Next Article in Journal
Astrocyte and Oligodendrocyte Cross-Talk in the Central Nervous System
Previous Article in Journal
Immunotherapy in Multiple Myeloma
Previous Article in Special Issue
Melatonin Orchestrates Lipid Homeostasis through the Hepatointestinal Circadian Clock and Microbiota during Constant Light Exposure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin’s Antineoplastic Potential Against Glioblastoma

by
Enrico Moretti
1,†,
Gaia Favero
1,†,
Luigi Fabrizio Rodella
1,2 and
Rita Rezzani
1,2,*
1
Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
2
Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2020, 9(3), 599; https://doi.org/10.3390/cells9030599
Submission received: 11 November 2019 / Revised: 25 February 2020 / Accepted: 1 March 2020 / Published: 3 March 2020
(This article belongs to the Special Issue Melatonin in Human Health and Diseases)

Abstract

:
Glioblastoma (GBM) is one of the most intransigent and aggressive brain tumors, and its treatment is extremely challenging and ineffective. To improve patients’ expectancy and quality of life, new therapeutic approaches were investigated. Melatonin is an endogenous indoleamine with an incredible variety of properties. Due to evidence demonstrating melatonin’s activity against several cancer hallmarks, there is growing interest in its use for preventing and treating cancer. In this review, we report on the potential effects of melatonin, alone or in combination with anticancer drugs, against GBM. We also summarize melatonin targets and/or the intracellular pathways involved. Moreover, we describe melatonin’s epigenetic activity responsible for its antineoplastic effects. To date, there are too few clinical studies (involving a small number of patients) investigating the antineoplastic effects of melatonin against GBM. Nevertheless, these studies described improvement of GBM patients’ quality of life and did not show significant adverse effects. In this review, we also report on studies regarding melatonin-like molecules with the tumor-suppressive properties of melatonin together with implemented pharmacokinetics. Melatonin effects and mechanisms of action against GBM require more research attention due to the unquestionably high potential of this multitasking indoleamine in clinical practice.

1. Introduction

1.1. Glioblastoma

Glioblastoma (GBM) is the most aggressive and common primary brain tumor in adults, with an incidence rate of 5–8 per 100,000 habitants, and it represents nearly 54% of all diagnosed gliomas. GBM has poor prognosis with an average life expectancy of less than one year after diagnosis, due to the high rate of tumor recurrences [1,2]. Males show a GBM incidence of 1.6 times higher as compared to females, and they have a significantly higher frequency of primary GBM. However, secondary GBM frequency is higher in females [2,3]. GBM contains a subpopulation of glioma stem-like cells (GSCs) that grow as spheres and induce an increase in the tumor-propagating and self-renewing capacity [4,5,6]. Numerous studies reported that these GSCs play a particularly essential role in maintaining tumor growth and recurrence [7,8,9]. According to these observations, therapeutic approaches that do not eradicate GSCs are more likely to fail. To date, the standard therapy is radiotherapy and adjuvant chemotherapy with temozolomide (TMZ) [10]. TMZ is an oral alkylating agent capable of entering the cerebrospinal fluid and central nervous system, which contributes to improving the overall survival rate of patients with GBM [10,11]. Other drugs were tried, for example, bevacizumab and cilengitide. The effects of bevacizumab, an anti-angiogenetic monoclonal antibody, versus GBM were studied with the hopes of improving the efficacy of chemotherapy. To date, results show an increase in the progression-free survival without an increase in the overall survival [12]. Cilengitide, an integrin inhibitor, is another molecule studied as a therapy against GBM, yet there is no evidence concerning an increase in the overall survival or progression-free survival [13].
Unfortunately, compelling evidence indicates that the treatment of patients with GBM is extremely challenging, as complete surgical resection of the tumor is very difficult, and most GBMs tend to recur after chemotherapy and radiation therapy regimens [14,15]. Hence, some researches focused on novel co-therapeutic strategies, preferably based upon natural resources, to provide anticancer compounds eligible for clinical application. Various studies described the effects of melatonin against GBM, which is not only a recognized antioxidant molecule, but evidence is revealing its anticancer effects. Melatonin is a promising candidate to overcome multidrug resistance in the treatment of GBM [16,17,18]. Additionally, the dipeptide carnosine (β-alanyl-l-histidine) demonstrated antineoplastic effects not only limited to proliferation and cell-cycle control, but also having the ability to decrease the migration of GBM cells [19,20]. Another possible approach to anticancer therapy is to artificially produce new tumor-specific molecules. The formulation of peptides specific for membrane receptors overexpressed or uniquely expressed in GBM cells seems to be a promising therapeutic approach; however, to date, results show poor and unsatisfactory outcomes [21].
These routes provide valuable and distinctive strategies for improving GBM treatment, one of the most untreatable and deadly malignant diseases. Further studies are required for obtaining a coherent picture and detailed knowledge of tumor membrane receptors and the underlying intracellular signaling pathways involved in GBM formation and progression. The standard of care should evolve to include patients’ maximum safety during resection with concurrent and adjuvant chemotherapy. Future studies are required that focus on the develop of new therapeutic molecules and strategies, various types of combination therapies, and dose-finding trials.

1.2. Melatonin

Melatonin, an endogenous indoleamine with numerous properties and functions, is synthesized from l-tryptophan by the pineal gland and potentially in any tissue [22,23]. Melatonin possesses lipophilic and hydrophilic properties, which permit its receptor-independent transfer into almost all tissues and fluids. Melatonin, through both receptor-dependent and receptor-independent signaling pathways, activates a broad spectrum of molecular pathways. For example, melatonin acts as a natural chronobiotic substance with immune-enhancing properties, as well as having the ability to reduce oxidative stress [24,25,26]. It is well established that melatonin possesses anti-inflammatory and antioxidant activities, as well as having an influence on metabolism, the sleep–wake cycle, and reproduction [27,28,29,30]. The antioxidant activity of melatonin is observed through the prevention of free radical formation, as well as the capacity to upregulate antioxidant enzyme activity and downregulate pro-oxidant enzymes [31,32,33]. Furthermore, in cancer cells, melatonin presents a paradoxical pro-oxidant effect, leading to cell death [34,35,36]. Therefore, melatonin may inhibit cancer development by increasing the induction of oxidative stress in tumor cells [37]. As concluded by several studies, melatonin seems to recognize a cell’s context (normal or tumoral) and modulates the appropriate actions, thus behaving akin to a “smart killer” [34,38]. This aspect of melatonin is fundamental in oncological therapies; however, an understanding of how such a paradoxical behavior occurs is needed from future studies.
Melatonin has three different receptors that belong to the family of G protein-coupled transmembrane receptors: human melatonin receptor 1 (MT1), 2 (MT2), and 3 (MT3) [39,40,41]. A study by Dillon et al. [42] indicated that breast cancer cells have higher MT1 expression than normal cells, thus supporting a different susceptibility of cancer cells to melatonin. Despite the existence of melatonin receptors, many of the melatonin actions are mainly mediated through receptor-independent activities, such as antioxidant, anti-inflammatory, and mitochondrial activity [43,44,45,46]. There may also be melatonin binding sites in the nucleus of some cells that serve to synergistically influence the activity of antioxidant enzymes [47].
In recent years, an increasing number of studies investigated the abilities of melatonin to modulate several cancer cell processes, including proliferation, differentiation, invasion, and apoptosis [48,49,50]. Melatonin’s anticancer activity was described in several cancer types, such as ovarian, lung, breast, colon, and brain cancers [51,52,53,54,55]. Notably, melatonin may be able to reduce the proliferation of GBM-inducing cells (GICs), stem-like cells isolated from GBM specimens, as well as their self-renewal and clonogenic capability, modulating different biological processes and signaling [16,18]. In addition, no significant side effects were described with the use of melatonin in animal and human studies, even in extremely high doses [56,57]. Andersen et al. [57] reported only mild adverse effects, such as dizziness, sleepiness, nausea, and headaches.
In this review, we analyze the current evidence of melatonin effects against GBM and GSCs, both in vitro and in vivo, as a promising multitasking indoleamine for improving the efficacy of GBM therapy. It is worth mentioning that the benefits of melatonin for cancer recovery in clinical trials are debatable, noting that this multitasking indoleamine is mainly used as an adjuvant molecule to reduce chemotherapy or radiotherapy side effects and to improve patients’ sleep [58,59]. Nevertheless, these advantageous effects of melatonin as an adjuvant therapeutic are important and should not be underestimated during chemotherapy-related illness because such effects may substantially improve patient’s quality of life.

