, and high tumor growth from each group (control: 0.9% NaCl; meldonium: 250 mg/kg) at the first day of pharmacologic intervention (day 12 postinjection) and at the end of the study (day 26 postinjection).

Close modal

Influence of the OCTN2/LC inhibitor meldonium on in vivo GBM growth

To finally show proof of the concept of the OCTN2/LC system as a therapeutic target for GBM treatment, we analyzed the in vivo tumor growth in an orthotopic murine GBM model using the cell line GL261, which was stereotactically injected into mice brain. Twelve days postinjection, tumor development was assessed by MRI. Afterwards, mice were treated with either 0.9% NaCl (control animals) or 250 mg/kg meldonium as an OCTN2/LC inhibitor (28) every day intraperitoneally until day 26 postinjection. To ensure normality of data, tumor volumes were log-transformed. Also, tumor growth was calculated as the difference between the transformed volumes. As seen in Fig. 5C–E at day 26 postinjection (14 days after starting treatment), a decrease in mean tumor size was observed in the meldonium-treated group (−2,366 log10[cm3]) compared with the starting time point (−2,306 log10[cm3]). In contrast, control animals showed significant progress in tumor size from −2,385 log10[cm3] (treatment starting point) to −1,618 log10[cm3] at day 26, which was significantly different from the corresponding time point of the meldonium group (P < 0.05). This significant difference was also present in the growth rate calculations of the tumors (Fig. 5E), which showed a mean tumor growth rate of 0,7671 log10[cm3] in control animals and −0,06014 log10[cm3] in meldonium-treated animals (P = 0.01). Three representative MRI recordings for the starting time point (12 days postinjection) and at day 26 postinjection per group (low, mid, high tumor growth) are shown in Fig. 5F.

Therapy failure caused by GBM resistance is the most common reason of tumor recurrence resulting in very poor survival rates. It has been previously shown that the OCTN2 substrate LC acts as a cytoprotector promoting cellular resistance and survivability (7, 30, 31). In this study, we demonstrate that OCTN2 expression and tissue LC concentrations are significantly higher in GBM compared with healthy brain. We observed a stepwise increase of OCTN2 expression from low-grade astrocytoma to primary and recurrent GBM. These findings are consistent with those of Bayraktar and colleagues who described a gradual increase in LC levels and some of its acyl derivates from low- to high-grade astrocytomas through to primary GBM (32).

In a recent study, Singer and colleagues described the development of therapeutic resistance in GBM stem cells by enhanced expression of an antioxidant response system referred to as Xc catalytic subunit xCT (SLC7A11) that partially operates through activation of the NRF2 transcriptional network (19). This network is also addressed by LC (15, 31) and regulates antioxidant response element (ARE)-containing genes (33) leading to attenuation of ROS-mediated cell damage, arrest of apoptosis, and resistance to radiochemotherapy (31, 34). ROS homeostasis is strictly regulated by cancer cells to promote tumorigenesis and malignant transformation (17, 35, 36). OCTN2 upregulation and LC accumulation could therefore help GBM cells to maintain conducive ROS levels both to drive mutagenesis and to mitigate unfavorable conditions evoked by ROS-generating processes, for example, high metabolic turnover and radiochemotherapy (19, 20).

These considerations are underlined by our findings that LN18 GBM cells, a well-accepted WHO grade IV GBM cell line (37), become more resistant or at least maintain their viability under hypoxic, metabolic, and cytotoxic stress when pretreated with LC. The results are consistent with several in vitro and in vivo studies on LC's antioxidant and antiapoptotic activities, which have been predominantly studied in nonmalignant compromised cells in context of neurodegenerative disorders (10, 11), but are also detectable in the malignant neuroblastoma cell line SH-SY5Y (12). However, LC enhanced the viability of the human LN18 GBM cells, but had no detectable effect on cell migration. Following administration of LC, we also observed a decreased caspase-3 activity in LN18 cells treated with BCNU and a restored viability in cells treated with temozolomide, both drugs used in the therapeutic regimen for patients with GBM. These results endorse the theory that LC strengthens the tumor cells through antiapoptotic mechanisms or perhaps through a combination of several cytoprotective effects. However, the cytoprotective LC concentrations given in literature are widely spread within a range of 9–25 μmol/L (38), 30–100 μmol/L (39), 0.1–1 mmol/L (40), and 1–10 mmol/L (41), respectively. In our current work, a LC concentration of 50 μmol/L was found to be adequate in detecting cytoprotective effects. Lower LC concentrations showed no significant benefit in our in vitro experiments while higher LC concentrations did not increase the survival-promoting effects (data not shown) even though other studies claim a concentration dependent effect of LC (6, 12, 30). Of note, the LC concentration used in our cell experiments was within the physiologic range of the LC plasma concentration, which is generally estimated to be 40–50 μmol/L (42).

