Open access peer-reviewed chapter

Antioxidant Supplementation during Glioma Therapy: Friend or Foe?

Written By

Duygu Harmanci

Submitted: 27 November 2017 Reviewed: 09 April 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.77079

From the Edited Volume

Glioma - Contemporary Diagnostic and Therapeutic Approaches

Edited by Ibrahim Omerhodžić and Kenan Arnautović

Chapter metrics overview

1,187 Chapter Downloads

View Full Metrics

Abstract

Gliomas which are one of the most common types of primary brain tumors are originated from glial cells. Type of tumor and tumor location are the most important factors to determine the treatment options. The treatment options might be surgery, radiation therapy, chemotherapy, targeted therapies, and experimental clinical studies. Especially, in course of chemotherapy and radiotherapy, antioxidant levels decrease. Antioxidants fight against the oxidants’ negative effects, which include cell damage, oxidative stress, and so on. Recent years, some researchers present that the antioxidant using could be harmful in some cases. A growing body of evidence suggests that antioxidant supplementation might increase the mortality. In this chapter, an overview of antioxidants and their functions has been presented to introduce researchers to the changes and effects of the antioxidants in glioma treatment. The evidence-based studies have been summarized. These experimental studies are important to understand the right option for the patient and transfer the solution from bench to bed.

Keywords

  • glioma
  • treatment
  • antioxidant
  • oxidative stress
  • experimental studies

1. Introduction

Central nervous system tumors start in the brain or spinal cord [1]. The common symptoms of these tumors are headache, seizures, weakness, nausea, vomiting, and altered mental status [1, 2]. Gliomas are one of the most common primary brain tumors, which are originated from glial cells. In general, gliomas are classified astrocytoma, oligodendrogliomas, and ependymomas. According to World Health Organization (WHO), the histological classification of gliomas consists of astrocytoma, oligodendroglioma, oligoastrocytoma (low-grade gliomas) and anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic oligoastrocytoma, anaplastic ependymoma, and glioblastoma (high-grade gliomas) [1, 2, 3, 4]. The histological type of the tumor is the most significant thing to determine the treatment option [2].

Antioxidants are present in plant-based foods, for instance, some types of vegetables and fruits: wine, blueberry, different types of tea, grape, and so on [5, 6]. The main role of antioxidants is prevention of oxidants’ harmful effects to human body. Principle regarding oxidant-antioxidant is related to a balance [7]. This balance’s side determines the human body reaction. In case of elevated oxidant levels in organism body homeostasis is lost and oxidative stress occurs. Loss of this balance and oxidative stress lead some pathological situations: cancer, neurodegenerative diseases, cardiovascular diseases, immunological diseases, and so on [8, 9, 10]. In terms of these diseases with the antioxidant supplementation, cell damage can be fixed.

In this chapter, two different stories will be told and these stories will be turned one story. We will discuss some of the basic concepts of antioxidants, antioxidant systems and antioxidants supplementation and explain how antioxidant supplementation can help with the cancer therapy, especially glioma therapy. Experimental studies are summarized and present evidences are collected under three headings: in vitro studies, animal studies, and clinical trials.

Advertisement

2. What is antioxidant?

To understand the term antioxidant, we have to tell the story from the beginning. The story begins with oxygen. Oxygen is the main source of the life, but in the body oxygen sometimes acts like a foe. Oxygen has two unpaired electrons, which spin in the same direction [9]. For this reason, oxygen is a biradical, so it is a free radical. In general, free radicals are highly reactive compounds, which are called as “reactive oxygen species” (ROS). ROS are intracellular compounds, which consist of oxygen [7, 11]. The most known ROS are listed in Table 1.

NameMolecule formula
SuperoxideO2
HydroxylOH
PeroxylROO
AlcoxylRO
HydroperoxylHO2
Lipid peroxylLOO
Hydrogen peroxideH2O2

Table 1.

Reactive oxygen species (ROS).

Oxygen is less dangerous than oxygen-derived free radical species (superoxide, hydroxyl radicals, hydrogen peroxide, etc.), and they react with lipids, proteins, and nucleic acids [12, 13]. Besides ROS, nitrogen-derived molecules are present in human body. These are known as reactive-nitrogen species [6, 14]. These molecules can get involved with oxidant molecules, but all oxidants are not free radicals. They produce endogenously or with some exogenous sources’ effects [9, 10]. Some endogenous and exogenous sources are shown in Table 2.