2. In Vitro Evidence of Melatonin Effects against Glioblastoma

As previously reported, GSCs are a cell subpopulation responsible for GBM tumor development, progression, relapse, and therapeutic resistance. Therefore, the eradication of GSCs and GICs is essential to achieve a long-lasting tumor remission. Martín et al. [60] evaluated the effects of melatonin alone in cells obtained from acute cell dissociation of human GBM postsurgical specimens. The authors found that melatonin treatment significantly reduced tumor cell proliferation and induced a decrease in self-renewal and clonogenic ability accompanied by a reduction in the expression of stem-cell markers, such as the transmembrane glycoprotein cluster of differentiation 133 (CD133). Interestingly, in the same study, similar effects did not appear during melatonin treatment of normal human neural cells, confirming, as previously reported, that cytotoxic effects of this indoleamine are specific for cancer cells [60]. Moreover, GBM cells treated with melatonin showed cell death with ultrastructural features of autophagy and a progressive accumulation of autophagosome vacuoles that led to the cellular membrane and organelle disruption.
Mitochondria are the main source of reactive oxygen species (ROS) production and are a known target of melatonin [61,62]. As previously reported, melatonin presented a pro-oxidant effect in cancer cells [34,35,36]. A high concentration of ROS can damage mitochondrial and nuclear DNA and modify the expression of oncogenes and tumor suppressor genes [63,64]. In the hopes of blocking tumor growth, Franco et al. [35] studied the effects of 3 mM melatonin on human GBM cells and observed a reduced expression of mitochondrial transcription factor A (TFAM), which is an essential protein that maintains mitochondrial DNA (mtDNA) integrity [65,66]. Moreover, melatonin selectively induced a surprisingly concentration-dependent increase in ROS production and delayed cell-cycle progression in GBM cancer cells as compared to the control group [35], while the presence of high levels of mitochondria-synthesized melatonin appears to be toxic for GBM and GSCs [67]. This effect of melatonin on ROS production may be due to the altered expression of respiratory chain genes, which could depend on the activity of the transcription factors TFAM, TFB1M, and TFB2M [35].
The concentration of melatonin required to inhibit cell growth in several tumor types is higher compared to its endogenous concentration in the bloodstream. In fact, pharmacological concentrations of this indoleamine are necessary to reduce neoplastic activity [68]. As described by Hevia et al. [69], the intracellular concentration of melatonin depend on the cell type; if the intracellular concentration of melatonin is high, its anticancer properties are improved. Together with the pharmacological concentration, the incubation time is crucial for melatonin therapeutic effects. It was observed that an incubation time less than 72 h did not induce effects in cell viability or intracellular ROS content [35]. Very few studies analyzed in detail if melatonin effects were receptor-mediated or -independent. Thus, more studies focused on these aspects could help to clarify melatonin’s mechanism of action against GBM. Interestingly, a study evaluating melatonin’s antineoplastic mechanism of action, carried out by Martín et al. [70], used both pertussis toxin (200 ng/mL), an inhibitor of G-coupled receptors, and luzindole (10 Amol/L), an MT1 and MT2 antagonist, and they observed that melatonin was effective in reducing glioma cell proliferation, confirming that melatonin may act through different molecular pathways. Specifically, the melatonin membrane receptor dissociation constant (Kd) is in the nanomolar range. As a result, such low concentrations of melatonin do not appear to have any significant effect on rat glioma cells. Therefore, Martín et al. [70] evaluated the possible mediation of melatonin membrane receptors in its antiproliferative effect using high doses (1 mM) of melatonin, observing that melatonin binding to its membrane receptors did not mediate inhibition of GBM cell proliferation. The direct and indirect antioxidant activities of melatonin also play a significant role in contrasting GBM cells. Rat glioma cells show basal activation of nuclear factor κB (NF-κB) and activator protein-1 (AP-1), both transcription factors regulated by cellular oxidative stress. The treatment with 1 mmol/L melatonin reduced NF-κB activation, while no effects on the basal activation of AP-1 were described [70].
In the tumor microenvironment, infiltrating immune cells were shown to interact with tumor progression [71], and GBM shows an abundant population of infiltrating monocytes/macrophages that constitute up to 40% of a tumor’s mass and provide protection from immune surveillance [72]. The reduction of monocyte-related tumor-promotion effects in GBM could be a potentially useful antineoplastic strategy. With this in mind, Lai et al. [73] reported that in vitro melatonin supplementation reduced monocyte adhesion molecules, as well as monocyte chemoattractant chemokine CCL2 expression in GBM, consequently attenuating monocyte recruitment and activity. Importantly, Lai et al. [73] also observed that administration of melatonin increased the expression of sirtuin 1, which, in turn, reduced the expression of monocyte adhesion molecules.
Notably, one of the key features of melatonin in anticancer therapy is its epigenetic activity [74,75]. Melatonin can alter the fate of a neoplastic cell by downregulating oncogenes and upregulating oncosuppressive genes and interacting with intracellular signaling pathways, for example, selectively inducing cancer cell apoptosis [76,77,78]. As previously described in other cancerous cells, mitochondrial membrane depolarization increased in cells incubated with melatonin, thus indicating the release of cytochrome c and other pro-apoptotic factors, activating the cell death pathway [61,79,80,81]. Melatonin can influence transcription factors, such as the c-Jun N-terminal kinase (JNK) pathway, and it can activate caspase-3, inducing cell death [82]. Melatonin treatment is also involved in modifying the expression of another important transcription factor, nuclear factor erythroid 2-related factor (Nrf2), which interacts with the expression of several antioxidant and anti-inflammatory genes [76,83,84].
The downregulation of enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) may in part explain the inhibitory effect of melatonin on GSCs. Zheng et al. [85], studying the knockdown of EZH2 in GBM cells, observed a significant downregulation of Notch1, a transmembrane protein with an important role in cell embryonic and postnatal development [85,86,87,88] that is overexpressed in many cancer types, like breast, lung, pancreatic, and colon cancer [88]. The depletion of EZH2 also reduced the quantity and self-renewal activity of GSCs. The treatment of GBM cells with melatonin induced remarkable growth inhibition in GBM, showing the role of melatonin in the inhibition of the EZH2–Notch1 signaling axis and its potential therapeutic significance [85].
It is known that the proprieties of cancerous cells are affected by surrounding environmental conditions. In particular, hypoxia is a well-characterized component of GBM microenvironment and a crucial factor in tumor aggressiveness and resistance to therapy [89,90]. In the study by Zhang et al. [91], hypoxic GBM cells showed an invasiveness increased more than two-fold compared with the respective controls under normoxia. Melatonin was observed to have anti-migratory and anti-invasive action in GBM not only under normoxia, but also under hypoxia [92]. Melatonin influenced hypoxia-inducible factor-1α (HIF-1α), a major oncogenic factor playing an essential role in tumor invasion and angiogenesis under hypoxia [93], as well as the activity of the enzymes responsible for the degradation of HIF-1α [91].

Melatonin and Chemotherapy

One of the main problems in GBM pharmacologic therapy is chemoresistance. The main goals for improving patients’ survival and quality of life concerning co-therapy are to improve the cytotoxic effects of chemotherapy, while reducing chemoresistance and chemotherapeutic side effects. Clinical and experimental studies showed that melatonin enhanced the antineoplastic activity of chemotherapy in various cancer types. This, along with significantly reducing chemotherapy side effects, improves the patient’s quality of life [36,53,59]. McConnell et al. [94] studied the effects of the association of melatonin to TMZ in cultured GBM cells and GSCs. They also studied TMZ association with other antioxidants, alpha-lipoic acid and vitamin D3, which are widely available via an oral route, and which can readily cross the blood–brain barrier. Notably, each molecule decreased the proliferation of GBM cells. Vitamin D3 showed the least dramatic effect [95], and higher doses could be necessary for more significant results; additionally, blood levels of 375 nmol/L vitamin D3 resulted in hypervitaminosis [96]. Interestingly, melatonin showed a robust anti-proliferative activity both alone and combined with TMZ. Surprisingly, alpha-lipoic acid showed the most pertinent results with significant anti-proliferative activity both alone and in combination with TMZ. Nevertheless, even if several studies described a delay in cancer cell proliferation and apoptosis under melatonin-treatment, there is no evidence of a clear cancer regression with a net reduction in the GBM neoplastic mass using melatonin alone in clinical trials. Notably, melatonin was shown to reduce the side effects related to of chemo- and radiotherapy and enhance their therapeutic efficacy in advanced cancer patients with poor clinical status, improving the clinical outcome of patients afflicted by cancer. Therefore, this indoleamine potentially presents both curative and palliative actions in the treatment of human neoplasms [97]. Further studies are needed in order to better understand melatonin’s mechanism of action against GBM and to determine which drugs melatonin should or should not be used with as an adjuvant therapy. Furthermore, future studies are needed to evaluate melatonin as a sole antineoplastic therapy, and not just as a supplement to more invasive therapies.
Multidrug resistance is the main defense mechanism developed by GBM against chemotherapy, leading to treatment failure. Aberrant expression and activity of the adenosine triphosphate-binding cassette (ABC) family transporters can induce multidrug resistance in cancer cells [98]. Interestingly, beneficial anti-GBM effects were reported for the combination treatment of melatonin and various drugs, including chemotherapeutic molecules [99]. Melatonin treatment of GBM cell lines and GSCs from surgery specimens increased methylation levels of the ABCG2 promoter, which appears to decrease the transporter expression, causing a synergism between melatonin and chemotherapeutic molecules [100]. The evidence of 5-azacitidine, a DNA methyl-transferase inhibitor, preventing melatonin effects on ABCG2/breast cancer resistance protein (BCRP) expression suggests an epigenetic effect of the indoleamine through the alteration of DNA methyltransferase activity [100].
GBM cells present an elevated expression of Nrf2, contributing to chemoresistance [101,102,103,104,105]. A study by Pan et al. [106] described an increased expression of the Nrf2–antioxidant responsive element (ARE) signaling pathway in TMZ-resistant GBM cells. Treatment with melatonin restored the chemosensitivity of TMZ-resistant GBM cells and increased the rate of cancer cell apoptosis [106]. Pharmacological concentrations of melatonin (1 mM) induced a significant reduction of Nrf2 expression in GBM cells when combined with TMZ [106]. However, a previous study suggested that at low concentration of melatonin may activate the Nrf2–ARE signaling pathway in glioma cells [107]. Further studies are required to confirm the activity of melatonin on the Nrf2–ARE pathway, including in vivo experiments and preclinical studies.
Furthermore, the expression of transcription factor EB (TFEB), a relevant molecule for chemoresistance that transcriptionally regulates several lysosomal genes, is significantly increased in GBM cells [108,109,110]. A recent study from Sung et al. [111] observed whether the combination of vorinostat, a histone deacetylases inhibitor, and melatonin could overcome the effects of TFEB and induce GBM and GSC apoptosis [111].