Recent NMR analyses of in situ glioma specimens as well as further in vitro and in vivo studies suggest that oxidative metabolism plays an integral role of cellular maintenance and proliferation in malignant glioma cells (5). Because LC is crucial for transporting middle- and long-chain fatty acids into mitochondria to drive β-oxidation, some consideration had to be given to how LC's metabolic effects could influence the tumor cells' behavior. We used the irreversible CPT1 inhibitor etomoxir (ETO) to block β-oxidation to analyze whether GBM cells rely on fatty acid oxidation and to what extent cytoprotection is attributable to LC when uncoupled from its native function as a mitochondrial shuttle. ETO caused both a significant loss of cell viability and migration in LN18 cells, which has been also shown in primary GBM cells (5, 29). Under both normoxic and hypoxic cell culture conditions, the loss of viability, but not the ETO-induced inhibition of migration, was abolished by simultaneous LC application. These results provide 3 valuable hints: First, LN18 relies to a remarkable extent on fatty acid metabolism as bioenergetic fuel and lacks viability and migration when β-oxidation is blocked. Second, LC stabilizes cell integrity independently of its metabolic function, but rather by its abovementioned extramitochondrial cytoprotective effects (31, 34). Third, LC did not reverse or alleviate the ETO-induced inhibition of migration suggesting that LC has no perceptible influence on GBM cell migration based on metabolic effects. It is quite possible, however, that malignant GBM cells gain survival advantages on both cytoprotective and metabolic levels through OCTN2 overexpression and LC accumulation, as it is also assumed that fatty acid oxidation provides NADPH for defense against oxidative stress and cell death in GBM cells (29). Nevertheless, the molecular details of malignant transformation and resistance of tumor cells need to be further elaborated in this context.

To our knowledge, no association studies on the role of the OCTN2/LC system in GBM survival have been published to date. Here, after adjustment of the HR on therapeutic regimen, resection status, age, and sex, we found a high OCTN2 protein expression to correlate with a 2.9-fold increased risk for death in patients with primary GBM. However, it has to be mentioned that further potential confounders (e.g., ECOG Performance Status, Karnofsky Index) were not recorded. The ANOVA II analyses revealed that differences in the patient survival between the low and high OCTN2–expressing group were highly pronounced in the respective subpopulation receiving RCTx and a total tumor resection, suggesting a prognostic relevance especially in holistically treated patients.

Although OCTN2 expression has been described in various tumor cells (43–45), its precise role in malignant transformation remains elusive. Contrary to our observations, Scalise and colleagues found a downregulation of OCTN2 expression in epithelial cancer cell lines, whereas an estrogen-mediated induction of OCTN2 expression in breast cancer was described (43, 44). For GBM, however, our data of increased OCTN2 expression may provide both a prognostic factor for GBM treatment as well as an option for optimizing GBM therapy by either specific blocking of OCTN2 or drug targeting via OCTN2. Very recently, it was shown that LC-conjugated nanoparticles promote permeation across the blood–brain barrier to target glioma cells via OCTN2 resulting in improved antiglioma therapy (46). Our preclinical studies using an orthotopic GBM mouse model demonstrate significantly reduced intracerebral tumor growth through inhibition of the OCTN2/LC system and suggest it as a potential target in GBM therapy. To date, our in vivo study is the first to show the antitumoral efficacy of the OCTN2/LC inhibitor meldonium. While no major side effects of meldonium have been observed so far, meldonium has even been shown to improve cardiovascular function (47, 48).