Endogenous sourcesExogenous sources
Normal cellular metabolism
  • Electron transport chain

  • Neutrophils, macrophages

  • Mitochondrial cytochrome oxidase

  • Smooth muscle cells

  • Cortisol, catecholamine

  • Immune system cells

UV
Ozone exposure
Hyperoxia
Burning organic foods
Smoking
Ionizing radiation
Air pollutants
Heavy metal ions

Table 2.

Endogenous and exogenous sources of ROS.

In human body, antioxidant systems are present to avoid cell damage due to free radicals. These antioxidant systems include a few enzymes for this reason they are called enzymatic antioxidants [9, 10, 11, 14]. The definition of antioxidant that it is a molecule reacts with free radicals and neutralizes them [6]. Except enzymatic antioxidants, generally, they occur naturally in foods, especially plant-based foods [15]. For instance, resveratrol is a very popular antioxidant in recent years, and it is found in grape, raspberry, blueberry, wine, and so on [16]. The most known non-enzymatic antioxidants are low-molecular-weight compounds such as vitamin C, vitamin E, beta-carotene, catechins, lycopene, glutathione, and coenzyme Q [5, 12, 17].

In summary, the story starts with oxygen and develops free radicals and stable molecules (DNA, protein, lipids, carbohydrates, etc.). Antioxidants are the good cops and they get involved the free radicals. In normal conditions, this is acceptable as happy ending. In terms of biological perspective, in course of normal metabolism energy production starts with consumption of oxygen and food nutrients. Oxygen and food enter the cell and mitochondria start to produce adenosine triphosphate (ATP). Free radicals form during cell’s energy production. These free radicals are neutralized by antioxidant enzyme systems (superoxide dismutase, catalase, glutathione peroxidase, etc.) and non-enzymatic antioxidants [6]. In the presence of any pathological conditions, ROS are highly produced and although antioxidant enzyme systems and antioxidants try to eliminate them to protect the cell, they remain incapable. Redox balance breaks down, oxidative stress increases, and antioxidant levels decrease [14]. In terms of cancer, ROS imbalance is one of the hallmarks of cancer [18].

Advertisement

3. Antioxidants and cancer

Cancer is a malign disease, which is characterized by abnormal cell proliferation [19, 20]. The uncontrolled situation in the cell is a result of endogenous or exogenous effects. According to multistep carcinogenesis theory, cancer originated from one cell, so cancer is a monoclonal disease and it develops in three stages. These stages are initiation, promotion, and progression [21]. Cells suffer damage with any endogenous or exogenous effects. Defects or mutations accumulate in the cell with these effects. The main effects are listed below [22]:

  • Environmental factors,

  • Lifestyle,

  • Infections,

  • Mutations,

  • Inherited genetic diseases,

  • Viruses

  • Reactive oxygen species (ROS).

Aforementioned before ROS cause some pathological situations due to their reactive features [10]. They react with nucleic acids, proteins, lipids, and carbohydrates. As a result of this interaction, it is possible that cancer development may be from one cell. ROS may take a role any stages of carcinogenesis [7]. Proven roles of ROS on cancer progression [23]:

  • Some genetic alterations are generated by ROS.

  • ROS promote cell migration via invadopodia formation in vitro.

  • ROS activate the PI3K/AKT/mTOR and MAPK/ERK mitogenic signaling pathways.

The main objectives of oncological treatment are increasing life-quality and extending survival time. When these objectives are considered, antioxidant supplementation brings to mind some questions. If clinicians add antioxidants to therapy:

  • Does the success of therapy increase or decrease?

  • Are some of side-effects related to current therapy eliminated by antioxidants?

  • Does antioxidant supplementation affect survival rates?

In accordance with these questions, lots of experimental and clinical studies were carried out to prove the role of antioxidants in cancer therapy [16, 24, 25, 26, 27, 28, 29, 30]. Results obtained from these studies were variable. This variation is basically related to cancer type and cancer grade. ROS-cancer relationship and antioxidant junction points are described Figure 1.