3. In Vivo and Clinical Trial Evidence of Melatonin Effects against Glioblastoma

Despite several in vitro findings of the antineoplastic effect of melatonin against GBM, to date, only a few articles described the effects of melatonin against GBM in animal models, with even fewer describing the effects in humans.
Martín et al. [70] described the efficacy of melatonin treatment in reducing tumor growth in vivo. In particular, glioma cells were pre-treated with melatonin and then injected into rats. Interestingly, at day 11 (six days after the start of the treatment), tumor growth was significantly reduced as compared to the control untreated group. Additionally, at day 14, the reduction in tumor growth reached 50% [70]. It must be mentioned that the concentration used in this study was greater than the concentration used in any human study (15 mg/kg body weight by subcutaneous injection in rats versus 20 mg per day administered orally in clinical trials). Therefore, for future clinical trials, new melatonin dosing schedules equivalent to that used successfully in in vivo studies will hopefully demonstrate the apparent effects of melatonin alone as a antineoplastic agent against GBM.
Cancer cell stemness is dynamically influenced by epigenetic mechanisms [112,113,114]. To determine the efficacy of five days of in vitro treatment with melatonin compared with TMZ (5 mmol/L) on GSCs, Chen et al. [16] performed an intracranial injection of GSCs into immunocompromised mice. The control GSCs showed no benefit of TMZ administration, and death usually resulted within 35–45 days. By contrast, the pre-treatment of GSCs with melatonin significantly raised the survival rate. These results suggest that short in vitro melatonin treatment may be able to reduce the number of tumor-initiating GSCs, allowing a delayed tumor development. Notably, Chen et al. [16] determined, for the first time, that the effects of melatonin on GSCs occur mainly through melatonin receptor-dependent signaling. In fact, the expression of GSC stem-cell markers was significantly impaired when GSCs were incubated with luzindole (10 mmol/L), prior to the addition of melatonin. [16]. More specifically, the inhibitory effect of melatonin on GSC growth resulted from alterations in the activity of EZH2, rather than its expression levels. EZH2, which is activated by protein kinase B (Akt) phosphorylation, catalyzes the methylation of histone H3 and induces signal transducer and activator of transcription 3 (STAT3) [16,115,116,117], which plays a critical role in the cell cycle and other pathways, such as tyrosine phosphorylation, serine phosphorylation, and DNA binding [16,118,119,120,121]. Notably, in GSCs isolated from patients, STAT3 showed specific interactions with EZH2, and Chen et al. [16] also observed that melatonin treatment significantly inhibited STAT3 activity. Therefore, by targeting the Akt–EZH2–STAT3 axis, melatonin can strongly impair GSC self-renewal and tumor-initiating capacity. Furthermore, Chen et al. [16] also observed that melatonin treatment in Akt-overexpressing GSCs inhibited Akt activation. Additionally, compared to normal human brain specimens, surgical GBM specimens showed a remarkable upregulation of EZH2 and Notch1 and, as previously reported, Zheng et al. [85] observed that Notch1 was significantly downregulated in knockdown EZH2 GBM cells, thus underlining the role of melatonin in the inhibition of the EZH2–Notch1 signaling axis. Figure 1 summarizes the melatonin mechanism of action on Akt–EZH2–STAT3 and EZH2–Notch1 signaling pathways.
Notably, the only clinical trial, carried out by Lissoni et al. [58], evaluated the effects of melatonin co-treatment in 30 GBM patients undergoing radical or adjuvant radiotherapy (RT). Patients were randomly divided in groups of RT alone or RT plus 20 mg/day oral melatonin. At one year from the start of the treatment, the patient survival percentage of the RT + melatonin group was significantly higher than the RT alone group (6/14 vs. 1/16 patients). Notably, as previously reported, this clinical trial also described in the RT + melatonin group a decrease in RT and steroid therapy adverse effects, such as infections and alopecia, together with reduced anxiety and improved quality of sleep. Even though future studies with a larger number of patients should be investigated, these results underline the potential utility of melatonin as co-treatment in GBM patients, as well as its capacity to improve patients’ quality of life. Another study from Lissoni et al. [122], tested the combination of melatonin and Aloe vera, which has anti-inflammatory effects [123,124,125]. The rationale of the study was to investigate the potential improvement of melatonin antineoplastic effects with the combination of Aloe vera. Thus, 50 neoplastic patients with GBM, breast cancer, lung cancer, or gastrointestinal tract tumor, unresponsive to chemotherapy, RT, or endocrine therapy or poor clinical condition precluding chemotherapy, were enlisted. After at least one month following the last chemotherapy session, patients received an oral dose of 20 mg/day of melatonin or the same dose of melatonin plus Aloe vera tincture until neoplastic regression. Unfortunately, in the group of melatonin alone, no tumor regression was observed at two months after the start of the treatment, whereas, in the melatonin plus Aloe vera group, 8% (two patients out of 24) showed a partial response. However, there were no melatonin-associated adverse events, whereas treatment with Aloe vera was associated with diarrhea, even if limited to just the first day of administration [122]. One concern, however, is that in vitro studies typically used a melatonin concentration (1 mM) that may not a be physiologically attainable. As McConnell et al. [94] reported, the concentration of 50 nM closely mimics physiological levels (54 nM) for patients taking 20 mg of melatonin orally, whereas the concentration of 1 mM melatonin may not be physiologically attainable. The use of melatonin as an adjuvant to chemotherapy showed promising results, both improving the efficacy of treatment and reducing side effects [17,94]. Clinical trials studying melatonin-integrated therapies in oncological patients, with tumors other than GBM, are generally conducted outside clinical guidelines after failing to respond to the gold-standard therapy and with an already compromised prognosis. This is certainly a limit in the study of a potential efficacy of melatonin treatment in increasing the healing rate or the tumor regression in such patients. Even with the evidence of antineoplastic effects in vitro and palliative effects in humans, the integration of this multitasking indoleamine in the first-line therapy of a malignancy would face logical ethical issues. However, improving quality of sleep and reducing the side effects from oncological therapies is already a very important result in critically ill patients that should not be undervalued [58,59,69].
Clinical treatment with melatonin faces some drawbacks, such as irregular absorption and a short circulating half-life [126,127]. In order to avoid these obstacles, melatonergic agonists were developed and used for sleep dysregulation and depression [128,129]. In comparison to melatonin, ramelteon and agomelatine, both melatonin receptor agonists, have significant advantages through higher receptor affinity, higher absorption concentration, and longer half-life. Agomelatine, licensed by the European Medicines Agency, is a melatonin agonist used in the treatment of depression [130]. It shows a higher affinity to melatonergic receptors, with longer half-life, and it better penetrates the blood–brain barrier [129,131,132]. Ramelteon, approved by the Food and Drug Administration of the United States (FDA), is an MT1 and MT2 agonist free of side effects and available for altered sleep initiation [133,134,135]. The affinity of this molecule to MT1 and MT2 is remarkably higher with respect to melatonin and shows improved oral absorption and circulating half-life [136]. These emerging findings suggest that melatonergic agonists present potentially essential roles as an approach against GBM. Recently, Zhou et al. [137] reported that agomelatine, also in addition to melatonin, may act as a common upstream signal between autophagy and apoptosis of GBM cells, with the effect attenuated by treatment with luzindole. These results may lead to the development of new therapeutic strategies for glioma.
Furthermore, using the ionic gelation method, chitosan/tripolyphosphate nanoparticles (CSNPs) were prepared with a size range from 115 to 740 nm in order to overcome the pharmacokinetic limitations of melatonin and improve its efficacy against GBM. CSNPs were tested in the human GBM cell line U87MG. The use of small-size particles as a carrier system led to higher cellular uptake and blood vessel penetration. GBM cells showed higher melatonin CSNP uptake compared to non-malignant cells [138]. The uptake of these nanoparticles depends on the cell surface charge and the metabolic activity of cancerous cells [139]. Previous studies showed that blank CSNPs increased ROS concentration in cancerous cells, leading to mitochondrial cytochrome c release and apoptosis through the caspase cascade [140]. Recently, de Oliveira Junior et al. [141] described, for the first time, the application of nanoparticle-mediated nose-to-brain delivery of melatonin for the treatment of GBM both in vitro and in vivo. Pharmacokinetic data revealed an increased melatonin concentration in the brain of rats administered intranasally with melatonin nanoparticles compared to melatonin suspension, in both nasally and orally administered groups. Furthermore, the nanoencapsulation of melatonin was crucial to promoting selective and strong cytotoxic effects on the U87MG human GBM cell line.
GBM cells were incubated with equivalent doses of melatonin and melatonin-loaded chitosan. Loading of blank nanoparticles without melatonin was evaluated as control. Both melatonin and melatonin-loaded chitosan reduced GBM cell viability with a marked time- and dose-dependent pattern [138]. Interestingly, the study described higher anticancer activity in the melatonin CSNP group than in the “pure” melatonin cells, leaving only 22% against 42% of viable cells after 24 h of incubation. With longer exposure (48 h and 72 h), the “pure” melatonin group did not show further reduction in the percentage of cell viability. This evidence suggests that melatonin CSNPs extend the melatonin anticancer activity with respect to “pure” melatonin. Differences in cellular metabolism and surface charge may explain the distinctive response to melatonin CSNPs between GBM cells and control cells [138].