Some limitations of our study need to be addressed. Our work represents a global expression analysis of the entire tumor mass without tumor region or cell type–specific differentiation, especially considering the heterogeneity of GBM. Taking into account the different types of GBM and their distinct genetic and epigenetic profiles (49), predictive molecular biomarkers for therapy response and outcome may vary in respective GBM subtypes. Our patient cohort was not subclassified and we therefore cannot draw any conclusion regarding differences in GBM subtypes. In our study, only vital tumor samples from the edge region were used. Within the same tumor, hypoxic, necrotic, and highly vascularized areas vary in their expression profiles and thus also in their OCTN2 expression. In addition, the use of only 1 cell line for in vitro and in vivo studies is a limitation of our investigations, arguing toward analyses in further human cell lines or primary GBM cells to validate the observed effects.

Nevertheless, this study stresses the role of OCTN2 and its substrate LC in GBM cytoprotection, suggests OCTN2 as a prognostic marker and offers a novel target that may aid in combatting tumor resistance or increasing its sensitivity to therapy. Although the precise mechanisms of the OCTN2/LC system in GBM biology remain to be completely determined, the promising results of meldonium in reducing tumor growth provide strong support for further research on the OCTN2/LC system, which is aimed at improving antiglioma therapy.

No potential conflicts of interest were disclosed.

Conception and design: M.A. Fink, H. Paland, S. Bien-Möller

Development of methodology: M.A. Fink, H. Paland

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.A. Fink, H. Paland, S. Herzog, M. Grube, S. Vogelgesang, A. Bialke, W. Hoffmann, H.W.S. Schroeder, S. Bien-Möller

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M.A. Fink, H. Paland, K. Weitmann, W. Hoffmann, B.H. Rauch, S. Bien-Möller

Writing, review, and/or revision of the manuscript: M.A. Fink, H. Paland, M. Grube, S. Vogelgesang, K. Weitmann, A. Bialke, W. Hoffmann, B.H. Rauch, S. Bien-Möller

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.A. Fink, H. Paland, S. Bien-Möller

Study supervision: M.A. Fink, H.W.S. Schroeder, S. Bien-Möller

This work was supported by national funding from the Forschungsverbund Neurowissenschaften. M.A. Fink received a scholarship of the Gerhard Domagk Program, funded by the Faculty of Medicine, Greifswald, Germany, and was further supported by the Germany Scholarship, funded by the German Federal Ministry for Education and Research.

We thank Prof. N. Hosten, head of the Institute for Radiology and Neuroradiology (University Medicine Greifswald) for providing the 7 Tesla MR imaging scanner and Stefan Hadlich for the excellent technical assistance with the MR imaging.