Figure 1.

ROS-cancer relationship and antioxidant junction points.

An antioxidant-cancer relationship is deeply discussed next part in terms of glioma.

Advertisement

4. Antioxidants and gliomas relationship

Gliomas are a class of primary central nervous system tumors and they originated from glial cells [1]. Glial progenitor cells have different subtypes: astrocyte, oligodendrocyte, and ependyma. In general, the classification of gliomas is based on these cell types [4]. The most detailed classification belongs to WHO. WHO suggests that four different grades (I–II–III–IV) are described for gliomas according to morphological and histological features [1]. Besides these features, some molecular and genetic features (epidermal growth factor upregulation, isocitrate dehydrogenase 1/2 mutations, p53 mutations, etc.) also alter the grading [2].

Tumor grade and class are major factors to determine the therapy options. Surgery, chemotherapy, and radiotherapy are preferred to treat the gliomas. After surgery, chemotherapy or radiotherapy is applied. For the glioma treatment, the most frequently encountered problems are the blood-brain barrier and drug resistance [31]. The blood-brain barrier is a control mechanism in relation to the transition of ions, molecules, and cells between the blood and brain. If a drug does not pass through the blood-brain barrier, it cannot reach the brain cells [32].

The second problem is drug resistance [33]. Temozolomide is the most common chemotherapeutic agent for gliomas. It is an alkylating agent [34]. In case of elevated levels of O6-metil guanine DNA methyltransferase expression, temozolomide meets with resistance [35]. On the other hand, increased levels of antioxidant response system SLC7A11 triggered the drug resistance [31].

Over the past decades, antioxidant supplementation becomes a necessity for cancer treatment. Basically, antioxidants use to eliminate the elevated levels of ROS, but cancer in question nothing is understandable. For this reason, researchers have carried out some studies. Understanding the beneficial or harmful roles of antioxidants in cancer treatment is essential. Further to that understanding of ROS effects in terms of cancer progression is really important. ROS is a reason for cancer progression, but in course of cancer development increased levels of ROS might be a cell-death option. Moreover, increased levels of ROS alter the cell signaling in cancer cell in consequence of acting as secondary messengers [17]. For instance, Akt overexpression is frequently showed in gliomas, and protein kinase C (PKC) activation stimulates some molecules like Akt, MAPK. All these molecules are under the control by cellular redox state [36]. As a result of these features, ROS antioxidants can be provided new approaches in order to treat glioma. It is still an unknown and questionable area for the researchers.

Accumulating data suggest two different approaches regarding antioxidant consumption. One is that antioxidants make tumor cells resistance against chemotherapy or radiotherapy and the survival rates are decreased. On the other hand, the second is that antioxidants protect the normal cells from oxidative damage and they are decreased side effects of therapy and provide better survival [8, 37, 38]. The next part of this chapter is related to evidence regarding these two opinions.

4.1. Evidence-based studies

Gliomagenesis is still an unknown, incurable, and lethal process. New and effective treatment strategies are the necessity and understanding the gliomagenesis is essential in order to develop these options. Experimental evidence indicates that antioxidants are sometimes friend, and in some cases, they are the foe.

4.1.1. In vitro studies

In 1995, Zhu et al. carried out a study to clarify the effects of selenium on rat and human glioblastoma multiforme cell lines. They used sodium selenite and showed that selenium had anti-proliferative effects on both A172 human glioblastoma cells and C6 rat glioblastoma cells, but it was more effective on human glioblastoma cells [39].

In 1997, Vartak et al. showed that some polyunsaturated fatty acids: gamma-linoleic acid (GLA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) supplementations increased the radiosensitivity and also radiation response on 36B10 rat astrocytoma cells [40].

In 1998, Vartak et al. compared the effects of GLA and linoleic acid (LA) on 36B10 malignant rat astrocytoma and normal rat astrocytes. They found that GLA was cytotoxic for astrocytoma cells, but not astrocytes. LA was not effective for both cells. It suggested that GLA might be used for astrocytoma treatment [41].