4. Conclusions

Melatonin is a promising agent in the field of antineoplastic research, and this multitasking indoleamine shows an incredible variety of properties against GBM (Figure 2). As described in this review, melatonin specifically interacts with cancer cells and interferes with GBM proliferative activity and aggressiveness. In vitro studies investigating the effects of GBM cells through therapy of melatonin in association with the standard chemotherapy TMZ showed relevant and notable antineoplastic effects together with contrasting TMZ chemoresistance. The association of melatonin with chemotherapy may enhance the cytotoxic effects against cancer and help to decrease its dosage, thereby reducing side effects and improving quality of life. Further studies observed also interesting epigenetic activity of melatonin against GBM. In addition, local drug delivery techniques or the improvement of the pharmacokinetics through melatonin-like molecules could achieve effective clinical outcomes. Despite the remarkable in vitro evidence of melatonin efficacy against GBM, only a few in vivo articles and even fewer clinical trials analyzed this specific topic. A scientific evidence consortium with in vivo and especially preclinical studies could be the first step toward the potential application of melatonin against GBM, as, at least, a combined therapy with TMZ.

Author Contributions

L.F.R. and R.R. conceptualized the review; E.M. and G.F. critically analyzed the bibliography; E.M. and G.F. wrote the manuscript draft and drew the figures; L.F.R. and R.R. improved the manuscript content; E.M. and G.F. contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

The relative research studies of our research group were supported by FLAMMA S.p.A., Chignolo d’Isola (BG), Italy (http://www.flammagroup.com) as well as by ex 60% grants of the University of Brescia, Brescia (Italy).