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
,
Mason
WP
,
van den Bent
MJ
,
Weller
M
,
Fisher
B
,
Taphoorn
MJB
, et al
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma
.
N Engl J Med
2005
;
352
:
987
96
.
2.
Liu
Y
,
Li
Q
,
Zhou
L
,
Xie
N
,
Nice
EC
,
Zhang
H
, et al
Cancer drug resistance: redox resetting renders a way
.
Oncotarget
2016
;
7
:
42740
61
.
3.
Holohan
C
,
Van Schaeybroeck
S
,
Longley
DB
,
Johnston
PG
. 
Cancer drug resistance: an evolving paradigm
.
Nat Rev Cancer
2013
;
13
:
714
26
.
4.
Rinaldi
M
,
Caffo
M
,
Minutoli
L
,
Marini
H
,
Abbritti
R
,
Squadrito
F
, et al
ROS and brain gliomas: an overview of potential and innovative therapeutic strategies
.
Int J Mol Sci
2016
;
17
:
984
15
.
5.
Lin
H
,
Patel
S
,
Affleck
VS
,
Wilson
I
,
Turnbull
DM
,
Joshi
AR
, et al
Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells
.
Neuro-Oncol
2016
;
19
:
43
54
.
6.
Li
J-L
,
Wang
Q-Y
,
Luan
H-Y
,
Kang
Z-C
,
Wang
C-B
. 
Effects of L-carnitine against oxidative stress in human hepatocytes: involvement of peroxisome proliferator-activated receptor alpha
.
J Biomed Sci
2012
;
19
:
32
.
7.
Gülçin
İ
. 
Antioxidant and antiradical activities of L-carnitine
Life Sci
2006
;
78
:
803
11
.
8.
Chao
H-H
,
Liu
J-C
,
Hong
H-J
,
Lin
J-W
,
Chen
C-H
,
Cheng
T-H
. 
L-carnitine reduces doxorubicin-induced apoptosis through a prostacyclin-mediated pathway in neonatal rat cardiomyocytes
.
Int J Cardiol
2011
;
146
:
145
52
.
9.
Chen
HH
,
Sue
YM
,
Chen
CH
,
Hsu
YH
,
Hou
CC
,
Cheng
CY
, et al
Peroxisome proliferator-activated receptor alpha plays a crucial role in L-carnitine anti-apoptosis effect in renal tubular cells
.
Nephrology Dialysis Transplant
2009
;
24
:
3042
9
.
10.
Ribas
GS
,
Vargas
CR
,
Wajner
M
. 
l-carnitine supplementation as a potential antioxidant therapy for inherited neurometabolic disorders
.
Gene
2014
;
533
:
469
76
.
11.
Hudson
SA
,
Tabet
N
. 
Acetyl-l-carnitine for dementia
.
Chichester, United Kingdom
:
John Wiley & Sons, Ltd
; 
2003
.
12.
Yu
J
,
Ye
J
,
Liu
X
,
Han
Y
,
Wang
C
. 
Protective effect of L-carnitine against H(2)O(2)-induced neurotoxicity in neuroblastoma (SH-SY5Y) cells
.
Neurol Res
2011
;
33
:
708
16
.
13.
Lee
B-J
,
Lin
J-S
,
Lin
Y-C
,
Lin
P-T
. 
Effects of L-carnitine supplementation on oxidative stress and antioxidant enzymes activities in patients with coronary artery disease: a randomized, placebo-controlled trial
.
Nutr J
2014
;
13
:
1360
7
.
14.
Cao
Y
,
Li
X
,
Shi
P
,
Wang
L-X
,
Sui
Z-G
. 
Effects of L-Carnitine on high glucose-induced oxidative stress in retinal ganglion cells
.
Pharmacology
2014
;
94
:
123
30
.
15.
Li
J
,
Zhang
Y
,
Luan
H
,
Chen
X
,
Han
Y
,
Wang
C
. 
l-carnitine protects human hepatocytes from oxidative stress-induced toxicity through Akt-mediated activation of Nrf2 signaling pathway
.
Can J Physiol Pharmacol
2016
;
94
:
517
25
.
16.
Salazar-Ramiro
A
,
Ramírez-Ortega
D
,
Pérez de la Cruz
V
,
Hérnandez-Pedro
NY
,
González-Esquivel
DF
,
Sotelo
J
, et al
Role of redox status in development of glioblastoma
.
Front Immunol
2016
;
7
:
157
15
.
17.
Cairns
RA
,
Harris
IS
,
Mak
TW
. 
Regulation of cancer cell metabolism
.
Nat Rev Cancer
2011
;
11
:
1
11
.
18.
Fruehauf
JP
,
Meyskens
FL
. 
Reactive oxygen species: a breath of life or death?
Clin Cancer Res
2007
;
13
:
789
94
.
19.
Singer
E
,
Judkins
J
,
Salomonis
N
,
Matlaf
L
,
Soteropoulos
P
,
McAllister
S
, et al
Reactive oxygen species-mediated therapeutic response and resistance in glioblastoma
.
Cell Death Dis
2015
;
6
:
e1601
1
.
20.
Wondrak
GT
. 
Redox-directed cancer therapeutics: molecular mechanisms and opportunities
.
Antioxid Redox Signal
2009
;
11
:
3013
69
.
21.
Tamai
I
,
Ohashi
R
,
Nezu
J
,
Yabuuchi
H
,
Oku
A
,
Shimane
M
, et al
Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2
.
J Biol Chem
1998
;
273
:
20378
82
.
22.
Bieber
LL
. 
Carnitine
.
Annu Rev Biochem
1988
;
57
:
261
83
.
23.
Maher
EA
,
Marin-Valencia
I
,
Bachoo
RM
,
Mashimo
T
,
Raisanen
J
,
Hatanpaa
KJ
, et al
Metabolism of [U-13C]glucose in human brain tumors in vivo
.
NMR Biomed
2012
;
25
:
1234
44
.
24.
Louis
DN
,
Ohgaki
H
,
Wiestler
OD
,
Cavenee
WK
,
Burger
PC
,
Jouvet
A
, et al
The 2007 WHO classification of tumours of the central nervous system
Acta Neuropathol
2007
;
114
:
97
109
.
25.
Newcomb
EW
,
Zagzag
D
. 
The murine GL261 glioma experimental model to assess novel brain tumor treatments
.
In:
Meir
EG
,
editor
.
CNS cancer: models, markers, prognostic factors, targets, and therapeutic approaches
,
Totowa, NJ
:
Humana Press
; 
2009
. p.
227
41
.
26.
Bien-Möller
S
,
Lange
S
,
Holm
T
,
Böhm
A
,
Paland
H
,
Küpper
J
, et al
Expression of S1P metabolizing enzymes and receptors correlate with survival time and regulate cell migration in glioblastoma multiforme
.
Oncotarget
2016
;
7
:
13031
46
.
27.
Grube
M
,
Meyer Zu Schwabedissen
H
,
Draber
K
,
Präger
D
,
Möritz
K-U
,
Linnemann
K
, et al
Expression, localization, and function of the carnitine transporter octn2 (slc22a5) in human placenta
.
Drug Metab Dispos
2005
;
33
:
31
7
.
28.
Grube
M
,
Meyer zu Schwabedissen
HEU
,
Präger
D
,
Haney
J
,
Möritz
K-U
,
Meissner
K
, et al
Uptake of cardiovascular drugs into the human heart: expression, regulation, and function of the carnitine transporter OCTN2 (SLC22A5)
.
Circulation
2006
;
113
:
1114
22
.
29.
Pike
LS
,
Smift
AL
,
Croteau
NJ
,
Ferrick
DA
,
Wu
M
. 
Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells
Biochim Biophys Acta
2011
;
1807
:
726
34
.
30.
Yildirim
S
,
Yildirim
A
,
Dane
S
,
Aliyev
E
,
Yigitoglu
R
. 
Dose-dependent protective effect of L-carnitine on oxidative stress in the livers of hyperthyroid rats
.
Eurasian J Med
2013
;
45
:
1
6
.
31.
Hota
KB
,
Hota
SK
,
Chaurasia
OP
,
Singh
SB
. 
Acetyl-L-carnitine-mediated neuroprotection during hypoxia is attributed to ERK1/2-Nrf2-regulated mitochondrial biosynthesis
.
Hippocampus
2012
;
22
:
723
36
.
32.
Bayraktar
N
. 
The relationship between carnitine levels and lipid peroxidation in glial brain tumors
.
Turkish J Med Sci
2008
;
38
:
293
9
.
33.
Kensler
TW
,
Wakabayashi
N
,
Biswal
S
. 
Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway
.
Annu Rev Pharmacol Toxicol
2007
;
47
:
89
116
.
34.
Zhu
J
,
Wang
H
,
Sun
Q
,
Ji
X
,
Zhu
L
,
Cong
Z
, et al
Nrf2 is required to maintain the self-renewal of glioma stem cells
.
BMC Cancer
2013
;
13
:
380
.
35.
DeNicola
GM
,
Karreth
FA
,
Humpton
TJ
,
Gopinathan
A
,
Wei
C
,
Frese
K
, et al
Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis
.
Nature
2011
;
475
:
106
9
.
36.
Gorrini
C
,
Harris
IS
,
Mak
TW
. 
Modulation of oxidative stress as an anticancer strategy
.
Nat Rev Drug Discov
2013
;
12
:
931
47
.
37.
Diserens
AC
,
de Tribolet
N
,
Martin-Achard
A
,
Gaide
AC
,
Schnegg
JF
,
Carrel
S
. 
Characterization of an established human malignant glioma cell line: LN-18
Acta Neuropathol
1981
;
53
:
21
8
.
38.
Yazaki
T
,
Hiradate
Y
,
Hoshino
Y
,
Tanemura
K
,
Sato
E
. 
L-carnitine improves hydrogen peroxide-induced impairment of nuclear maturation in porcine oocytes
.
Animal Sci J
2012
;
84
:
395
402
.
39.
Liu
F
,
Patterson
TA
,
Sadovova
N
,
Zhang
X
,
Liu
S
,
Zou
X
, et al
Ketamine-induced neuronal damage and altered N-methyl-d-aspartate receptor function in rat primary forebrain culture
.
Toxicol Sci
2012
;
131
:
548
57
.
40.
Mescka
CP
,
Wayhs
CAY
,
Guerreiro
G
,
Manfredini
V
,
Dutra-Filho
CS
,
Vargas
CR
. 
Prevention of DNA damage by l-carnitine induced by metabolites accumulated in maple syrup urine disease in human peripheral leukocytes in vitro
.
Gene
2014
;
548
:
294
8
.
41.
Tastekin
A
,
Gepdiremen
A
,
Ors
R
,
Emin Buyukokuroglu
M
,
Halici
Z
. 
l-carnitine protects against glutamate- and kainic acid-induced neurotoxicity in cerebellar granular cell culture of rats
.
Brain Develop
2005
;
27
:
570
3
.
42.
Reuter
SE
,
Evans
AM
. 
Carnitine and acylcarnitines: pharmacokinetic, pharmacological and clinical aspects
.
Clin Pharmacokinet
2012
;
51
:
553
72
.
43.
Wang
C
,
Uray
IP
,
Mazumdar
A
,
Mayer
JA
,
Brown
PH
. 
SLC22A5/OCTN2 expression in breast cancer is induced by estrogen via a novel intronic estrogen-response element (ERE)
.
Breast Cancer Res Treat
2012
;
134
:
101
15
.
44.
Scalise
M
,
Galluccio
M
,
Accardi
R
,
Cornet
I
,
Tommasino
M
,
Indiveri
C
. 
Human OCTN2 (SLC22A5) is down-regulated in virus- and nonvirus-mediated cancer
.
Cell Biochem Funct
2012
;
30
:
419
25
.
45.
Qu
Q
,
Qu
J
,
Zhan
M
,
Wu
L-X
,
Zhang
Y-W
,
Lou
X-Y
, et al
Different involvement of promoter methylation in the expression of organic cation/carnitine transporter 2 (OCTN2) in cancer cell lines
.
PLoS ONE
2013
;
8
:
e76474
10
.
46.
Kou
L
,
Hou
Y
,
Yao
Q
,
Guo
W
,
Wang
G
,
Wang
M
, et al
L-Carnitine-conjugated nanoparticles to promote permeation across blood-brain barrier and to target glioma cells for drug delivery via the novel organic cation/carnitine transporter OCTN2
.
Artif Cells Nanomed Biotechnol
2018
;
46
:
1605
16
.
47.
Dambrova
M
,
Liepinsh
E
,
Kalvinsh
I
. 
Mildronate: cardioprotective action through carnitine-lowering effect
.
Trends Cardiovasc Med
2002
;
12
:
275
9
.
48.
Sjakste
N
,
Gutcaits
A
,
Kalvinsh
I
. 
Mildronate: an antiischemic drug for neurological indications
.
CNS Drug Rev
2005
;
11
:
151
68
.
49.
Masui
K
,
Mischel
PS
,
Reifenberger
G
. 
Molecular classification of gliomas
.
Handb Clin Neurol
2016
;
134
:
97
120
.