In 1999, Arora-Kuruganti et al. examined roles of oxidant (H2O2) and antioxidant (N-acetyl-cysteine, NAC) on U373-MG astrocytoma cell line. They observed that tumor cell proliferation was inhibited by NAC. NAC also induced H2O2 [25].

In the beginning 2000s, the first study came from Rooprai et al. They checked some anti-invasive and anti-proliferative agents: swainsonine, captopril, tangeretin, and nobiletin on four different glioma cell lines: ependymoma, oligoastrocytoma, anaplastic astrocytoma, and glioblastoma multiforme. Firstly, they observed that each cell line showed difference response against agents. They found that the most effective agent was nobiletin on four different cell lines and it was decreased the MMP-2 and -9 secretions [42].

In 2001, Naidu et al. studied the effects of ascorbyl stearate (Asc-S) on human glioblastoma multiforme cells. They used different doses of Asc-S on T98G human glioblastoma cell lines for 24 h. They showed that Asc-S inhibited insulin-like growth factor-dependent cell proliferation in a dose-dependent manner. Asc-S modulated IGF-R expression, in consequence of this situation programmed cell death was triggered on T98G cells by Asc-S [43].

In 2007, Rooprai et al. studied on IPSB-18 human astrocytoma cells. They treated cells with selenite and found that selenite was altered the expressions of matrix metalloproteinases and their inhibitors. It was also decreased the epidermal growth factor (EGFR) expression. This was suggested that selenite had anti-metastatic effects [44].

In 2013, Pozsgai et al. studied on quercetin effects on glioblastoma standard treatment. They found that combination treatment provided significant reduction in cell viability in U251 and DBTRG-05MG glioblastoma multiforme cell lines. They also showed that quercetin alone, or in a combination with IR triggered the apoptosis [29]. In 2016, Lou et al. found that quercetin nanoparticles stimulated the autophagy and apoptosis by activating AKT/erk/caspase 3 signaling pathway [45].

In 2017, increasing cell proliferation of glioblastoma multiforme cell lines with low doses of selenomethionine was showed by Harmanci et al. [46].

The combination of berbamine and paclitaxel were decreased the cell proliferation on U87 glioblastoma multiforme cells [47].

Higher levels of ascorbate led the DNA strand breakages by creating genotoxic and metabolic stress on glioma cells, but it also caused the development of radioresistance [48].

4.1.2. Animal studies

In 1981, Newell et al. used a mixture of vitamins C and B12 in high dose on rats with glioma. They observed no difference in survival time between experimental and control groups [49].

In 1989, Wang et al. showed that retinoids (retinal, retinoic acid, retinyl acetate, and retinyl palmitate) and carotenoids (beta-carotene, lycopene, and crocetin) inhibited the tumor growth in C6 glioma cells inoculated rats [50].

A study regarding naringenin using was carried out on rats by Sabarinathan et al. [30]. With supplementation of naringenin in glioma induced rats the status of lipid peroxidation was decreased, on the contrary antioxidant status increased. Besides this, naringenin also modulate the glial-tumor cell proliferation [30].

In 2013, Perez de la Ossa et al. examined that Δ9-tetrahidrocannabinol (THC) and cannabidiol (CBD) effects on tumor growth in xenograft glioblastoma multiforme model. THC and CBD loaded on microparticles and delivered locally. At the end of the study they found that THC and CBD stimulated apoptosis and induced cell proliferation and angiogenesis [51].

In 2013, Hervouet et al. found that using SUVIMAX-like diet (supplementation en vitamins et minéraux antioxydants), which was enriched with beta carotene, alpha tocopherol, vitamin C, zinc, and sodium selenite, was delayed the clinical signs on ethyl-nitrosourea induced glioma rat model, but gliomagenesis occurred. This diet just decreased the tumor aggressiveness [52].

In 2017, prolonged survival time was showed treatment with coptis chinensis on glioma induced mice model by Li et al. [53].

Combination of berbamine and paxitaxel was delayed the development of tumor U87 xenograft model [47].

4.1.3. Clinical trials

In 1990, Philipov et al. carried out a limited clinical study with 15 patients with malignant brain tumors. There was no significant survival prolongation with selenium addition on patients’ diet [28].

In 1996, Lissoni et al. evaluated the effects of melatonin using with radiotherapy on 30 patients with glioblastoma multiforme. They showed that the melatonin addition in normal therapy provided prolonged survival time, decreased side-effects. Based on these results they suggested that concomitant therapy may be more effective for glioblastoma patients [27].

In 2010, Delorenze et al. exhibited that the relationship between daily intake of antioxidants and survival rate was variable depends on tumor grade. They also showed that the supplementation of higher dose vitamin E has increased the survival in grade III gliomas; otherwise, vitamin C and genistein were decreased the survival rate [26].

In 2010, the side-effects welding from radiotherapy were decreased with lycopene supplementation in patients with high-grade glioma [54].

In 2015, Mulpur et al. carried out a study to check complementary therapy options among glioblastoma multiforme patients. They found that multivitamins or omega-3-fatty acids did not affect survival, but for Vitamin D and E further investigations are necessity [55].

Advertisement

5. Conclusions

Cancer is a personal disease, for this reason, it needs special attention. The above-mentioned evidences have shown that antioxidant supplementation cannot safe at times. Researchers advocate two different opinions regarding using antioxidant in course of cancer treatment. The first opinion is traditional approach. It says antioxidants prevent the normal cell from oxidative damage and they induce toxicity and provide better survival rates. The second one shows the dark side of antioxidants. According to the second opinion: antioxidants are decreased the survival rates by triggering drug-resistance. When we consider these two opinions we can easily understand the requirement of this area.

The most urgent thing is clinical trials with larger sample size and long-term following. Evidence obtained from in vitro or in vivo studies cannot representative for the 3D organism. The effects of antioxidant supplementation can determine appropriate clinical trials. Antioxidants’ interference in chemotherapeutic mechanisms is still unknown and clinical fails of therapeutic approaches regarding redox modulation are obvious.

In summary, antioxidant cancer therapy remains incapable. ROS scavengers must give place to antioxidant inhibitors. ROS-related cell death mechanism is a novel approach to provide the selective cell death. Further investigations will need to see the effectiveness of pro-oxidant cancer therapy.

References

  1. 1. Ostrom QT, Gittleman H, Liao P, Vecchione-Koval T, Wolinsky Y, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010-2014. Neuro-Oncology. 2017;19(suppl_5):v1-v88. DOI: 10.1093/neuonc/nox158
  2. 2. Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, Nishikawa R, Rosenthal M, Wen PY, Stupp R, et al. Glioma. Nature Reviews Disease Primers. 2015;1:15017. DOI: 10.1038/nrdp.2015.17
  3. 3. Pisapia DJ. The updated World Health Organization glioma classification: Cellular and molecular origins of adult infiltrating gliomas. Archives of Pathology & Laboratory Medicine. 2017;141(12):1633-1645. DOI: 10.5858/arpa.2016-0493-RA
  4. 4. Schneider T, Mawrin C, Scherlach C, Skalej M, Firsching R. Gliomas in adults. Deutsches Ärzteblatt International. 2010;107(45):799-807. quiz 808. DOI: 10.3238/arztebl.2010.0799
  5. 5. Dai J, Mumper RJ. Plant phenolics: Extraction, analysis and their antioxidant and anticancer properties. Molecules. 2010;15(10):7313-7352. DOI: 10.3390/molecules15107313
  6. 6. Halliwell B. Free radicals and antioxidants—Quo vadis? Trends in Pharmacological Sciences. 2011;32(3):125-130. DOI: 10.1016/j.tips.2010.12.002
  7. 7. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chemico-Biological Interactions. 2006;160(1):1-40. DOI: 10.1016/j.cbi.2005.12.009
  8. 8. Benfeito S, Oliveira C, Soares P, Fernandes C, Silva T, Teixeira J, Borges F. Antioxidant therapy: Still in search of the ‘magic bullet’. Mitochondrion. 2013;13(5):427-435. DOI: 10.1016/j.mito.2012.12.002
  9. 9. Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organization Journal. 2012;5(1):9-19. DOI: 10.1097/WOX.0b013e3182439613
  10. 10. Ziech D, Franco R, Georgakilas AG, Georgakila S, Malamou-Mitsi V, Schoneveld O, Pappa A, Panayiotidis MI. The role of reactive oxygen species and oxidative stress in environmental carcinogenesis and biomarker development. Chemico-Biological Interactions. 2010;188(2):334-339. DOI: 10.1016/j.cbi.2010.07.010
  11. 11. Sies H. Oxidative stress: Oxidants and antioxidants. Experimental Physiology. 1997;82(2):291-295
  12. 12. Bicas JL, Neri-Numa IA, Ruiz AL, De Carvalho JE, Pastore GM. Evaluation of the antioxidant and antiproliferative potential of bioflavors. Food and Chemical Toxicology. 2011;49(7):1610-1615. DOI: 10.1016/j.fct.2011.04.012
  13. 13. Murphy MP. How mitochondria produce reactive oxygen species. The Biochemical Journal. 2009;417(1):1-13. DOI: 10.1042/BJ20081386
  14. 14. Cooke MS, Evans MD, Dizdaroglu M, Lunec J. Oxidative DNA damage: Mechanisms, mutation, and disease. The FASEB Journal. 2003;17(10):1195-1214. DOI: 10.1096/fj.02-0752rev
  15. 15. Hasani-Ranjbar S, Larijani B, Abdollahi M. A systematic review of the potential herbal sources of future drugs effective in oxidant-related diseases. Inflammation & Allergy Drug Targets. 2009;8(1):2-10
  16. 16. de la Lastra CA, Villegas I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical implications. Biochemical Society Transactions. 2007;35(Pt 5):1156-1160. DOI: 10.1042/BST0351156
  17. 17. Cragg GM, Newman DJ. Plants as a source of anti-cancer agents. Journal of Ethnopharmacology. 2005;100(1-2):72-79. DOI: 10.1016/j.jep.2005.05.011
  18. 18. Glasauer A, Chandel NS. Targeting antioxidants for cancer therapy. Biochemical Pharmacology. 2014;92(1):90-101. DOI: 10.1016/j.bcp.2014.07.017
  19. 19. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57-70
  20. 20. Floor SL, Dumont JE, Maenhaut C, Raspe E. Hallmarks of cancer: Of all cancer cells, all the time? Trends in Molecular Medicine. 2012;18(9):509-515. DOI: 10.1016/j.molmed.2012.06.005
  21. 21. Minamoto T, Mai M, Ronai Z. Environmental factors as regulators and effectors of multistep carcinogenesis. Carcinogenesis. 1999;20(4):519-527
  22. 22. Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M. Comparative risk assessment collaborating g: Causes of cancer in the world: Comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005;366(9499):1784-1793. DOI: 10.1016/S0140-6736(05)67725-2
  23. 23. Chio IIC, Tuveson DA. ROS in cancer: The burning question. Trends in Molecular Medicine. 2017;23(5):411-429. DOI: 10.1016/j.molmed.2017.03.004
  24. 24. Elmaci I, Altinoz MA. Thymoquinone: An edible redox-active quinone for the pharmacotherapy of neurodegenerative conditions and glial brain tumors. A short review. Biomedicine & Pharmacotherapy. 2016;83:635-640. DOI: 10.1016/j.biopha.2016.07.018
  25. 25. Arora-Kuruganti P, Lucchesi PA, Wurster RD. Proliferation of cultured human astrocytoma cells in response to an oxidant and antioxidant. Journal of Neuro-Oncology. 1999;44(3):213-221
  26. 26. DeLorenze GN, McCoy L, Tsai AL, Quesenberry CP Jr, Rice T, Il'yasova D, Wrensch M. Daily intake of antioxidants in relation to survival among adult patients diagnosed with malignant glioma. BMC Cancer. 2010;10:215. DOI: 10.1186/1471-2407-10-215
  27. 27. Lissoni P, Meregalli S, Nosetto L, Barni S, Tancini G, Fossati V, Maestroni G. Increased survival time in brain glioblastomas by a radioneuroendocrine strategy with radiotherapy plus melatonin compared to radiotherapy alone. Oncology. 1996;53(1):43-46. DOI: 10.1159/000227533
  28. 28. Philipov P, Tzatchev K. Selenium in the treatment of patients with brain gliomas. A pilot study. Zentralblatt für Neurochirurgie. 1990;51(3):145-146
  29. 29. Pozsgai E, Bellyei S, Cseh A, Boronkai A, Racz B, Szabo A, Sumegi B, Hocsak E. Quercetin increases the efficacy of glioblastoma treatment compared to standard chemoradiotherapy by the suppression of PI-3-kinase-Akt pathway. Nutrition and Cancer. 2013;65(7):1059-1066. DOI: 10.1080/01635581.2013.810291
  30. 30. Sabarinathan D, Mahalakshmi P, Vanisree AJ. Naringenin, a flavanone inhibits the proliferation of cerebrally implanted C6 glioma cells in rats. Chemico-Biological Interactions. 2011;189(1-2):26-36. DOI: 10.1016/j.cbi.2010.09.028
  31. 31. Rinaldi M, Caffo M, Minutoli L, Marini H, Abbritti RV, Squadrito F, Trichilo V, Valenti A, Barresi V, Altavilla D, et al. ROS and brain gliomas: An overview of potential and innovative therapeutic strategies. International Journal of Molecular Sciences. 2016;17(6). DOI: 10.3390/ijms17060984
  32. 32. Daneman R, Prat A. The blood-brain barrier. Cold Spring Harbor Perspectives in Biology. 2015;7(1):a020412. DOI: 10.1101/cshperspect.a020412
  33. 33. Haar CP, Hebbar P, Wallace GC, Das A, Vandergrift WA 3rd, Smith JA, Giglio P, Patel SJ, Ray SK, Banik NL. Drug resistance in glioblastoma: A mini review. Neurochemical Research. 2012;37(6):1192-1200. DOI: 10.1007/s11064-011-0701-1
  34. 34. Perazzoli G, Prados J, Ortiz R, Caba O, Cabeza L, Berdasco M, Gonzalez B, Melguizo C. Temozolomide resistance in glioblastoma cell lines: Implication of MGMT, MMR, P-glycoprotein and CD133 expression. PLoS One. 2015;10(10):e0140131. DOI: 10.1371/journal.pone.0140131
  35. 35. Chang KY, Hsu TI, Hsu CC, Tsai SY, Liu JJ, Chou SW, Liu MS, Liou JP, Ko CY, Chen KY, et al. Specificity protein 1-modulated superoxide dismutase 2 enhances temozolomide resistance in glioblastoma, which is independent of O(6)-methylguanine-DNA methyltransferase. Redox Biology. 2017;13:655-664. DOI: 10.1016/j.redox.2017.08.005
  36. 36. Martin V, Herrera F, Garcia-Santos G, Antolin I, Rodriguez-Blanco J, Rodriguez C. Signaling pathways involved in antioxidant control of glioma cell proliferation. Free Radical Biology & Medicine. 2007;42(11):1715-1722. DOI: 10.1016/j.freeradbiomed.2007.02.028
  37. 37. Abdollahi SSM. Antioxidants: Friends or foe in prevention or treatment of cancer: The debate of the century. Toxicology and Applied Pharmacology. 2013;271:49-63
  38. 38. Firuzi O, Miri R, Tavakkoli M, Saso L. Antioxidant therapy: Current status and future prospects. Current Medicinal Chemistry. 2011;18(25):3871-3888
  39. 39. Zhu Z, Kimura M, Itokawa Y, Nakatsu S, Oda Y, Kikuchi H. Effect of selenium on malignant tumor cells of brain. Biological Trace Element Research. 1995;49(1):1-7. DOI: 10.1007/BF02788998
  40. 40. Vartak S, Robbins ME, Spector AA. Polyunsaturated fatty acids increase the sensitivity of 36B10 rat astrocytoma cells to radiation-induced cell kill. Lipids. 1997;32(3):283-292
  41. 41. Vartak S, McCaw R, Davis CS, Robbins ME, Spector AA. Gamma-linolenic acid (GLA) is cytotoxic to 36B10 malignant rat astrocytoma cells but not to 'normal' rat astrocytes. British Journal of Cancer. 1998;77(10):1612-1620
  42. 42. Rooprai HK, Kandanearatchi A, Maidment SL, Christidou M, Trillo-Pazos G, Dexter DT, Rucklidge GJ, Widmer W, Pilkington GJ. Evaluation of the effects of swainsonine, captopril, tangeretin and nobiletin on the biological behaviour of brain tumour cells in vitro. Neuropathology and Applied Neurobiology. 2001;27(1):29-39
  43. 43. Naidu KA, Tang JL, Naidu KA, Prockop LD, Nicosia SV, Coppola D. Antiproliferative and apoptotic effect of ascorbyl stearate in human glioblastoma multiforme cells: Modulation of insulin-like growth factor-I receptor (IGF-IR) expression. Journal of Neuro-Oncology. 2001;54(1):15-22
  44. 44. Rooprai HK, Kyriazis I, Nuttall RK, Edwards DR, Zicha D, Aubyn D, Davies D, Gullan R, Pilkington GJ. Inhibition of invasion and induction of apoptosis by selenium in human malignant brain tumour cells in vitro. International Journal of Oncology. 2007;30(5):1263-1271
  45. 45. Lou M, Zhang LN, Ji PG, Feng FQ, Liu JH, Yang C, Li BF, Wang L. Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/Caspase-3 signaling pathway in human neuroglioma cells: In vitro and in vivo. Biomedicine & Pharmacotherapy. 2016;84:1-9. DOI: 10.1016/j.biopha.2016.08.055
  46. 46. Harmanci D. In vitro effects of selenium on human glioblastoma multiforme cell lines: A preliminary study. Acta Clinica Croatica. 2017;56(1):48-57
  47. 47. Jia F, Ruan S, Liu N, Fu L. Synergistic antitumor effects of berbamine and paclitaxel through ROS/Akt pathway in glioma cells. Evidence-based Complementary and Alternative Medicine. 2017;2017:8152526. DOI: 10.1155/2017/8152526
  48. 48. Castro ML, Carson GM, McConnell MJ, Herst PM. High dose ascorbate causes both genotoxic and metabolic stress in glioma cells. Antioxidants (Basel). 2017;6(3). DOI: 10.3390/antiox6030058
  49. 49. Newell SD Jr, Kapp J, Romfh JH. Evaluation of megadose vitamin therapy in an experimental brain tumor. Surgical Neurology. 1981;16(2):161-164
  50. 50. Wang CJ, Lin JK. Inhibitory effects of carotenoids and retinoids on the in vitro growth of rat C-6 glioma cells. Proceedings of the National Science Council, Republic of China, Part B. 1989;13(3):176-183
  51. 51. Hernan Perez de la Ossa D, Lorente M, Gil-Alegre ME, Torres S, Garcia-Taboada E, Aberturas Mdel R, Molpeceres J, Velasco G, Torres-Suarez AI. Local delivery of cannabinoid-loaded microparticles inhibits tumor growth in a murine xenograft model of glioblastoma multiforme. PLoS One. 2013;8(1):e54795. DOI: 10.1371/journal.pone.0054795
  52. 52. Hervouet E, Staehlin O, Pouliquen D, Debien E, Cartron PF, Menanteau J, Vallette FM, Olivier C. Antioxidants delay clinical signs and systemic effects of ENU induced brain tumors in rats. Nutrition and Cancer. 2013;65(5):686-694. DOI: 10.1080/01635581.2013.789541
  53. 53. Li J, Ni L, Li B, Wang M, Ding Z, Xiong C, Lu X. Coptis Chinensis affects the function of glioma cells through the down-regulation of phosphorylation of STAT3 by reducing HDAC3. BMC Complementary and Alternative Medicine. 2017;17(1):524. DOI: 10.1186/s12906-017-2029-0
  54. 54. Puri T, Goyal S, Julka PK, Nair O, Sharma DN, Rath GK. Lycopene in treatment of high-grade gliomas: A pilot study. Neurology India. 2010;58(1):20-23. DOI: 10.4103/0028-3886.60389
  55. 55. Mulpur BH, Nabors LB, Thompson RC, Olson JJ, LaRocca RV, Thompson Z, Egan KM. Complementary therapy and survival in glioblastoma. Neuro-oncology Practice. 2015;2(3):122-126. DOI: 10.1093/nop/npv008

Written By

Duygu Harmanci

Submitted: 27 November 2017 Reviewed: 09 April 2018 Published: 05 November 2018