Acknowledgments

The authors sincerely thank FLAMMA S.p.A., Chignolo d’Isola (BG, Italy) for the precious research support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Crocetti, E.; Trama, A.; Stiller, C.; Caldarella, A.; Soffietti, R.; Jaal, J.; Weber, D.C.; Ricardi, U.; Slowinski, J.; Brandes, A. RARECARE working group. Epidemiology of glial and non-glial brain tumours in Europe. Eur. J. Cancer 2012, 48, 1532–1542. [Google Scholar] [CrossRef] [PubMed]
  2. Ostrom, Q.T.; Gittleman, H.; Liao, P.; Vecchione-Koval, T.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010–2014. Neuro. Oncol. 2017, 19, v1–v88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wrensch, M.; Minn, Y.; Chew, T.; Bondy, M.; Berger, M.S. Epidemiology of primary brain tumors: Current concepts and review of the literature. Neuro Oncol. 2002, 4, 278–299. [Google Scholar] [CrossRef] [PubMed]
  4. Dirks, P.B. Brain tumor stem cells: The cancer stem cell hypothesis writ large. Mol. Oncol. 2010, 4, 420–430. [Google Scholar] [CrossRef] [Green Version]
  5. Liebelt, B.D.; Shingu, T.; Zhou, X.; Ren, J.; Shin, S.A.; Hu, J. Glioma stem cells: Signaling, microenvironment, and therapy. Stem Cells Int. 2016, 2016, 1–10. [Google Scholar] [CrossRef] [Green Version]
  6. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef] [Green Version]
  7. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
  8. Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [Green Version]
  9. Gilbert, C.A.; Ross, A.H. Cancer stem cells: Cell culture, markers, and targets for new therapies. J. Cell Biochem. 2009, 108, 1031–1038. [Google Scholar] [CrossRef] [Green Version]
  10. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  11. Friedman, H.S.; Kerby, T.; Calvert, H. Temozolomide and treatment of malignant glioma. Clin. Cancer Res. 2000, 6, 2585–2597. [Google Scholar] [PubMed]
  12. Haines, I.E.; Gabor Miklos, G.L. Bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 2048. [Google Scholar] [CrossRef] [PubMed]
  13. Stupp, R.; Hegi, M.E.; Gloria, T.; Erridge, S.C.; Perry, J.; Hong, Y.K.; Aldape, K.D.; Lhermitte, B.; Pietsch, T.; Grujicic, D. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma and methylated O6-methylguanine-DNA methyltransferase (MGMT) gene promoter: Key results of the multicenter, randomized, open-label, controlled, phase III CENTRIC study. J. Clin. Oncol. 2013, 15, 1100–1108. [Google Scholar] [CrossRef] [Green Version]
  14. Carson, K.A.; Grossman, S.A.; Fisher, J.D.; Shaw, E.G. Prognostic factors for survival in adult patients with recurrent glioma enrolled onto the new approaches to brain tumor therapy CNS consortium phase I and II clinical trials. J. Clin. Oncol. 2007, 25, 2601–2606. [Google Scholar] [CrossRef] [PubMed]
  15. Qazi, M.A.; Vora, P.; Venugopal, C.; Sidhu, S.S.; Moffat, J.; Swanton, C.; Singh, S.K. Intratumoral heterogeneity: Pathways to treatment resistance and relapse in human glioblastoma. Ann. Oncol. 2017, 28, 1448–1456. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, X.; Hao, A.; Li, X.; Du, Z.; Li, H.; Wang, H.; Yang, H.; Fang, Z. Melatonin inhibits tumorigenicity of glioblastoma stem-like cells via the AKT-EZH2-STAT3 signaling axis. J. Pineal Res. 2016, 61, 208–217. [Google Scholar] [CrossRef] [PubMed]
  17. Maitra, S.; Bhattacharya, D.; Das, S.; Bhattacharya, S. Melatonin and its anti-glioma functions: A comprehensive review. Rev. Neurosci. 2019, 30, 527–541. [Google Scholar] [CrossRef]
  18. Neamati, F.; Asemi, Z. The effects of melatonin on signaling pathways and molecules involved in glioma. Fundam. Clin. Pharmacol. 2019. [Google Scholar] [CrossRef]
  19. Oppermann, H.; Faust, H.; Yamanishi, U.; Meixensberger, J.; Gaunitz, F. Carnosine inhibits glioblastoma growth independent from PI3K/Akt/mTOR signaling. PLoS ONE 2019, 14, e0218972. [Google Scholar] [CrossRef] [Green Version]
  20. Oppermann, H.; Purcz, K.; Birkemeyer, C.; Baran-Schmidt, R.; Meixensberger, J.; Gaunitz, F. Carnosine’s inhibitory effect on glioblastoma cell growth is independent of its cleavage. Amino Acids 2019, 51, 761–772. [Google Scholar] [CrossRef]
  21. Raucher, D. Tumor targeting peptides: Novel therapeutic strategies in glioblastoma. Curr. Opin. Pharmacol. 2019, 47, 14–19. [Google Scholar] [CrossRef] [PubMed]
  22. Acuña Castroviejo, D.; Escames, G.; Venegas, C.; Díaz Casado, M.E.; Lima Cabello, E.; López, L.C.; Rosales Corral, S.; Tan, D.X.; Reiter, R.J. Extrapineal melatonin: Sources, regulation, and potential functions. Cell Mol. Life Sci. 2014, 71, 2997–3025. [Google Scholar] [CrossRef] [PubMed]
  23. Bubenik, G.A.; Konturek, S.J. Melatonin and aging: Prospects for human treatment. J. Physiol. Pharmacol. 2011, 62, 13–19. [Google Scholar] [PubMed]
  24. Carrillo-Vico, A.; Lardone, P.J.; Alvarez-Sanchez, N.; Rodríguez-Rodríguez, A.; Guerrero, J.M. Melatonin: Buffering the immune system. Int. J. Mol. Sci. 2013, 14, 8638–8683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Galano, A.; Reiter, R.J. Melatonin and its metabolites versus oxidative stress: From individual actions to collective protection. J. Pineal Res. 2018, 65, e12514. [Google Scholar] [CrossRef] [Green Version]
  26. Lewy, A.J. Melatonin and human chronobiology. Cold Spring Harb. Sym. 2007, 72, 623–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kleszczyński, K.; Bilska, B.; Stegemann, A.; Flis, D.J.; Ziolkowski, W.; Pyza, E.; Luger, T.A.; Reiter, R.J.; Böhm, M.; Slominski, A.T. Melatonin and its metabolites ameliorate UVR-induced mitochondrial oxidative stress in human MNT-1 melanoma cells. Int. J. Mol. Sci. 2018, 19, 1–19. [Google Scholar] [CrossRef] [Green Version]
  28. Moretti, R.; Zanin, A.; Pansiot, J.; Spiri, D.; Manganozzi, L.; Kratzer, I.; Favero, G.; Vasiljevic, A.; Rinaldi, V.E.; Pic, I.; et al. Melatonin reduces excitotoxic blood-brain barrier breakdown in neonatal rats. Neuroscience 2015, 311, 382–397. [Google Scholar] [CrossRef]
  29. Ostadmohammadi, V.; Soleimani, A.; Bahmani, F.; Aghadavod, E.; Ramezani, R.; Reiter, R.J.; Mansournia, M.A.; Banikazemi, Z.; Soleimani, M.; Zaroudi, M.; et al. The effects of melatonin supplementation on parameters of mental health, glycemic control, markers of cardiometabolic risk, and oxidative stress in diabetic hemodialysis patients: A randomized, double-blind, placebo-controlled trial. J. Ren. Nutr. 2019. [Google Scholar] [CrossRef]
  30. Sánchez-López, A.L.; Ortiz, G.G.; Pacheco-Moises, F.P.; Mireles-Ramírez, M.A.; Bitzer-Quintero, O.K.; Delgado-Lara, D.L.C.; Ramírez-Jirano, L.J.; Velázquez-Brizuela, I.E. Efficacy of melatonin on serum pro-inflammatory cytokines and oxidative stress markers in relapsing remitting multiple sclerosis. Arch. Med. Res. 2018, 49, 391–398. [Google Scholar] [CrossRef]
  31. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Reiter, R.J. Clinical aspects of melatonin in the acute coronary syndrome. Curr. Vasc. Pharmacol. 2009, 7, 367–373. [Google Scholar] [CrossRef] [PubMed]
  32. Halladin, N.L.; Busch, S.E.; Jensen, S.E.; Hansen, H.S.; Zaremba, T.; Aarøe, J.; Rosenberg, J.; Gögenur, I. Intracoronary and systemic melatonin to patients with acute myocardial infarction: Protocol for the IMPACT trial. Dan. Med. J. 2014, 61, A4773. [Google Scholar] [PubMed]
  33. Reiter, R.J.; Mayo, J.C.; Tan, D.X.; Sainz, R.M.; Alatorre-Jimenez, M.; Qin, L. Melatonin as an antioxidant: Under promises but over delivers. J. Pineal Res. 2016, 61, 253–278. [Google Scholar] [CrossRef]
  34. Bizzarri, M.; Proietti, S.; Cucina, A.; Reiter, R.J. Molecular mechanisms of the pro-apoptotic actions of melatonin in cancer: A review. Expert Opin. Ther. Targets 2013, 17, 1483–1496. [Google Scholar] [CrossRef] [PubMed]
  35. Franco, D.G.; Moretti, I.F.; Marie, S.K.N. Mitochondria transcription factor A: A putative target for the effect of melatonin on U87MG malignant glioma cell line. Molecules 2018, 23, 1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Pourhanifeh, M.H.; Sharifi, M.; Reiter, R.J.; Davoodabadi, A.; Asemi, Z. Melatonin and non-small cell lung cancer: New insights into signaling pathways. Cancer Cell. Int. 2019, 19, 131. [Google Scholar] [CrossRef]
  37. Fan, C.; Pan, Y.; Yang, Y.; Di, S.; Jiang, S.; Ma, Z.; Li, T.; Zhang, Z.; Li, W.; Li, X.; et al. HDAC1 inhibition by melatonin leads to suppression of lung adenocarcinoma cells via induction of oxidative stress and activation of apoptotic pathways. J. Pineal Res. 2015, 59, 321–333. [Google Scholar] [CrossRef]
  38. Lanoix, D.; Lacasse, A.A.; Reiter, R.J.; Vaillancourt, C. Melatonin: The smart killer: The human trophoblast as a model. Mol. Cell Endocrinol. 2012, 348, 1–11. [Google Scholar] [CrossRef]
  39. Dubocovich, M.L.; Rivera-Bermudez, M.A.; Gerdin, M.J.; Masana, M.I. Molecular pharmacology, regulation and function of mammalian melatonin receptors. Front. Biosci. 2003, 8, 1093–1108. [Google Scholar] [CrossRef] [Green Version]
  40. Slominski, R.M.; Reiter, R.J.; Schlabritz-Loutsevitch, N.; Ostrom, R.S.; Slominski, A.T. Melatonin membrane receptors in peripheral tissues: Distribution and functions. Mol. Cell Endocrinol. 2012, 351, 152–166. [Google Scholar] [CrossRef] [Green Version]
  41. Witt-Enderby, P.A.; Bennett, J.; Jarzynka, M.J.; Firestine, S.; Melan, M.A. Melatonin receptors and their regulation: Biochemical and structural mechanisms. Life Sci. 2003, 72, 2183–2198. [Google Scholar] [CrossRef]
  42. Dillon, D.C.; Easley, S.E.; Asch, B.B.; Cheney, R.T.; Brydon, L.; Jockers, R.; Winston, J.S.; Brooks, J.S.; Hurd, T.; Asch, H.L. Differential expression of high-affinity melatonin receptors (MT1) in normal and malignant human breast tissue. Am. J. Clin. Pathol. 2002, 118, 451–458. [Google Scholar] [CrossRef] [PubMed]
  43. Cutando, A.; Aneiros-Fernández, J.; López-Valverde, A.; Arias-Santiago, S.; Aneiros-Cachaza, J.; Reiter, R.J. A new perspective in oral health: Potential importance and actions of melatonin receptors MT1, MT2, MT3, and RZR/ROR in the oral cavity. Arch. Oral Biol. 2011, 56, 944–950. [Google Scholar] [CrossRef] [PubMed]
  44. Ghosh, A.K.; Naaz, S.; Bhattacharjee, B.; Ghosal, N.; Chattopadhyay, A.; Roy, S.; Reiter, R.J.; Bandyopadhyay, D. Mechanism of melatonin protection against copper-ascorbate-induced oxidative damage in vitro through isothermal titration calorimetry. Life Sci. 2017, 180, 123–136. [Google Scholar] [CrossRef] [PubMed]
  45. Rusanova, I.; Martínez-Ruiz, L.; Florido, J.; Rodríguez-Santana, C.; Guerra-Librero, A.; Acuña-Castroviejo, D.; Escames, G. Protective effects of melatonin on the skin: Future perspectives. Int. J. Mol. Sci. 2019, 20, 4948. [Google Scholar] [CrossRef] [Green Version]
  46. Tan, D.X.; Reiter, R.J.; Manchester, L.C.; Yan, M.T.; El-Sawi, M.; Sainz, R.M.; Mayo, J.C.; Kohen, R.; Allegra, M.; Hardeland, R. Chemical and physical properties and potential mechanisms: Melatonin as a broad spectrum antioxidant and free radical scavenger. Curr. Top Med. Chem. 2002, 2, 181–197. [Google Scholar] [CrossRef] [Green Version]
  47. Tomás-Zapico, C.; Coto-Montes, A. A proposed mechanism to explain the stimulatory effect of melatonin on antioxidative enzymes. J. Pineal Res. 2005, 39, 99–104. [Google Scholar] [CrossRef]
  48. Liu, L.; Xu, Y.; Reiter, R.J.; Pan, Y.; Chen, D.; Liu, Y.; Pu, X.; Jiang, L.; Li, Z. Inhibition of ERK1/2 signaling pathway is involved in melatonin’s antiproliferative effect on human MG-63 osteosarcoma cells. Cell Physiol Biochem. 2016, 39, 2297–2307. [Google Scholar] [CrossRef]
  49. Loureiro, R.; Magalhães-Novais, S.; Mesquita, K.A.; Baldeiras, I.; Sousa, I.S.; Tavares, L.C.; Barbosa, I.A.; Oliveira, P.J.; Vega-Naredo, I. Melatonin antiproliferative effects require active mitochondrial function in embryonal carcinoma cells. Oncotarget 2015, 6, 17081–17096. [Google Scholar] [CrossRef] [Green Version]
  50. Shen, C.J.; Chang, C.C.; Chen, Y.T.; Lai, C.S.; Hsu, Y.C. Melatonin suppresses the growth of ovarian cancer cell lines (OVCAR-429 and PA-1) and potentiates the effect of G1 arrest by targeting CDKs. Int. J. Mol. Sci. 2016, 17, 176. [Google Scholar] [CrossRef] [Green Version]
  51. Bejarano, I.; Redondo, P.C.; Espino, J.; Rosado, J.A.; Paredes, S.D.; Barriga, C.; Reiter, R.J.; Pariente, J.A.; Rodríguez, A.B. Melatonin induces mitochondrial-mediated apoptosis in human myeloid HL-60 cells. J. Pineal Res. 2009, 46, 392–400. [Google Scholar] [CrossRef] [PubMed]
  52. Chovancova, B.; Hudecova, S.; Lencesova, L.; Babula, P.; Rezuchova, I.; Penesova, A.; Grman, M.; Moravcik, R.; Zeman, M.; Krizanova, O. Melatonin-induced changes in cytosolic calcium might be responsible for apoptosis induction in tumour cells. Cell Physiol. Biochem. 2017, 44, 763–777. [Google Scholar] [CrossRef] [PubMed]
  53. Favero, G.; Moretti, E.; Bonomini, F.; Reiter, R.J.; Rodella, L.F.; Rezzani, R. Promising antineoplastic actions of melatonin. Front. Pharmacol. 2018, 2017, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gatti, G.; Lucini, V.; Dugnani, S.; Calastretti, A.; Spadoni, G.; Bedini, A.; Rivara, S.; Mor, M.; Canti, G.; Scaglione, F.; et al. Antiproliferative and pro-apoptotic activity of melatonin analogues on melanoma and breast cancer cells. Oncotarget 2017, 8, 68338–68353. [Google Scholar] [CrossRef] [Green Version]
  55. Nooshinfar, E.; Bashash, D.; Safaroghli-Azar, A.; Bayati, S.; Rezaei-Tavirani, M.; Ghaffari, S.H.; Akbari, M.E. Melatonin promotes ATO-induced apoptosis in MCF-7 cells: Proposing novel therapeutic potential for breast cancer. Biomed. Pharmacother. 2016, 83, 83–456. [Google Scholar] [CrossRef]
  56. Altun, A.; Ugur-Altun, B. Melatonin: Therapeutic and clinical utilization. Int. J. Clin. Pract. 2007, 61, 835–845. [Google Scholar] [CrossRef]
  57. Andersen, L.P.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The safety of melatonin in humans. Clin. Drug Investig. 2016, 36, 169–175. [Google Scholar] [CrossRef]
  58. 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, 43–46. [Google Scholar] [CrossRef]
  59. Sanchez-Barcelo, E.J.; Mediavilla, M.D.; Alonso-Gonzalez, C.; Reiter, R.J. Melatonin uses in oncology: Breast cancer prevention and reduction of the side effects of chemotherapy and radiation. Expert Opin. Investig. Drugs 2012, 21, 819–931. [Google Scholar] [CrossRef]
  60. Martín, V.; Sanchez-Sanchez, A.M.; Puente-Moncada, N.; Gomez-Lobo, M.; Alvarez-Vega, M.A.; Antolín, I.; Rodriguez, C. Involvement of autophagy in melatonin-induced cytotoxicity in glioma-initiating cells. J. Pineal Res. 2014, 57, 308–316. [Google Scholar] [CrossRef]
  61. Acuña Castroviejo, D.; Escames, G.; Carazo, A.; León, J.; Khaldy, H.; Reiter, R.J. Melatonin, mitochondrial homeostasis and mitochondrial-related diseases. Curr. Top Med. Chem. 2002, 2, 133–151. [Google Scholar] [CrossRef] [PubMed]
  62. Reiter, R.J.; Rosales-Corral, S.; Tan, D.X.; Jou, M.J.; Galano, A.; Xu, B. Melatonin as a mitochondria-targeted antioxidant: One of evolution’s best ideas. Cell Mol. Life Sci. 2017, 7, 3863–3881. [Google Scholar] [CrossRef] [PubMed]
  63. Luchetti, F.; Canonico, B.; Betti, M.; Arcangeletti, M.; Pilolli, F.; Piroddi, M.; Canesi, L.; Papa, S.; Galli, F. Melatonin signaling and cell protection function. FASEB J. 2010, 24, 3603–3624. [Google Scholar] [CrossRef] [PubMed]
  64. Thannickal, V.J.; Fanburg, B.L. Reactive oxygen species in cell signaling. Am. J. Physiol. Lung Cell Mol. Physiol. 2000, 279, L1005–L1028. [Google Scholar] [CrossRef] [Green Version]
  65. Alam, T.I.; Kanki, T.; Muta, T.; Ukaji, K.; Abe, Y.; Nakayama, H.; Takio, K.; Hamasaki, N.; Kang, D. Human mitochondrial DNA is packaged with TFAM. Nucleic Acids Res. 2003, 31, 1640–1645. [Google Scholar] [CrossRef]
  66. Campbell, C.T.; Kolesar, J.E.; Kaufman, B.A. Mitochondrial transcription factor A regulates mitochondrial transcription initiation, DNA packaging, and genome copy number. Biochim. Biophys. Acta 2012, 1819, 921–929. [Google Scholar] [CrossRef]
  67. Anderson, G.; Reiter, R.J. Glioblastoma: Role of mitochondria n-acetylserotonin/melatonin ratio in mediating effects of mir-451 and aryl hydrocarbon receptor and in coordinating wider biochemical changes. Int. J. Tryptophan. Res. 2019, 12, 1–9. [Google Scholar] [CrossRef] [Green Version]
  68. Sainz, R.M.; Mayo, J.C.; Tan, D.X.; León, J.; Manchester, L.; Reiter, R.J. Melatonin reduces prostate cancer cell growth leading to neuroendocrine differentiation via a receptor and PKA independent mechanism. Prostate 2005, 63, 29–43. [Google Scholar] [CrossRef]
  69. Hevia, D.; Mayo, J.C.; Quiros, I.; Gomez-Cordoves, C.; Sainz, R.M. Monitoring intracellular melatonin levels in human prostate normal and cancer cells by HPLC. Anal. Bioanal. Chem. 2010, 397, 1235–1244. [Google Scholar] [CrossRef]
  70. Martín, V.; Herrera, F.; Carrera-Gonzalez, P.; García-Santos, G.; Antolín, I.; Rodriguez-Blanco, J.; Rodriguez, C. Intracellular signaling pathways involved in the cell growth inhibition of glioma cells by melatonin. Cancer Res. 2006, 66, 1081–1088. [Google Scholar] [CrossRef] [Green Version]
  71. Gil-Bernabé, A.M.; Ferjancic, S.; Tlalka, M.; Zhao, L.; Allen, P.D.; Im, J.H.; Watson, K.; Hill, S.A.; Amirkhosravi, A.; Francis, J.L.; et al. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice. Blood 2012, 119, 3164–3175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Charles, N.A.; Holland, E.C.; Gilbertson, R.; Glass, R.; Kettenmann, H. The brain tumor microenvironment. Glia 2012, 60, 502–514. [Google Scholar] [CrossRef] [PubMed]
  73. Lai, S.W.; Liu, Y.S.; Lu, D.Y.; Tsai, C.F. Melatonin modulates the microenvironment of glioblastoma multiforme by targeting sirtuin 1. Nutrients 2019, 11, 1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Haim, A.; Zubidat, A.E. Artificial light at night: Melatonin as a mediator between the environment and epigenome. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 1–7. [Google Scholar] [CrossRef] [Green Version]
  75. Korkmaz, A.; Sanchez-Barcelo, E.J.; Tan, D.X.; Reiter, R.J. Role of melatonin in the epigenetic regulation of breast cancer. Breast Cancer Res. Treat. 2009, 115, 13–27. [Google Scholar] [CrossRef]
  76. Bondy, S.C. and Campbell, A. Mechanisms underlying tumor suppressive properties of melatonin. Int. J. Mol. Sci. 2018, 19, 2205. [Google Scholar] [CrossRef] [Green Version]
  77. Gao, Y.; Xiao, X.; Zhang, C.; Yu, W.; Guo, W.; Zhang, Z.; Li, Z.; Feng, X.; Hao, J.; Zhang, K.; et al. Melatonin synergizes the chemotherapeutic effect of 5-fluorouracil in colon cancer by suppressing PI3K/AKT and NF-B/iNOS signaling pathways. J. Pineal Res. 2017, 62, 1–15. [Google Scholar] [CrossRef]
  78. Lee, S.E.; Kim, S.J.; Yoon, H.J.; Yu, S.Y.; Yang, H.; Jeong, S.I.; Hwang, S.Y.; Park, C.S.; Park, Y.S. Genome-wide profiling in melatonin-exposed human breast cancer cell lines identifies differentially methylated genes involved in the anticancer effect of melatonin. J. Pineal Res. 2013, 54, 80–88. [Google Scholar] [CrossRef]
  79. Halestrap, A.P.; McStay, G.P.; Clarke, S.J. The permeability transition pore complex: Another view. Biochimie 2002, 84, 153–166. [Google Scholar] [CrossRef]
  80. Kosar, P.A.; Razirogler, M.; Ovey, I.S.; Ciq, B. Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells: Involvement of TRPV1 channels. J. Membr. Biol. 2016, 249, 129–214. [Google Scholar] [CrossRef]
  81. León, J.; Acuña-Castroviejo, D.; Escames, G.; Tan, D.X.; Reiter, R.J. Melatonin mitigates mitochondrial malfunction. J. Pineal Res. 2005, 38, 1–9. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, S.; Hoffmann, K.; Gao, C.; Petrulionis, M.; Herr, I.; Schemmer, P. Melatonin promotes sorafenib-induced apoptosis through synergistic activation of JNK/c-jun pathway in human hepatocellular carcinoma. J. Pineal Res. 2017, 62, 1–9. [Google Scholar] [CrossRef] [PubMed]
  83. Santofimia-Castaño, P.; Clea Ruy, D.; Garcia-Sanchez, L.; Jimenez-Blasco, D.; Fernandez-Bermejo, M.; Bolaños, J.P.; Salido, G.M.; Gonzalez, A. Melatonin induces the expression of Nrf2-regulated antioxidant enzymes via PKC and Ca2+ influx activation in mouse pancreatic acinar cells. Free Radic. Biol. Med. 2015, 87, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Trivedi, P.P.; Jena, G.B.; Tikoo, K.B.; Kumar, V. Melatonin modulated autophagy and Nrf2 signaling pathways in mice with colitis-associated colon carcinogenesis. Mol. Carcinog. 2016, 55, 255–267. [Google Scholar] [CrossRef] [PubMed]
  85. Zheng, X.; Pang, B.; Gu, G.; Gao, T.; Zhang, R.; Pang, Q.; Liu, Q. Melatonin Inhibits Glioblastoma Stem-like cells through Suppression of EZH2-NOTCH1 Signaling Axis. Int. J. Biol. Sci. 2017, 13, 245–253. [Google Scholar] [CrossRef] [Green Version]
  86. Artavanis-Tsakonas, S.; Rand, M.D.; Lake, R.J. Notch signaling: Cell fate control and signal integration in development. Science 1999, 284, 770–776. [Google Scholar] [CrossRef] [Green Version]
  87. Nickoloff, B.J.; Osborne, B.A.; Miele, L. Notch signaling as a therapeutic target in cancer: A new approach to the development of cell fate modifying agents. Oncogene 2003, 22, 6598–6608. [Google Scholar] [CrossRef] [Green Version]
  88. Shao, S.; Zhao, X.; Zhang, X.; Luo, M.; Zuo, X.; Huang, S.; Wang, Y.; Gu, S.; Zhao, X. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol. Cancer 2015, 14, 28. [Google Scholar] [CrossRef] [Green Version]
  89. Bar, E.E.; Lin, A.; Mahairaki, V.; Matsui, W.; Eberhart, C.G. Hypoxia increases the expression of stem-cell markers and promotes clonogenicity in glioblastoma neurospheres. Am. J. Pathol. 2010, 177, 1491–1502. [Google Scholar] [CrossRef]
  90. Lee, O.H.; Xe, J.; Fueyo, J.; Alonso, M.M.; Liu, D.; Martin, V.; Jiang, H.; Piao, Y.; Liu, T.J.; Gomez-Manzano, C. Angiopoietin-2 decreases vascular endothelial growth factor expression by modulating HIF-1 alpha levels in gliomas. Oncogene 2008, 27, 1310–1314. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, Y.; Liu, Q.; Wang, F.; Ling, E.A.; Liu, S.; Wang, L.; Yang, Y.; Yao, L.; Chen, X.; Wang, F.; et al. Melatonin antagonizes hypoxia-mediated glioblastoma cell migration and invasion via inhibition of HIF-1α. J. Pineal Res. 2013, 55, 121–130. [Google Scholar] [CrossRef] [PubMed]
  92. Mauriz, J.L.; Collado, P.S.; Veneroso, C.; Reiter, R.J.; González-Gallego, J. A review of the molecular aspects of melatonin’s anti-inflammatory actions: Recent insights and new perspectives. J. Pineal Res. 2013, 54, 1–14. [Google Scholar] [CrossRef] [PubMed]
  93. Semenza, G.L. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol. Sci. 2012, 33, 207–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. McConnell, D.D.; McGreevy, J.W.; Williams, M.N.; Litofsky, N.S. Do anti-oxidants vitamin D(3,) melatonin, and alpha-lipoic acid have synergistic effects with temozolomide on cultured glioblastoma cells? Medicines 2018, 5, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Van Ginkel, P.R.; Yang, W.; Marcet, M.M.; Chow, C.C.; Kulkarni, A.D.; Darjatmoko, S.; Lindstrom, M.J.; Lokken, J.; Bhattacharya, S.; Albert, D.M. 1 alpha-hydroxyvitamin D2 inhibits growth of human neuroblastoma. J. Neurooncol. 2007, 85, 255–262. [Google Scholar] [CrossRef] [PubMed]
  96. Goodwin, P.J. Vitamin D in cancer patients: Above all, do no harm. J. Clin. Oncol. 2009, 27, 2117–2119. [Google Scholar] [CrossRef] [Green Version]
  97. Lissoni, P. Is there a role for melatonin in supportive care? Support Care Cancer 2002, 10, 110–116. [Google Scholar] [CrossRef]
  98. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP-dependent transporters. Nat. Rev. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef] [Green Version]
  99. Reiter, R.J.; Tan, D.X.; Sainz, R.M.; Mayo, J.C.; Lopez-Burillo, S. Melatonin: Reducing the toxicity and increasing the efficacy of drugs. J. Pharm. Pharmacol. 2002, 54, 1299–1321. [Google Scholar] [CrossRef] [Green Version]
  100. Martín, V.; Sanchez-Sanchez, A.M.; Herrera, F.; Gomez-Manzano, C.; Fueyo, J.; Alvarez-Vega, M.A.; Antolín, I.; Rodriguez, C. Melatonin-induced methylation of the ABCG2/BCRP promoter as a novel mechanism to overcome multidrug resistance in brain tumour stem cells. Br. J. Cancer 2013, 108, 2005–2012. [Google Scholar] [CrossRef] [Green Version]
  101. Ji, X.J.; Chen, S.H.; Zhu, L.; Pan, H.; Zhou, Y.; Li, W.; You, W.C.; Gao, C.C.; Zhu, J.H.; Jiang, K.; et al. Knockdown of NF-E2-related factor 2 inhibits the proliferation and growth of U251MG human glioma cells in a mouse xenograft model. Oncol. Rep. 2013, 30, 157–164. [Google Scholar] [CrossRef] [PubMed]
  102. Ji, X.; Wang, H.; Zhu, J.; Zhu, L.; Pan, H.; Li, W.; Zhou, Y.; Cong, Z.; Yan, F.; Chen, S. Knockdown of Nrf2 suppresses glioblastoma angiogenesis by inhibiting hypoxia-induced activation of HIF-1α. Int. J. Cancer 2014, 135, 574–584. [Google Scholar] [CrossRef] [PubMed]
  103. Pan, H.; Wang, H.; Zhu, L.; Mao, L.; Qiao, L.; Su, X. The role of Nrf2 in migration and invasion of human glioma cell U251. World Neurosurg. 2013, 80, 363–370. [Google Scholar] [CrossRef] [PubMed]
  104. Pan, H.; Wang, H.; Zhu, L.; Wang, X.; Cong, Z.; Sun, K.; Fan, Y. The involvement of Nrf2-ARE pathway in regulation of apoptosis in human glioblastoma cell U251. Neurol. Res. 2013, 35, 71–78. [Google Scholar] [CrossRef]
  105. Zhou, Y.; Wang, H.D.; Zhu, L.; Cong, Z.X.; Li, N.; Ji, X.J.; Pan, H.; Wang, J.W.; Li, W.C. Knockdown of Nrf2 enhances autophagy induced by temozolomide in U251 human glioma cell line. Oncol. Rep. 2013, 29, 394–400. [Google Scholar] [CrossRef]
  106. Pan, H.; Wang, H.; Jia, Y.; Wang, Q.; Li, L.; Wu, Q.; Chen, L. VPA and MEL induce apoptosis by inhibiting the Nrf2-ARE signaling pathway in TMZ-resistant U251 cells. Mol. Med. Rep. 2017, 16, 908–914. [Google Scholar] [CrossRef] [Green Version]
  107. Jumnongprakhon, P.; Govitrapong, P.; Tocharus, C.; Pinkaew, D.; Tocharus, J. Melatonin protects methamphetamine-induced neuroinflammation through NF-κB and Nrf2 pathways in glioma cell line. Neurochem. Res. 2015, 40, 1448–1456. [Google Scholar] [CrossRef]
  108. Giatromanolaki, A.; Sivridis, E.; Mitrakas, A.; Kalamida, D.; Zois, C.E.; Haider, S.; Piperidou, C.; Pappa, A.; Gatter, K.C.; Harris, A.L.; et al. Autophagy and lysosomal related protein expression patterns in human glioblastoma. Cancer Biol. Ther. 2014, 15, 1468–1478. [Google Scholar] [CrossRef] [Green Version]
  109. Martina, J.A.; Chen, Y.; Gucek, M.; Puertollano, R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 2012, 8, 903–914. [Google Scholar] [CrossRef] [Green Version]
  110. Song, W.; Wang, F.; Savini, M.; Ake, A.; di Ronza, A.; Sardiello, M.; Segatori, L. TFEB regulates lysosomal proteostasis. Hum. Mol. Genet. 2013, 22, 1994–2009. [Google Scholar] [CrossRef] [Green Version]
  111. Sung, G.J.; Kim, S.H.; Kwak, S.; Park, S.H.; Song, J.H.; Jung, J.H.; Kim, H.; Choi, K.C. Inhibition of TFEB oligomerization by co-treatment of melatonin with vorinostat promotes the therapeutic sensitivity in glioblastoma and glioma stem cells. J. Pineal Res. 2019, 66, e12556. [Google Scholar] [CrossRef] [PubMed]
  112. Avgustinova, A.; Benitah, S.A. The epigenetics of tumour initiation: Cancer stem cells and their chromatin. Curr. Opin. Genet. Dev. 2016, 36, 8–15. [Google Scholar] [CrossRef] [PubMed]
  113. Hammoud, S.S.; Cairns, B.R.; Jones, D.A. Epigenetic regulation of colon cancer and intestinal stem cells. Curr. Opin. Cell Biol. 2013, 25, 177–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wang, Y.; Cardenas, H.; Fang, F.; Condello, S.; Taverna, P.; Segar, M.; Liu, Y.; Nephew, K.P.; Matei, D. Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 2014, 74, 4922–4936. [Google Scholar] [CrossRef] [Green Version]
  115. Cao, R.; Wang, L.; Wang, H.; Erdjument-Bromage, H.; Tempst, P.; Jones, R.S.; Zhang, Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 2002, 298, 1039–1043. [Google Scholar] [CrossRef] [Green Version]
  116. Kondo, Y. Targeting histone methyltransferase EZH2 as cancer treatment. J. Biochem. 2014, 156, 249–257. [Google Scholar] [CrossRef]
  117. Yoo, K.H.; Hennighausen, L. EZH2 methyltransferase and H3K27 methylation in breast cancer. Int. J. Biol. Sci. 2012, 8, 59–65. [Google Scholar] [CrossRef] [Green Version]
  118. Deb, G.; Thakur, V.S.; Gupta, S. Multifaceted role of EZH2 in breast and prostate tumorigenesis: Epigenetics and beyond. Epigenetics 2013, 8, 464–476. [Google Scholar] [CrossRef] [Green Version]
  119. Xu, K.; Wu, Z.J.; Groner, A.C.; He, H.H.; Cai, C.; Lis, R.T.; Wu, X.; Stack, E.C.; Loda, M.; Liu, T. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science 2012, 338, 1465–1469. [Google Scholar] [CrossRef] [Green Version]
  120. Yang, J.; Stark, G.R. Roles of unphosphorylated STATs in signaling. Cell Res. 2008, 18, 443–451. [Google Scholar] [CrossRef]
  121. Zhong, Z.; Wen, Z.; Darnell, J.E., Jr. Stat3: A STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 1994, 264, 95–98. [Google Scholar] [CrossRef] [PubMed]
  122. Lissoni, P.; Giani, L.; Zerbini, S.; Trabattoni, P.; Rovelli, F. Biotherapy with the pineal immunomodulating hormone melatonin versus melatonin plus aloe vera in untreatable advanced solid neoplasms. Nat. Immun. 1998, 16, 27–33. [Google Scholar] [CrossRef] [PubMed]
  123. Davis, R.H.; Parker, W.L.; Sampson, R.T.; Murdoch, D.P. Isolation of a stimulatory system in an aloe extract. J. Am. Pediatr. Med. Assoc. 1991, 81, 473–478. [Google Scholar] [CrossRef] [PubMed]
  124. Hart, L.A.; Van Enckevort, P.H.; Van Dijk, H.; Zaat, R.; De Silva, K.T. Two functionally and chemically distinct immunomodulatory compounds in the gel of Aloe Vera. J. Ethnopharmacol. 1988, 23, 61–71. [Google Scholar] [CrossRef]
  125. Soeda, M. Extract of Cape aloes inhibited sarcoma 180 and Ehrlich ascites tumours. J. Med. Soc. Jpn. 1969, 16, 365–369. [Google Scholar]
  126. DeMuro, R.L.; Nafziger, A.N.; Blask, D.E.; Menhinick, A.M.; Bertino, J.S., Jr. The absolute bioavailability of oral melatonin. J. Clin. Pharmacol. 2000, 40, 781–7844. [Google Scholar] [CrossRef]
  127. Kast, R.E. Agomelatine or ramelteon as treatment adjuncts in glioblastoma and other M1- or M2-expressing cancers. Contemp. Oncol. 2015, 19, 157–162. [Google Scholar] [CrossRef]
  128. Hardeland, R. New approaches in the management of insomnia: Weighing the advantages of prolonged release melatonin and synthetic melatoninergic agonists. Neuropsychiatr. Dis. Treat. 2009, 5, 341–354. [Google Scholar] [CrossRef] [Green Version]
  129. Hardeland, R.; Poeggeler, B.; Srinivasan, V.; Trakht, I.; Pandi-Perumal, S.R.; Cardinali, D.P. Melatonergic drugs in clinical practice. Arzneimittelforschung 2008, 58, 1–10. [Google Scholar] [CrossRef]
  130. Owen, R.T. Agomelatine: A novel pharmacological approach to treating depression. Drugs Today 2009, 45, 599–608. [Google Scholar] [CrossRef]
  131. Cardinali, D.P.; Srinivasan, V.; Brzezinski, A.; Brown, G.M. Melatonin and its analogs in insomnia and depression. J. Pineal Res. 2012, 52, 365–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Pompili, M.; Serafini, G.; Innamorati, M.; Venturini, P.; Fusar-Poli, P.; Sher, L.; Amore, M.; Girardi, P. Agomelatine, a novel intriguing antidepressant option enhancing neuroplasticity: A critical review. World J. Biol. Psychiatry 2013, 14, 412–431. [Google Scholar] [CrossRef] [PubMed]
  133. McGechan, A.; Wellington, K. Ramelteon. CNS Drugs 2005, 19, 1057–1065. [Google Scholar] [CrossRef] [PubMed]
  134. Pandi-Perumal, S.R.; Srinivasan, V.; Spence, D.W.; Moscovitch, A.; Hardeland, R.; Brown, G.M.; Cardinali, D.P. Ramelteon: A review of its therapeutic potential in sleep disorders. Adv. Ther. 2009, 26, 613–626. [Google Scholar] [CrossRef] [Green Version]
  135. Simpson, D.; Curran, M.P. Ramelteon: A review of its use in insomnia. Drugs 2008, 68, 1901–1919. [Google Scholar] [CrossRef]
  136. Greenblatt, D.J.; Harmatz, J.S.; Karim, A. Age and gender effects on the pharmacokinetics and pharmacodynamics of ramelteon, a hypnotic agent acting via melatonin receptors MT1 and MT2. J. Clin. Pharmacol. 2007, 47, 485–496. [Google Scholar] [CrossRef]
  137. Zhou, N.; Wei, Z.X.; Qi, Z.X. Inhibition of autophagy triggers melatonin-induced apoptosis in glioblastoma cells. BMC Neurosci. 2019, 20, 63. [Google Scholar] [CrossRef] [Green Version]
  138. Kumar Yadav, S.; Kumar Srivastava, A.; Dev, A.; Kaundal, B.; Roy Choudhury, S.; Karmakar, S. Nanomelatonin triggers superior anticancer functionality in a human malignant glioblastoma cell line. Nanotechnology 2017, 28, 365102. [Google Scholar] [CrossRef]
  139. Costa, E.C.; Gaspar, V.M.; Marques, J.G.; Coutinho, P.; Correia, I.J. Evaluation of nanoparticle uptake in coculture cancer models. PLoS ONE 2013, 8, e70072. [Google Scholar] [CrossRef]
  140. Zhao, Y.; Zhang, S.; Wang, P.; Fu, S.; Wu, D.; Liu, A. Seleno-short-chain chitosan induces apoptosis in human non-small-cell lung cancer A549 cells through ROS-mediated mitochondrial pathway. Cytotechnology 2017, 69, 851–863. [Google Scholar] [CrossRef]
  141. De Oliveira Junior, E.R.; Nascimento, T.L.; Salomão, M.A.; Da Silva, A.C.G.; Valadares, M.C.; Lima, E.M. Increased nose-to-brain delivery of melatonin mediated by polycaprolactone nanoparticles for the treatment of glioblastoma. Pharm. Res. 2019, 1, 131. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The interaction of melatonin with its membrane receptors induces alterations in the activity of protein kinase B (Akt)–enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2)–signal transducer and activator of transcription 3 (STAT3) and EZH2–Notch1 pathways in Akt-overexpressing glioma stem-like cells (GSCs). Melatonin prevents EZH2 activation and reduces EZH2 pro-oncotic effects such as the upregulation of Notch1. (A) Scheme summarizing the basal activity of Akt–EZH2–STAT3 and EZH2–Notch1 pathways. (B) Scheme summarizing the effects induced by melatonin on Akt–EZH2–STAT3 and EZH2–Notch1 pathways. Mel: melatonin; MT: melatonin receptor; P: phosphorylation.
Figure 1. The interaction of melatonin with its membrane receptors induces alterations in the activity of protein kinase B (Akt)–enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2)–signal transducer and activator of transcription 3 (STAT3) and EZH2–Notch1 pathways in Akt-overexpressing glioma stem-like cells (GSCs). Melatonin prevents EZH2 activation and reduces EZH2 pro-oncotic effects such as the upregulation of Notch1. (A) Scheme summarizing the basal activity of Akt–EZH2–STAT3 and EZH2–Notch1 pathways. (B) Scheme summarizing the effects induced by melatonin on Akt–EZH2–STAT3 and EZH2–Notch1 pathways. Mel: melatonin; MT: melatonin receptor; P: phosphorylation.
Cells 09 00599 g001
Figure 2. Melatonin may induce, in glioblastoma (GBM) cells, an intracellular transcription cascade, which, in turn, leads to an antineoplastic response reducing/blocking oncogene expression, chemoresistance, and other cellular mechanisms. Mel: melatonin; MT: melatonin receptor; mtDNA: mitochondrial DNA; TMZ: temozolomide.
Figure 2. Melatonin may induce, in glioblastoma (GBM) cells, an intracellular transcription cascade, which, in turn, leads to an antineoplastic response reducing/blocking oncogene expression, chemoresistance, and other cellular mechanisms. Mel: melatonin; MT: melatonin receptor; mtDNA: mitochondrial DNA; TMZ: temozolomide.
Cells 09 00599 g002

Share and Cite

MDPI and ACS Style

Moretti, E.; Favero, G.; Rodella, L.F.; Rezzani, R. Melatonin’s Antineoplastic Potential Against Glioblastoma. Cells 2020, 9, 599. https://doi.org/10.3390/cells9030599

AMA Style

Moretti E, Favero G, Rodella LF, Rezzani R. Melatonin’s Antineoplastic Potential Against Glioblastoma. Cells. 2020; 9(3):599. https://doi.org/10.3390/cells9030599

Chicago/Turabian Style

Moretti, Enrico, Gaia Favero, Luigi Fabrizio Rodella, and Rita Rezzani. 2020. "Melatonin’s Antineoplastic Potential Against Glioblastoma" Cells 9, no. 3: 599. https://doi.org/10.3390/cells9030599

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop