Next Article in Journal
AI-Based Glioma Grading for a Trustworthy Diagnosis: An Analytical Pipeline for Improved Reliability
Next Article in Special Issue
Diffuse Gliomas with FGFR3-TACC3 Fusions: Oncogenic Mechanisms, Hallmarks, and Therapeutic Perspectives
Previous Article in Journal
Liver Fibrosis in Non-Alcoholic Fatty Liver Disease and Progression to Hepatocellular Carcinoma in Patients with Inflammatory Bowel Disease: A Systematic Review
Previous Article in Special Issue
Medical Device Advances in the Treatment of Glioblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Could Metformin and Resveratrol Support Glioblastoma Treatment? A Mechanistic View at the Cellular Level

by
Raghad Sabaawi Ibrahim
1,†,
Shahad Sabaawi Ibrahim
1,†,
Ahmed El-Naas
1,
Lenka Koklesová
2,
Peter Kubatka
3 and
Dietrich Büsselberg
1,*
1
Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
2
Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
3
Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(13), 3368; https://doi.org/10.3390/cancers15133368
Submission received: 12 April 2023 / Revised: 14 June 2023 / Accepted: 15 June 2023 / Published: 27 June 2023
(This article belongs to the Special Issue Hot Topics in Neuro-Oncology)

Abstract

:

Simple Summary

Glioblastoma’s poor prognosis calls for the discovery of newer, more efficacious management and treatment methods. This review collates and examines how the antidiabetic drug metformin and nonflavonoid polyphenol resveratrol, a dietary supplement with antidiabetic effects, can complement current treatment methods. Specifically, metformin and resveratrol exert anticancer effects on major metabolic pathways in glioblastoma cells, resulting in reduced proliferation, increased apoptosis, and reduced tumor growth and volume. The shown effects suggest that metformin and resveratrol can potentially aid in treating glioblastoma. The novel delivery methods and a lack of clinical studies endorse further clinical investigations.

Abstract

Glioblastoma, a malignant brain tumor, is a common primary brain tumor in adults, with diabetes mellitus being a crucial risk factor. This review examines how the antidiabetic drug metformin and dietary supplement resveratrol can benefit the treatment of glioblastoma. Metformin and resveratrol have demonstrated action against relevant pathways in cancer cells. Metformin and resveratrol inhibit cell proliferation by downregulating the PI3K/Akt pathway, activating mTOR, and increasing AMPK phosphorylation, resulting in lower proliferation and higher apoptosis levels. Metformin and resveratrol both upregulate and inhibit different cascades in the MAPK pathway. In vivo, the drugs reduced tumor growth and volume. These actions show how metformin and resveratrol can combat cancer with both glucose-dependent and glucose-independent effects. The pre-clinical results, alongside the lack of clinical studies and the rise in novel delivery mechanisms, warrant further clinical investigations into the applications of metformin and resveratrol as both separate and as a combination complement to current glioblastoma therapies.

1. Metformin and Resveratrol in Glioblastoma

Glioblastoma (GBM) is a malignant brain tumor with a poor prognosis and is a common primary brain tumor in adults, accounting for approximately 49% of malignant brain/CNS tumors [1,2]. Current glioblastoma management involves surgery followed by weeks of radiotherapy and concomitant daily temozolomide [3]. While this approach once doubled the two-year survival rate for glioblastoma patients, glioblastoma’s prognosis remains very poor; its low survival rate, with only 5.5% of patients surviving five years post-diagnosis [4], creates a necessity for not only more research on current efforts in the management of the disease but for a widening of scope involving the search for efficient and efficacious alternatives or complements. Steps in this direction include the investigation of metformin (MET) and resveratrol (RES)—due to their displays of anticancer activities—and their effects on major metabolic, proliferative, and apoptotic pathways involved in glioblastoma.
Phytochemicals, bioactive plant-derivatives, exert many health-promoting effects, including antioxidant, genoprotective, antineoplastic, anti-inflammatory, and antiangiogenic efficacy in various cancer types [5,6]. They also have a role in carcinogenesis by modulating miRNA expression, which regulates tumors by acting as tumor suppressors or oncogenes [7]. Metformin (1,1-dimethylbiguanide) is a synthetic derivative of galegine and/or guanidine originating from the plant Galega officinalis or French lilac [8]. It is an FDA-approved antidiabetic drug—the first-line treatment of type 2 diabetes mellitus—and is widely studied. It shows potential against various cancers and also exhibits antineoplastic effects in brain tumors specifically [9]. Resveratrol (3, 5, 4′-trihydroxystilbene) is a nonflavonoid polyphenol that is found in grapes, peanuts, and other plant sources. It has various therapeutic physiological effects on the human body, ranging from cardioprotective and antidiabetic effects to antioxidant potential and antitumor effects [10].
Hyperglycemia, a defining symptom of diabetes mellitus, is a risk factor for some cancers, including gliomas [11]. This review investigates how metformin and resveratrol, two drugs with both antidiabetic and anticancer effects, can benefit the treatment of glioblastoma, and whether such benefits would be primarily dependent on their effects on glucose metabolism or if their anticancer effects arise from affecting other pathways. Both metformin and resveratrol have promising anticancer effects because they decrease the development of some cancer cell lines in in vitro studies through pathways, including the PI3K-Akt, AMPK-mTOR, and MAPK cascades [12,13,14,15]. Reducing proliferation and inducing apoptosis through these pathways cause a decrease in cell development and, thus, reduce cancer progression and symptoms.
This review aims to create a framework that will aid in investigating metformin and resveratrol as possible separate and/or combined complements to traditional glioblastoma treatment by discussing the in vitro, in vivo, clinical, and pharmacokinetic profiles of metformin and resveratrol in glioblastoma.

2. Metformin and Resveratrol on Glioblastoma’s Proliferative and Apoptotic Pathways

Signaling pathways have been a therapeutic target in cancer treatment as they exhibit major variations between different types of cancer and, more importantly, between healthy and diseased individuals. Some significant pathways activated and deactivated in cancer cells include PI3K/Akt, mTOR, AMPK, and MAPK.

2.1. PI3K/Akt Pathway

The PI3K/Akt signaling pathway is significant in cancer therapy as its activation results in downstream proteins and pathways that enhance cell proliferation and survival. MET inhibits the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway when applied to glioblastoma cell lines, primarily by inhibiting PI3K or phosphorylated Akt, as seen in Table 1. Inhibition of this pathway results in a decrease in the cellular invasion, migration, cell survival, and cell proliferation [16,17,18]. Additionally, it inhibits the cell cycle by arresting cells in the G1, S, and M phases and increasing the number of cells in the G0 phase preceding apoptosis induction [18].
RES, like MET, downregulates the PI3K/Akt and NF-κB pathways by decreasing the expression of PI3K class III, p-Akt, NF-κB, and miR-21. Consequently, cell proliferation, invasion, migration, and autophagy are diminished [19,20,21,22,23,24]. RES increases p53 expression, which increases apoptosis in U87 cell lines, specifically through SIRT1-dependent apoptosis [19,21,22]. An S-G2/M cell cycle arrest is also observed when glioblastoma cell lines are treated with resveratrol [23].
MET and RES inhibit PI3K/Akt signaling primarily by inhibiting the two PI3K phosphorylated subunits, the regulatory domain p85 and the catalytic domain p110, triggering a cascade of events that inhibits PIP2, PIP3, and PDK1, which are usually found to be upregulated in cancers as seen in Figure 1 [25,26,27,28,29].

2.2. mTOR Pathway

The target of rapamycin (mTOR) is crucial in several signaling pathways involved in glioblastoma cell growth, proliferation, and survival, making mTOR an exciting target for drugs such as MET and RES. It involves two complexes—mTORC1 (modulates growth and metabolism) and mTORC2 (modulates cell proliferation and survival)—and, when upregulated, it leads to tumor progression [30]. MET decreases mTOR expression alongside increased AMPK expression (an mTOR inhibitor) in human glioma cells [31]. Table 2 shows how the decreased phosphorylation of mTOR and increased AMPK expression were accompanied by increased apoptosis rates and apoptosis enzyme caspase-3 activity, aligning with mTOR’s role in cell proliferation [18,32,33].
RES shows similar effects of increased apoptosis and caspase activity through mTOR pathways. Additionally, it effectively synergized with temozolomide (TMZ) in SHG44 cells to inhibit mTOR through the AMPK/mTOR pathway, where the combination of TMZ and RES had a significantly greater inhibitory effect on mTOR phosphorylation than TMZ alone [34].
Due to mTOR’s vast signaling reach, the effects of MET and RES on the mTOR pathway occur primarily through the effects of MET and RES on signaling intermediates, including Akt, Hsp27, AMPK, Redd1, TSC1/2, and Bcl-2, as seen in Figure 2.

2.3. RAS/RAF/MAPK Pathway

The mitogen-activated protein kinase (MAPK) signaling pathway constitutes a kinase cascade with many signaling proteins to target and regulate. In Table 3 it can be seen that treatment with MET downregulates this pathway at many points, decreasing the expression of the MAP4K RAF, the MAP3K RAS, the MAP2K MEK-1, and the MAPK ERK-1 [36]. This decreased expression comes alongside decreased antiapoptotic Bcl-2. Another study on glioblastoma stem cells (GSC) suggests that MET induces autophagy and apoptosis by stimulating the MAPK pathways instead, likely through the MAPKs p38 and JNK [37].
Treatment with RES induces activation of the MAPK subfamily—including p-ERK, p-p38, and p-JNK—through ROS generation, inducing apoptosis as seen in Figure 3 [38]. This activation of the MAPK subfamily aligns with studies in which RES treatment activated p38-MAPK and thus increased autophagy rates in other cancer types [29]. Although not in glioblastoma cells specifically, RES also seems to block the MAPK pathway to induce apoptosis, likely through the pathway’s proliferative distributaries (such as the ERK1/2 cascade). Note that the MAPKs interact with many downstream proteins, and MET and RES’s effects on this pathway could be carried out through factors such as HSF1, Hsp27, Hsp70, and more [22].
Although the MAPK pathway contains cascades that are some of the most dysregulated in human cancers, the studied effects of MET and RES on this vital signaling pathway in glioblastoma cells are challenging to profile completely; this is due to the span of this large protein family and its overlapping upstream tributaries and downstream cascades. Further studies investigating distinct cascades under the MAPK umbrella (such as ERK1/2, p38, or the JNK1/2/3 pathways) and investigating the further downstream MAPKAPKs and transcription factors will allow a better understanding of the effects of MET and RES on this critical protein family and its many interactions.

2.4. AMPK Pathway

When activated by MET or RES, the AMP-activated protein kinase (AMPK) signaling pathway hinders viability and proliferation in glioblastoma cell lines, as seen in Table 4. MET primarily activates the AMPK signaling pathway, increasing the intracellular AMP to ATP ratio [39,40]. MET also activates FOXO3 through AMPK activation, eliminating glioma-initiating cells [41].
The activation of the AMPK signaling pathway by MET or RES leads to several effects on lipid and glucose metabolism: increased fatty acid catabolism through adipose triglyceride lipase (ATGL), regulation of fatty acid synthesis/oxidation through regulation of acetyl-CoA carboxylase (ACC), reduction of cholesterol synthesis through the reduction of HMG-CoA activity, inhibition of glycogenesis (GS), and the regulation of glucose uptake and glycolysis through TBC1D1, as seen in Figure 4 [40,43,44]. These effects, alongside other mTOR-mediated effects, improve glioblastoma cell metabolic programming, suppressing tumor growth.

2.5. Mitochondrial Pathway

Mitochondria are the powerhouses of the cell, involved in ATP production, proliferation, apoptosis, and calcium homeostasis in glioblastoma cells. MET reduces oxygen consumption and the activity of the electron transport chain complex I, thereby decreasing mitochondrial ATP production [18,45]. MET reduces membrane potential, mitochondrial transcription factor A (mtTFA), and coactivator PGC-1a, thus decreasing mitochondrial biogenesis [46]. As seen in Table 5, MET elevates lactate production, glucose consumption, ROS levels, and mitochondrial depolarization, resulting in mitochondrial apoptosis [47,48]. The increased lactate production (by reducing pyruvate) prevents pyruvate from feeding into the Krebs cycle and decreases ATP production.
Figure 5 shows how RES decreases mitochondrial ATP production and results in mitochondrial apoptosis and cell sensitivity by increasing caspase-3 activity, ROS production, and calcium ion influx [45,49]. RES also induces mitochondrial apoptosis by causing the collapse of the mitochondrial membrane potential, much like MET [50].

2.6. In Vivo

In vivo studies on xenograft mice models involved injecting them with glioblastoma cell lines and treating them with MET or RES. Table 6 describes the altered signaling pathways and how MET decreases tumor growth, volume, and cell proliferation, increasing cell death and model survival. It increases p-AMPK and active Caspase-3 and decreases Ki-67 and fatty acid synthase (FASN) [18,33,47].
RES also decreases tumor growth, volume, and the Ki-67 staining index and increases apoptosis, autophagy, and model survival. RES decreased Bcl-2, EGFR, NF-κB, COX-2, and VEGF levels [21,34,51].
The complete molecular pathway targeted at different locations by both MET and RES is seen in Figure 6.

3. Metformin and Resveratrol on Glucose in Glioblastoma

A positive correlation between elevated glucose levels and glioblastoma is described in Table 7. The interaction of glucose with different pathways, such as the nuclear factor kappa beta (NF-κB) pathway and glycolytic pathway, possibly increased cell proliferation, cell viability, tumorigenesis, NF-κB phosphorylation, and the expression of Bcl-2, Mcl-1, FPR1, EGFR, VEGF, ERK, EGF, ROS production, STAT3, PDK1, PDK3, ECH, and HADH [11,52,53,54]. These effects arose through multiple signaling intermediates and interactions. For example, high glucose upregulates the expression of a G-protein coupled chemoattractant receptor (GPCR), formyl peptide receptor 1 (FPR1), and epidermal growth factor receptor (EGFR) on GBM cells [11]. Similarly, high glucose promotes the expression of interleukin-1β, an upstream regulator of the NF-κB pathway. Glucose also contributes to chemoresistance by increasing the expression of Mcl-1 and antiapoptotic agents [55]. FR and STAT3 expression is upregulated with increased glucose levels, as happens in tumors [53,56,57].
A high glucose-induced increase in the Warburg effect is also observed: where cancer cells alter molecular pathways and switch from oxidative phosphorylation to aerobic glycolysis [5,53]. This alteration—the Warburg phenotype—then composes numerous selective molecular advantages for the growth and proliferation of tumor cells.
Clinically, hyperglycemia is associated with poor prognoses of glioblastoma and represents an independent prognostic factor for reduced survival in glioblastoma patients [58,59,60,61,62,63,64]. This is likely due to the effects of glucose levels on glioblastoma cells and tumor growth (as shown in Table 7), or due to the hyperinsulinemia that often accompanies hyperglycemia (insulin, a member of a family of growth factors, may itself promote tumor growth) [58,64,65].
Since in vitro glucose concentrations and blood glucose levels clinically correlate with glioblastoma cell survival and tumor growth, metformin and resveratrol’s blood glucose-decreasing functions—increasing glucose uptake and insulin sensitivity—cause glucose-dependent anticancer effects of MET and RES.

4. Clinical Considerations and Relevance

While in vitro and in vivo studies show potent and promising effects of MET and RES on glioblastoma cells, these models do not appropriately reflect human conditions due to apparent differences in available concentrations, metabolism, and delivery. To further understand the mechanistic effects displayed by the drugs and to discuss further the value of MET and RES in glioblastoma treatment, the drugs’ bioavailability, delivery methods, and clinical studies and indications must be discussed.

4.1. Bioavailibility

Despite their common usage, metformin is associated with low gastrointestinal absorption (40–60%) and bioavailability upon oral administration [66]. This naturally necessitates higher dose administrations and/or dose frequencies and could increase dose-related side effects, not to forget negatively affecting patient compliance. Steady-state plasma levels of metformin have been reported to range from 10 µmol/L to 40 µmol/L; a 850 mg dose thrice daily (with standard doses ranging from 500 to 2550 mg daily) leads to steady-state plasma concentrations of around 1.35 mg/L in both healthy and diabetic patients [67,68].
Resveratrol, when taken orally, is absorbed well with a bioavailability of around 70%; however, the availability of RES itself is minimal [69]. This is due to extensive liver and intestines metabolism that produces lower activity metabolites than RES through glucuronidation and sulfation [70]. For example, with a 25 mg oral dose, peak plasma levels of unchanged resveratrol were negligible (<5 ng/mL), while peak plasma levels of resveratrol and its metabolites reached about 2 µmol/L (491 ng/mL), showing 70% absorption.
In the case of glioblastoma, it is vital to consider the blood–brain barrier (BBB). The BBB comprises brain tissue capillaries’ endothelial cells and surrounding pericytes and astrocytes that form a barrier that tightly regulates the transfer of substances to the neural tissue [71]. Interestingly, six hours after administration to rat models, metformin had a brain-to-plasma ratio of 0.99, indicating that the drug concentration in brain tissue was equal to plasma levels, suggesting that metformin can somewhat penetrate the blood–brain barrier [72]. Resveratrol, however, showed minimal BBB permeability in a study concerned with polyphenol permeability [73].
The concentrations of metformin and resveratrol used in both in vitro and in vivo studies far exceed these serum concentrations of the drugs in humans. It is interesting to note, however, that both drugs accumulate in tissues, with metformin possibly accumulating in different tissues in concentrations 100 times greater than plasma levels and resveratrol and its metabolites accumulating in epithelial cells [70,74]. Nevertheless, this mismatch in concentrations necessitates the development of more efficacious delivery methods for both drugs and more clinical studies to complement the vast codex of in vitro and in vivo studies.

4.2. Delivery

To combat the drugs’ low bioavailability, decrease adverse reactions and metabolism, and increase targeting to the brain, several novel drug delivery methods of metformin and resveratrol have been studied. Since transport through the BBB depends on the lipid solubility of substances and phytochemicals, these methods involve lipid-designed drug delivery systems, nanotechnologies, and encapsulation methods [75].
To enhance delivery to the bloodstream and tissues in general, surface-modified nanostructured lipid carriers (PEGylated NLCs) have shown promise in improving metformin’s release, delivery, and bioavailability in in vivo enhancing pharmacokinetics, even at reduced doses [66]. Similarly, lipid nanocarriers, nanocrystals, and other technologies enhanced the in vivo pharmacokinetic profile and delivery of resveratrol [69,76,77].
Similar advancements showed promising results when targeting the brain specifically, which is vital in potential glioblastoma treatments. Lipid nanoparticles, borneol W/O/W composite submicron emulsions, and exosomes have shown increased metformin delivery to brain tissue compared to typical free metformin drug administration [78,79,80]. The exosome technology involved removing naturally created exosomes from the blood, loading them with metformin or the desired drug, and injecting the loaded exosomes as a therapeutic: not only did the exosomes cross the BBB, but they seemed to specifically accumulate in glioblastoma cells [80,81]. Similarly, nanocarriers, nanoparticles modified with brain-targeted peptides, and liposomes increased BBB crossing and the delivery of resveratrol to the brain [82,83,84].

4.3. Clinical Trials

There are very few clinical studies on metformin in glioblastoma, and none on resveratrol, despite the promising in vitro and in vivo research.
A phase I clinical study investigating the use of metformin with TMZ in newly diagnosed glioblastoma patients found that doses of 2250 mg of metformin per day were tolerable, with no dose-limiting toxicities, although manageable adverse effects included appetite loss, nausea, and diarrhea [85]. A similar phase I trial found that 850 mg of metformin twice daily was the tolerable dose with TMZ; the most significant dose-limiting toxicity of metformin in the study was anorexia, and the dose was decreased from a target of 1000 mg twice daily to 850 mg because of the adverse effects of nausea and dysgeusia [86]. A study exploring the combination of metformin with a modified Atkins diet and radiotherapy in patients with newly diagnosed and recurrent glioma found that 850 mg of metformin twice daily to be the tolerable dose to proceed into phase II trials [87].
Although there is a lack of clinical studies investigating resveratrol use in glioblastoma, clinical trials concerned with the pharmacokinetics of resveratrol and its effects in other cancers are relevant to this discussion. A phase I trial investigating resveratrol’s pharmacological effects in healthy humans found that consumption of given doses (0.5 to 5 g) did not cause any serious adverse effects; peak levels of resveratrol were 2.4 µmol/L, but the peak levels of three of its metabolites were three to eight times higher [88]. Another trial found that eight daily doses of 1 g of resveratrol were well tolerated in patients with colorectal cancer and slightly reduced tumor cell proliferation [89]. In patients with colorectal cancer and hepatic metastases, 5 g of resveratrol daily was well tolerated. Cleaved caspase-3 levels (an apoptosis marker) significantly increased by 39% in malignant hepatic tissue following resveratrol treatment compared to placebo treatments [90].

5. Conclusions

Metformin and resveratrol antidiabetic drugs show antineoplastic mechanisms against glioblastoma cells by increasing apoptosis and autophagy and decreasing proliferation by altering the PI3K/Akt, mTOR, AMPK, MAPK, and mitochondrial pathways. Alongside heavily affecting cancer cell glucose metabolism with potent effects in vitro and in vivo, and having new delivery mechanisms that ameliorate their poor bioavailability, metformin and resveratrol show promise in glioblastoma treatment. The large pool of evidence warrants further clinical research to profile the drugs’ pharmacokinetics in glioblastoma patients and investigate the effectiveness of metformin and resveratrol as separate or combined complements to the current glioblastoma treatments of chemotherapy or radiotherapy. As there is currently no literature available on the combined effects of metformin and resveratrol on glioblastoma cells, the combination of metformin and resveratrol treatment on GBM cell cultures must be investigated in vitro by employing varying concentrations of both metformin and resveratrol, and comparing these to a resveratrol-only control, a metformin-only control, and proper control. Appropriate analyses (viability assays, migration assays, gene expression analyses, and cell cycle analyses) must then be undertaken to confirm or deny the possibility of synergy between metformin and resveratrol on glioblastoma cells. Based on the results of the in vitro investigations, possible in vivo studies and clinical trials (phase I, non-inferiority, etc.) can be duly planned and executed. Such studies on glioblastoma cells and patients may lead to the discovery of a more effective and powerful therapy that can be implemented in future treatments.

Author Contributions

Conceptualization, R.S.I. and S.S.I.; resources, D.B.; writing—original draft preparation, R.S.I., S.S.I. and A.E.-N.; writing—review and editing, A.E.-N., D.B., R.S.I., S.S.I., P.K. and L.K.; visualization, R.S.I.; supervision, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a National Priorities Research Program grant (NPRP 11S- 1214-170101; awarded to Dietrich Büsselberg, June 2019–Current). The statements made herein are solely the responsibility of the authors.

Data Availability Statement

The data presented in this study are available in this article.

Acknowledgments

The publication of this article was funded by the Weill Cornell Medicine—Qatar Health Science Library.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

ACCAcetyl-CoA carboxylase
AKTProtein Kinase B
AMPKAdenosine Monophosphate-Activated Protein Kinase
ATGLAdipose triglyceride lipase
Bcl-2B Cell Lymphoma 2 Proteins
COX-2Cyclooxygenase-2
EGFREpidermal growth factor receptor
FASNFatty acid synthase
FOXO3Forkhead box protein O3
HSF1Heat shock factor 1
Hsp27small heat shock protein
JNK1/2/3c-Jun N-terminal protein kinase
MAPKMitogen-Activated Protein Kinase
METMetformin
mTORMammalian Target of Rapamycin
NF-κBNuclear factor kappa beta pathway
PI3KPhosphatidylinositol 3-Kinase
RAF/RAS/MAPK/MEK/ERKRapidly accelerated fibrosarcoma/Rat sarcoma/Mitogen activated protein kinase/ Mitogen activated protein kinase kinase/Extracellular signal regulated kinases.
RESResveratrol
ROSReactive Oxygen Species
SIRT1NAD-dependent deacetylase sirtuin-1
TMZTemozolomide
VEGFVascular endothelial growth factors

References

  1. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and Molecular Prognostic Review of Glioblastoma. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Miller, K.D.; Ostrom, Q.T.; Kruchko, C.; Patil, N.; Tihan, T.; Cioffi, G.; Fuchs, H.E.; Waite, K.A.; Jemal, A.; Siegel, R.L.; et al. Brain and Other Central Nervous System Tumor Statistics, 2021. CA Cancer J. Clin. 2021, 71, 381–406. [Google Scholar] [CrossRef] [PubMed]
  3. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; 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] [Green Version]
  4. Ostrom, Q.T.; Gittleman, H.; Xu, J.; Kromer, C.; 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 2009–2013. Neuro Oncol. 2016, 18, v1–v75. [Google Scholar] [CrossRef] [Green Version]
  5. Samec, M.; Liskova, A.; Koklesova, L.; Samuel, S.M.; Zhai, K.; Buhrmann, C.; Varghese, E.; Abotaleb, M.; Qaradakhi, T.; Zulli, A.; et al. Flavonoids against the Warburg Phenotype—Concepts of Predictive, Preventive and Personalised Medicine to Cut the Gordian Knot of Cancer Cell Metabolism. EPMA J. 2020, 11, 377. [Google Scholar] [CrossRef] [PubMed]
  6. Koklesova, L.; Liskova, A.; Samec, M.; Qaradakhi, T.; Zulli, A.; Smejkal, K.; Kajo, K.; Jakubikova, J.; Behzadi, P.; Pec, M.; et al. Genoprotective Activities of Plant Natural Substances in Cancer and Chemopreventive Strategies in the Context of 3P Medicine. EPMA J. 2020, 11, 261–287. [Google Scholar] [CrossRef] [PubMed]
  7. Samec, M.; Liskova, A.; Kubatka, P.; Uramova, S.; Zubor, P.; Samuel, S.M.; Zulli, A.; Pec, M.; Bielik, T.; Biringer, K.; et al. The Role of Dietary Phytochemicals in the Carcinogenesis via the Modulation of MiRNA Expression. J. Cancer Res. Clin. Oncol. 2019, 145, 1665–1679. [Google Scholar] [CrossRef]
  8. Hardie, D.G. A New Understanding of Metformin. In Comprehensive Pharmacology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 280–300. [Google Scholar] [CrossRef]
  9. Takhwifa, F.; Aninditha, T.; Setiawan, H.; Sauriasari, R. The Potential of Metformin as an Antineoplastic in Brain Tumors: A Systematic Review. Heliyon 2021, 7, E06558. [Google Scholar] [CrossRef]
  10. Rauf, A.; Imran, M.; Butt, M.S.; Nadeem, M.; Peters, D.G.; Mubarak, M.S. Resveratrol as an Anticancer Agent: A Review. Crit. Rev. Food Sci. Nutr. 2017, 58, 1428–1447. [Google Scholar] [CrossRef]
  11. Bao, Z.; Chen, K.; Krepel, S.; Tang, P.; Gong, W.; Zhang, M.; Liang, W.; Trivett, A.; Zhou, M.; Wang, J.M. High Glucose Promotes Human Glioblastoma Cell Growth by Increasing the Expression and Function of Chemoattractant and Growth Factor Receptors. Transl. Oncol. 2019, 12, 1155–1163. [Google Scholar] [CrossRef]
  12. Tseng, H.W.; Li, S.C.; Tsai, K.W. Metformin Treatment Suppresses Melanoma Cell Growth and Motility through Modulation of MicroRNA Expression. Cancers 2019, 11, 209. [Google Scholar] [CrossRef] [Green Version]
  13. Chen, K.; Qian, W.; Jiang, Z.; Cheng, L.; Li, J.; Sun, L.; Zhou, C.; Gao, L.; Lei, M.; Yan, B.; et al. Metformin Suppresses Cancer Initiation and Progression in Genetic Mouse Models of Pancreatic Cancer. Mol. Cancer 2017, 16, 131. [Google Scholar] [CrossRef] [Green Version]
  14. Wu, X.P.; Xiong, M.; Xu, C.S.; Duan, L.N.; Dong, Y.Q.; Luo, Y.; Niu, T.H.; Lu, C.R. Resveratrol Induces Apoptosis of Human Chronic Myelogenous Leukemia Cells in Vitro through P38 and JNK-Regulated H2AX Phosphorylation. Acta Pharmacol. Sin. 2015, 36, 353–361. [Google Scholar] [CrossRef]
  15. Kueck, A.; Opipari, A.W.; Griffith, K.A.; Tan, L.; Choi, M.; Huang, J.; Wahl, H.; Liu, J.R. Resveratrol Inhibits Glucose Metabolism in Human Ovarian Cancer Cells. Gynecol. Oncol. 2007, 107, 450–457. [Google Scholar] [CrossRef]
  16. Würth, R.; Pattarozzi, A.; Gatti, M.; Bajetto, A.; Corsaro, A.; Parodi, A.; Sirito, R.; Massollo, M.; Marini, C.; Zona, G.; et al. Metformin Selectively Affects Human Glioblastoma Tumor-Initiating Cell Viability. Cell Cycle 2012, 12, 145–156. [Google Scholar] [CrossRef] [Green Version]
  17. Hassan, M.A.; Fakhoury, I.; El Masri, Z.; Ghazale, N.; Dennaoui, R.; Atat, O.E.; Kanaan, A.; El-Sibai, M. Metformin Treatment Inhibits Motility and Invasion of Glioblastoma Cancer Cells. Anal. Cell. Pathol. 2018, 2018, 5917470. [Google Scholar] [CrossRef] [Green Version]
  18. Sesen, J.; Dahan, P.; Scotland, S.J.; Saland, E.; Dang, V.T.; Lemarié, A.; Tyler, B.M.; Brem, H.; Toulas, C.; Moyal, E.C.J.; et al. Metformin Inhibits Growth of Human Glioblastoma Cells and Enhances Therapeutic Response. PLoS ONE 2015, 10, e0123721. [Google Scholar] [CrossRef] [Green Version]
  19. Liu, L.; Zhang, Y.; Zhu, K.; Song, L.; Tao, M.; Huang, P.; Pan, Y. Resveratrol Inhibits Glioma Cell Growth via Targeting LRIG1. JBUON 2018, 23, 403–409. [Google Scholar]
  20. Jiao, Y.; Li, H.; Liu, Y.; Guo, A.; Xu, X.; Qu, X.; Wang, S.; Zhao, J.; Li, Y.; Cao, Y. Resveratrol Inhibits the Invasion of Glioblastoma-Initiating Cells via Down-Regulation of the PI3K/Akt/NF-ΚB Signaling Pathway. Nutrients 2015, 7, 4383–4402. [Google Scholar] [CrossRef] [Green Version]
  21. Clark, P.A.; Bhattacharya, S.; Elmayan, A.; Darjatmoko, S.R.; Thuro, B.A.; Yan, M.B.; van Ginkel, P.R.; Polans, A.S.; Kuo, J.S. Resveratrol Targeting of AKT and P53 in Glioblastoma and Glioblastoma Stem-like Cells to Suppress Growth and Infiltration. J. Neurosurg. 2017, 126, 1448–1460. [Google Scholar] [CrossRef] [Green Version]
  22. Önay Uçar, E.; Şengelen, A. Resveratrol and SiRNA in Combination Reduces Hsp27 Expression and Induces Caspase-3 Activity in Human Glioblastoma Cells. Cell Stress Chaperones 2019, 24, 763–775. [Google Scholar] [CrossRef] [PubMed]
  23. Filippi-Chiela, E.C.; Villodre, E.S.; Zamin, L.L.; Lenz, G. Autophagy Interplay with Apoptosis and Cell Cycle Regulation in the Growth Inhibiting Effect of Resveratrol in Glioma Cells. PLoS ONE 2011, 6, e20849. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, G.; Dai, F.; Yu, K.; Jia, Z.; Zhang, A.; Huang, Q.; Kang, C.; Jiang, H.; Pu, P. Resveratrol Inhibits Glioma Cell Growth via Targeting Oncogenic MicroRNAs and Multiple Signaling Pathways. Int. J. Oncol. 2015, 46, 1739–1747. [Google Scholar] [CrossRef] [Green Version]
  25. Xia, C.; Liu, C.; He, Z.; Cai, Y.; Chen, J. Metformin Inhibits Cervical Cancer Cell Proliferation by Modulating PI3K/Akt-Induced Major Histocompatibility Complex Class I-Related Chain A Gene Expression. J. Exp. Clin. Cancer Res. 2020, 39, 127. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, T.S.; Lee, M.; Park, M.; Kim, S.Y.; Shim, M.S.; Lee, C.Y.; Choi, D.H.; Cho, Y. Metformin and Dichloroacetate Suppress Proliferation of Liver Cancer Cells by Inhibiting MTOR Complex 1. Int. J. Mol. Sci. 2021, 22, 10027. [Google Scholar] [CrossRef]
  27. Liu, Y.; Zhang, Y.; Jia, K.; Dong, Y.; Ma, W. [Corrigendum] Metformin Inhibits the Proliferation of A431 Cells by Modulating the PI3K/Akt Signaling Pathway. Exp. Ther. Med. 2022, 24, 445. [Google Scholar] [CrossRef]
  28. Innets, B.; Thongsom, S.; Petsri, K.; Racha, S.; Yokoya, M.; Moriue, S.; Chaotham, C.; Chanvorachote, P. Akt/MTOR Targeting Activity of Resveratrol Derivatives in Non-Small Lung Cancer. Molecules 2022, 27, 8268. [Google Scholar] [CrossRef]
  29. Wang, J.; Li, J.; Cao, N.; Li, Z.; Han, J.; Li, L. Resveratrol, an Activator of SIRT1, Induces Protective Autophagy in Non-Small-Cell Lung Cancer via Inhibiting Akt/MTOR and Activating P38-MAPK. Onco. Targets Ther. 2018, 11, 7777–7786. [Google Scholar] [CrossRef] [Green Version]
  30. Zou, Z.; Tao, T.; Li, H.; Zhu, X. MTOR Signaling Pathway and MTOR Inhibitors in Cancer: Progress and Challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef] [Green Version]
  31. Xiong, Z.S.; Gong, S.F.; Si, W.; Jiang, T.; Li, Q.L.; Wang, T.J.; Wang, W.J.; Wu, R.Y.; Jiang, K. Effect of Metformin on Cell Proliferation, Apoptosis, Migration and Invasion in A172 Glioma Cells and Its Mechanisms. Mol. Med. Rep. 2019, 20, 887–894. [Google Scholar] [CrossRef]
  32. Yu, Z.; Zhao, G.; Xie, G.; Zhao, L.; Chen, Y.; Yu, H.; Zhang, Z.; Li, C.; Li, Y.; Yu, Z.; et al. Metformin and Temozolomide Act Synergistically to Inhibit Growth of Glioma Cells and Glioma Stem Cells in Vitro and in Vivo. Oncotarget 2015, 6, 32930–32943. [Google Scholar] [CrossRef] [Green Version]
  33. Lee, J.E.; Lim, J.H.; Hong, Y.K.; Yang, S.H. High-Dose Metformin Plus Temozolomide Shows Increased Antitumor Effects in Glioblastoma In Vitro and In Vivo Compared with Monotherapy. Cancer Res. Treat. 2018, 50, 1331–1342. [Google Scholar] [CrossRef] [Green Version]
  34. Yuan, Y.; Xue, X.; Guo, R.B.; Sun, X.L.; Hu, G. Resveratrol Enhances the Antitumor Effects of Temozolomide in Glioblastoma via ROS-Dependent AMPK-TSC-MTOR Signaling Pathway. CNS Neurosci. Ther. 2012, 18, 536–546. [Google Scholar] [CrossRef]
  35. Jiang, H.; Shang, X.; Wu, H.; Gautam, S.C.; Al-Holou, S.; Li, C.; Kuo, J.; Zhang, L.; Chopp, M. Resveratrol Downregulates PI3K/Akt/MTOR Signaling Pathways in Human U251 Glioma Cells. J. Exp. Ther. Oncol. 2009, 8, 25. [Google Scholar]
  36. Guarnaccia, L.; Navone, S.E.; Masseroli, M.M.; Balsamo, M.; Caroli, M.; Valtorta, S.; Moresco, R.M.; Campanella, R.; Schisano, L.; Fiore, G.; et al. Effects of Metformin as Add-On Therapy against Glioblastoma: An Old Medicine for Novel Oncology Therapeutics. Cancers 2022, 14, 1412. [Google Scholar] [CrossRef]
  37. Carmignani, M.; Volpe, A.R.; Aldea, M.; Soritau, O.; Irimie, A.; Florian, I.S.; Tomuleasa, C.; Baritchii, A.; Petrushev, B.; Crisan, G.; et al. Glioblastoma Stem Cells: A New Target for Metformin and Arsenic Trioxide. J. Biol. Regul. Homeost. Agents 2014, 28, 1–15. [Google Scholar]
  38. Jung, J.S.; Woo, J.S. Resveratrol Induces Cell Death through ROS-Dependent MAPK Activation in A172 Human Glioma Cells. J. Life Sci. 2016, 26, 212–219. [Google Scholar] [CrossRef] [Green Version]
  39. Mihaylova, M.M.; Shaw, R.J. The AMPK Signalling Pathway Coordinates Cell Growth, Autophagy and Metabolism. Nat. Cell Biol. 2011, 13, 1016–1023. [Google Scholar] [CrossRef]
  40. Mazurek, M.; Litak, J.; Kamieniak, P.; Kulesza, B.; Jonak, K.; Baj, J.; Grochowski, C. Metformin as Potential Therapy for High-Grade Glioma. Cancers 2020, 12, 210. [Google Scholar] [CrossRef] [Green Version]
  41. Sato, A.; Sunayama, J.; Okada, M.; Watanabe, E.; Seino, S.; Shibuya, K.; Suzuki, K.; Narita, Y.; Shibui, S.; Kayama, T.; et al. Glioma-Initiating Cell Elimination by Metformin Activation of FOXO3 via AMPK. Stem Cells Transl. Med. 2012, 1, 811. [Google Scholar] [CrossRef]
  42. Xing, J.; Wang, Z.; Xu, H.; Liu, C.; Wei, Z.; Zhao, L.; Ren, L. Pak2 Inhibition Promotes Resveratrol-Mediated Glioblastoma A172 Cell Apoptosis via Modulating the AMPK-YAP Signaling Pathway. J. Cell. Physiol. 2020, 235, 6563–6573. [Google Scholar] [CrossRef] [PubMed]
  43. Foretz, M.; Carling, D.; Guichard, C.; Ferre, P.; Foufelle, F. AMP-Activated Protein Kinase Inhibits the Glucose-Activated Expression of Fatty Acid Synthase Gene in Rat Hepatocytes. J. Biol. Chem. 1998, 273, 14767–14771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Hardie, D.G.; Ross, F.A.; Hawley, S.A. AMPK: A Nutrient and Energy Sensor That Maintains Energy Homeostasis. Nat. Rev. Mol. Cell Biol. 2012, 13, 251–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wu, Z.; Ho, W.S.; Lu, R. Targeting Mitochondrial Oxidative Phosphorylation in Glioblastoma Therapy. Neuromol. Med. 2022, 24, 18–22. [Google Scholar] [CrossRef] [PubMed]
  46. Feng, S.W.; Chang, P.C.; Chen, H.Y.; Hueng, D.Y.; Li, Y.F.; Huang, S.M. Exploring the Mechanism of Adjuvant Treatment of Glioblastoma Using Temozolomide and Metformin. Int. J. Mol. Sci. 2022, 23, 8171. [Google Scholar] [CrossRef]
  47. Valtorta, S.; Dico, A.L.; Raccagni, I.; Gaglio, D.; Belloli, S.; Politi, L.S.; Martelli, C.; Diceglie, C.; Bonanomi, M.; Ercoli, G.; et al. Metformin and Temozolomide, a Synergic Option to Overcome Resistance in Glioblastoma Multiforme Models. Oncotarget 2017, 8, 113090–113104. [Google Scholar] [CrossRef] [Green Version]
  48. Isakovic, A.; Harhaji, L.; Stevanovic, D.; Markovic, Z.; Sumarac-Dumanovic, M.; Starcevic, V.; Micic, D.; Trajkovic, V. Dual Antiglioma Action of Metformin: Cell Cycle Arrest and Mitochondria-Dependent Apoptosis. Cell. Mol. Life Sci. 2007, 64, 1290–1302. [Google Scholar] [CrossRef]
  49. Öztürk, Y.; Günaydın, C.; Yalçın, F.; Nazıroğlu, M.; Braidy, N. Resveratrol Enhances Apoptotic and Oxidant Effects of Paclitaxel through TRPM2 Channel Activation in DBTRG Glioblastoma Cells. Oxid. Med. Cell. Longev. 2019, 2019, 4619865. [Google Scholar] [CrossRef]
  50. Li, J.; Qin, Z.; Liang, Z. The Prosurvival Role of Autophagy in Resveratrol-Induced Cytotoxicity in Human U251 Glioma Cells. BMC Cancer 2009, 9, 215. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, L.; Long, L.; Wang, W.; Liang, Z. Resveratrol, a Potential Radiation Sensitizer for Glioma Stem Cells Both in Vitro and in Vivo. J. Pharmacol. Sci. 2015, 129, 216–225. [Google Scholar] [CrossRef] [Green Version]
  52. McKelvey, K.J.; Wilson, E.B.; Short, S.; Melcher, A.A.; Biggs, M.; Diakos, C.I.; Howell, V.M. Glycolysis and Fatty Acid Oxidation Inhibition Improves Survival in Glioblastoma. Front. Oncol. 2021, 11, 570. [Google Scholar] [CrossRef]
  53. Supabphol, S.; Seubwai, W.; Wongkham, S.; Saengboonmee, C. High Glucose: An Emerging Association between Diabetes Mellitus and Cancer Progression. J. Mol. Med. 2021, 99, 1175–1193. [Google Scholar] [CrossRef]
  54. Bielecka-Wajdman, A.M.; Ludyga, T.; Smyk, D.; Smyk, W.; Mularska, M.; Świderek, P.; Majewski, W.; Mullins, C.S.; Linnebacher, M.; Obuchowicz, E. Glucose Influences the Response of Glioblastoma Cells to Temozolomide and Dexamethasone. Cancer Control 2022, 29, 1–15. [Google Scholar] [CrossRef]
  55. Wu, X.; Luo, Q.; Liu, Z. Ubiquitination and Deubiquitination of MCL1 in Cancer: Deciphering Chemoresistance Mechanisms and Providing Potential Therapeutic Options. Cell Death Dis. 2020, 11, 556. [Google Scholar] [CrossRef]
  56. Hatanpaa, K.J.; Burma, S.; Zhao, D.; Habib, A.A. Epidermal Growth Factor Receptor in Glioma: Signal Transduction, Neuropathology, Imaging, and Radioresistance. Neoplasia 2010, 12, 675–684. [Google Scholar] [CrossRef] [Green Version]
  57. Piperi, C.; Papavassiliou, K.A.; Papavassiliou, A.G. Pivotal Role of STAT3 in Shaping Glioblastoma Immune Microenvironment. Cells 2019, 8, 1398. [Google Scholar] [CrossRef] [Green Version]
  58. Lu, V.M.; Goyal, A.; Vaughan, L.S.; McDonald, K.L. The Impact of Hyperglycemia on Survival in Glioblastoma: A Systematic Review and Meta-Analysis. Clin. Neurol. Neurosurg. 2018, 170, 165–169. [Google Scholar] [CrossRef]
  59. Mayer, A.; Vaupel, P.; Struss, H.G.; Giese, A.; Stockinger, M.; Schmidberger, H. Strong Adverse Prognostic Impact of Hyperglycemic Episodes during Adjuvant Chemoradiotherapy of Glioblastoma Multiforme. Strahlenther. Und Onkol. 2014, 190, 933–938. [Google Scholar] [CrossRef]
  60. Tieu, M.T.; Lovblom, L.E.; McNamara, M.G.; Mason, W.; Laperriere, N.; Millar, B.A.; Ménard, C.; Kiehl, T.R.; Perkins, B.A.; Chung, C. Impact of Glycemia on Survival of Glioblastoma Patients Treated with Radiation and Temozolomide. J. Neurooncol. 2015, 124, 119–126. [Google Scholar] [CrossRef] [Green Version]
  61. Adeberg, S.; Bernhardt, D.; Foerster, R.; Bostel, T.; Koerber, S.A.; Mohr, A.; Koelsche, C.; Rieken, S.; Debus, J. The Influence of Hyperglycemia during Radiotherapy on Survival in Patients with Primary Glioblastoma. Acta Oncol. 2015, 55, 201–207. [Google Scholar] [CrossRef] [Green Version]
  62. Chaichana, K.L.; McGirt, M.J.; Woodworth, G.F.; Datoo, G.; Tamargo, R.J.; Weingart, J.; Olivi, A.; Brem, H.; Quinones-Hinojosa, A. Persistent Outpatient Hyperglycemia Is Independently Associated with Survival, Recurrence and Malignant Degeneration Following Surgery for Hemispheric Low Grade Gliomas. Neurol. Res. 2010, 32, 442–448. [Google Scholar] [CrossRef] [PubMed]
  63. Hagan, K.; Bhavsar, S.; Arunkumar, R.; Grasu, R.; Dang, A.; Carlson, R.; Cowles, C.; Arnold, B.; Potylchansky, Y.; Rahlfs, T.F.; et al. Association between Perioperative Hyperglycemia and Survival in Patients with Glioblastoma. J. Neurosurg. Anesthesiol. 2017, 29, 21–29. [Google Scholar] [CrossRef] [PubMed]
  64. Derr, R.L.; Ye, X.; Islas, M.U.; Desideri, S.; Saudek, C.D.; Grossman, S.A. Association between Hyperglycemia and Survival in Patients with Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2009, 27, 1082–1086. [Google Scholar] [CrossRef] [PubMed]
  65. Gupta, K.; Krishnaswamy, G.; Karnad, A.; Peiris, A.N. Insulin: A Novel Factor in Carcinogenesis. Am. J. Med. Sci. 2002, 323, 140–145. [Google Scholar] [CrossRef]
  66. Kenechukwu, F.C.; Isaac, G.T.; Nnamani, D.O.; Momoh, M.A.; Attama, A.A. Enhanced Circulation Longevity and Pharmacodynamics of Metformin from Surface-Modified Nanostructured Lipid Carriers Based on Solidified Reverse Micellar Solutions. Heliyon 2022, 8, e09100. [Google Scholar] [CrossRef]
  67. Graham, G.G.; Punt, J.; Arora, M.; Day, R.O.; Doogue, M.P.; Duong, J.K.; Furlong, T.J.; Greenfield, J.R.; Greenup, L.C.; Kirkpatrick, C.M.; et al. Clinical Pharmacokinetics of Metformin. Clin. Pharm. 2011, 50, 81–99. [Google Scholar] [CrossRef]
  68. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-Activated Protein Kinase in Mechanism of Metformin Action. J. Clin. Investig. 2001, 108, 1167. [Google Scholar] [CrossRef]
  69. Li, C.; Wang, Z.; Lei, H.; Zhang, D. Recent Progress in Nanotechnology-Based Drug Carriers for Resveratrol Delivery. Drug Deliv. 2023, 30, 2174206. [Google Scholar] [CrossRef]
  70. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High Absorption but Very Low Bioavailability of Oral Resveratrol in Humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
  71. Zenaro, E.; Piacentino, G.; Constantin, G. The Blood-Brain Barrier in Alzheimer’s Disease. Neurobiol. Dis. 2017, 107, 41. [Google Scholar] [CrossRef] [Green Version]
  72. Łabuzek, K.; Suchy, D.; Gabryel, B.; Bielecka, A.; Liber, S.; Okopień, B. Quantification of Metformin by the HPLC Method in Brain Regions, Cerebrospinal Fluid and Plasma of Rats Treated with Lipopolysaccharide. Pharmacol. Rep. 2010, 62, 956–965. [Google Scholar] [CrossRef]
  73. Shimazu, R.; Anada, M.; Miyaguchi, A.; Nomi, Y.; Matsumoto, H. Evaluation of Blood-Brain Barrier Permeability of Polyphenols, Anthocyanins, and Their Metabolites. J. Agric. Food Chem. 2021, 69, 11676–11686. [Google Scholar] [CrossRef]
  74. Wilcock, C.; Bailey, C.J. Accumulation of Metformin by Tissues of the Normal and Diabetic Mouse. Xenobiotica 1994, 24, 49–57. [Google Scholar] [CrossRef]
  75. Kafoud, A.; Salahuddin, Z.; Ibrahim, R.S.; Al-Janahi, R.; Mazurakova, A.; Kubatka, P.; Büsselberg, D. Potential Treatment Options for Neuroblastoma with Polyphenols through Anti-Proliferative and Apoptotic Mechanisms. Biomolecules 2023, 13, 563. [Google Scholar] [CrossRef]
  76. Poonia, N.; Lather, V.; Narang, J.K.; Beg, S.; Pandita, D. Resveratrol-Loaded Folate Targeted Lipoprotein-Mimetic Nanoparticles with Improved Cytotoxicity, Antioxidant Activity and Pharmacokinetic Profile. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111016. [Google Scholar] [CrossRef]
  77. Argenziano, M.; Ansari, I.A.; Muntoni, E.; Spagnolo, R.; Scomparin, A.; Cavalli, R. Lipid-Coated Nanocrystals as a Tool for Improving the Antioxidant Activity of Resveratrol. Antioxidants 2022, 11, 1007. [Google Scholar] [CrossRef]
  78. Ebrahimi, H.; Kazem Nezhad, S.; Farmoudeh, A.; Babaei, A.; Ebrahimnejad, P.; Akbari, E.; Siahposht-Khachaki, A. Design and Optimization of Metformin-Loaded Solid Lipid Nanoparticles for Neuroprotective Effects in a Rat Model of Diffuse Traumatic Brain Injury: A Biochemical, Behavioral, and Histological Study. Eur. J. Pharm. Biopharm. 2022, 181, 122–135. [Google Scholar] [CrossRef]
  79. Hong, L.; Li, X.; Bao, Y.; Duvall, C.L.; Zhang, C.; Chen, W.; Peng, C. Preparation, Preliminary Pharmacokinetic and Brain Targeting Study of Metformin Encapsulated W/O/W Composite Submicron Emulsions Promoted by Borneol. Eur. J. Pharm. Sci. 2019, 133, 160–166. [Google Scholar] [CrossRef]
  80. Felker, J.; Agnihotri, S. Hurdling over the Blood–Brain Barrier with Exosome Technology. Neuro Oncol. 2022, 24, 1884–1885. [Google Scholar] [CrossRef]
  81. Zhan, Q.; Yi, K.; Cui, X.; Li, X.; Yang, S.; Wang, Q.; Fang, C.; Tan, Y.; Li, L.; Xu, C.; et al. Blood Exosomes-Based Targeted Delivery of CPLA2 SiRNA and Metformin to Modulate Glioblastoma Energy Metabolism for Tailoring Personalized Therapy. Neuro Oncol. 2022, 24, 1871–1883. [Google Scholar] [CrossRef]
  82. Vijayakumar, M.R.; Vajanthri, K.Y.; Balavigneswaran, C.K.; Mahto, S.K.; Mishra, N.; Muthu, M.S.; Singh, S. Pharmacokinetics, Biodistribution, in Vitro Cytotoxicity and Biocompatibility of Vitamin E TPGS Coated Trans Resveratrol Liposomes. Colloids Surf. B Biointerfaces 2016, 145, 479–491. [Google Scholar] [CrossRef] [PubMed]
  83. Jalili, C.; Kiani, A.; Gholami, M.; Bahrehmand, F.; Fakhri, S.; Kakehbaraei, S.; Kakebaraei, S. Brain Targeting Based Nanocarriers Loaded with Resveratrol in Alzheimer’s Disease: A Review. IET Nanobiotechnol. 2023, 17, 154–170. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, L.; Wang, Y.; Li, Z.; Wu, X.; Mei, J.; Zheng, G. Brain Targeted Peptide-Functionalized Chitosan Nanoparticles for Resveratrol Delivery: Impact on Insulin Resistance and Gut Microbiota in Obesity-Related Alzheimer’s Disease. Carbohydr. Polym. 2023, 310, 120714. [Google Scholar] [CrossRef] [PubMed]
  85. Ohno, M.; Kitanaka, C.; Miyakita, Y.; Tanaka, S.; Sonoda, Y.; Mishima, K.; Ishikawa, E.; Takahashi, M.; Yanagisawa, S.; Ohashi, K.; et al. Metformin with Temozolomide for Newly Diagnosed Glioblastoma: Results of Phase I Study and a Brief Review of Relevant Studies. Cancers 2022, 14, 4222. [Google Scholar] [CrossRef]
  86. Maraka, S.; Groves, M.D.; Mammoser, A.G.; Melguizo-Gavilanes, I.; Conrad, C.A.; Tremont-Lukats, I.W.; Loghin, M.E.; O’Brien, B.J.; Puduvalli, V.K.; Sulman, E.P.; et al. Phase 1 Lead-in to a Phase 2 Factorial Study of Temozolomide Plus Memantine, Mefloquine, and Metformin as Postradiation Adjuvant Therapy for Newly Diagnosed Glioblastoma. Cancer 2019, 125, 424. [Google Scholar] [CrossRef] [Green Version]
  87. Porper, K.; Shpatz, Y.; Plotkin, L.; Pechthold, R.G.; Talianski, A.; Champ, C.E.; Furman, O.; Shimoni-Sebag, A.; Symon, Z.; Amit, U.; et al. A Phase I Clinical Trial of Dose-Escalated Metabolic Therapy Combined with Concomitant Radiation Therapy in High-Grade Glioma. J. Neurooncol. 2021, 153, 487–496. [Google Scholar] [CrossRef]
  88. Boocock, D.J.; Faust, G.E.S.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I Dose Escalation Pharmacokinetic Study in Healthy Volunteers of Resveratrol, a Potential Cancer Chemopreventive Agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [Green Version]
  89. Patel, K.R.; Brown, V.A.; Jones, D.J.L.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical Pharmacology of Resveratrol and Its Metabolites in Colorectal Cancer Patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef] [Green Version]
  90. Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.P.; Gescher, A.J. Phase I Randomized, Double-Blind Pilot Study of Micronized Resveratrol (SRT501) in Patients with Hepatic Metastases—Safety, Pharmacokinetics, and Pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Metformin and resveratrol’s action on PI3K/Akt pathway. Metformin and resveratrol inhibit PI3K and Akt, decreasing cell proliferation. Resveratrol decreases NF−κB expression, resulting in cell cycle arrest and decreased cell proliferation. Metformin and resveratrol also arrest the cell cycle directly. Resveratrol upregulates p53, increasing apoptosis. Generated using BioRender.
Figure 1. Metformin and resveratrol’s action on PI3K/Akt pathway. Metformin and resveratrol inhibit PI3K and Akt, decreasing cell proliferation. Resveratrol decreases NF−κB expression, resulting in cell cycle arrest and decreased cell proliferation. Metformin and resveratrol also arrest the cell cycle directly. Resveratrol upregulates p53, increasing apoptosis. Generated using BioRender.
Cancers 15 03368 g001
Figure 2. Metformin and resveratrol’s action on mTOR. Metformin and resveratrol inhibit Akt and activate AMPK, activating TSC1/2 and inhibiting mTOR and cell proliferation. Metformin increases Redd1 expression, activating TSC1/2 and reducing cell proliferation. Resveratrol decreases Hsp27 expression, which decreases Akt activation. Resveratrol also increases Bcl−2, which increases apoptosis. Metformin and resveratrol inhibit mTOR, decreasing cell proliferation, survival, and cytoskeletal organization. Generated using BioRender.
Figure 2. Metformin and resveratrol’s action on mTOR. Metformin and resveratrol inhibit Akt and activate AMPK, activating TSC1/2 and inhibiting mTOR and cell proliferation. Metformin increases Redd1 expression, activating TSC1/2 and reducing cell proliferation. Resveratrol decreases Hsp27 expression, which decreases Akt activation. Resveratrol also increases Bcl−2, which increases apoptosis. Metformin and resveratrol inhibit mTOR, decreasing cell proliferation, survival, and cytoskeletal organization. Generated using BioRender.
Cancers 15 03368 g002
Figure 3. Metformin and resveratrol’s interactions with the mitogen-activated protein kinase (MAPK) pathway. Primarily, metformin and resveratrol inhibit the MAPK/ERK cascade, decreasing levels of MAPK4K’s (RAF), MAP3K’s (RAF), MAP2K’s (MEK1/2), MAPK’s (ERK1/2), and associated proteins (Bcl−2). Resveratrol activates MAPK’s, including p−38 and JNK. Generated using BioRender.
Figure 3. Metformin and resveratrol’s interactions with the mitogen-activated protein kinase (MAPK) pathway. Primarily, metformin and resveratrol inhibit the MAPK/ERK cascade, decreasing levels of MAPK4K’s (RAF), MAP3K’s (RAF), MAP2K’s (MEK1/2), MAPK’s (ERK1/2), and associated proteins (Bcl−2). Resveratrol activates MAPK’s, including p−38 and JNK. Generated using BioRender.
Cancers 15 03368 g003
Figure 4. Metformin and resveratrol’s action on AMPK pathway. Metformin and resveratrol activate AMPK and FOXO3, downregulating Akt, mTOR, and cell growth. AMPK regulates HMG−CoA, GS, ATGL, ACC, and TBC1D1. Generated using BioRender.
Figure 4. Metformin and resveratrol’s action on AMPK pathway. Metformin and resveratrol activate AMPK and FOXO3, downregulating Akt, mTOR, and cell growth. AMPK regulates HMG−CoA, GS, ATGL, ACC, and TBC1D1. Generated using BioRender.
Cancers 15 03368 g004
Figure 5. Metformin and resveratrol’s action on the mitochondria. Metformin increases lactate production, decreases oxygen feeding into the ETC, and inhibits complex 1 of the ETC. Additionally, metformin and resveratrol decrease ATP levels, ultimately reducing cell proliferation. Metformin increases mitochondrial membrane depolarization and ROS levels, causing mitochondrial dysfunction and mitochondrial apoptosis. Resveratrol increases ROS levels and the influx of calcium ions into the mitochondria, causing mitochondrial membrane depolarization, which leads to mitochondrial apoptosis, and increases caspase−3 levels, leading to apoptosis. Generated using BioRender.
Figure 5. Metformin and resveratrol’s action on the mitochondria. Metformin increases lactate production, decreases oxygen feeding into the ETC, and inhibits complex 1 of the ETC. Additionally, metformin and resveratrol decrease ATP levels, ultimately reducing cell proliferation. Metformin increases mitochondrial membrane depolarization and ROS levels, causing mitochondrial dysfunction and mitochondrial apoptosis. Resveratrol increases ROS levels and the influx of calcium ions into the mitochondria, causing mitochondrial membrane depolarization, which leads to mitochondrial apoptosis, and increases caspase−3 levels, leading to apoptosis. Generated using BioRender.
Cancers 15 03368 g005
Figure 6. An overview of metformin and resveratrol’s actions on glioblastoma. Metformin and resveratrol inhibit PI3K, Akt, and the MAPK/ERK pathway and increase AMPK. Consequently, they increase TSC1/2, decrease mTOR, cell proliferation, cell survival, and cytoskeletal organization, and trigger apoptosis and autophagy. Metformin also increases Redd1 levels, which increases TSC1/2. Resveratrol decreases Hsp27 and Bcl-2 and increases p53 and MAPKs, which include JNK and p38, increasing apoptosis and decreasing NF-κB, inhibiting the cell cycle and cell proliferation. Metformin and resveratrol both inhibit the cell cycle halting cell proliferation. Metformin inhibits the electron transport chain by increasing lactate production, decreasing oxygen levels fed into the electron transport chain, and inhibiting complex 1. Metformin and resveratrol decrease ATP levels, decreasing cell proliferation. Resveratrol and metformin increase ROS levels, which cause mitochondrial apoptosis associated with mitochondrial membrane depolarization. Resveratrol increases caspase-3 levels, leading to apoptosis. Generated using BioRender.
Figure 6. An overview of metformin and resveratrol’s actions on glioblastoma. Metformin and resveratrol inhibit PI3K, Akt, and the MAPK/ERK pathway and increase AMPK. Consequently, they increase TSC1/2, decrease mTOR, cell proliferation, cell survival, and cytoskeletal organization, and trigger apoptosis and autophagy. Metformin also increases Redd1 levels, which increases TSC1/2. Resveratrol decreases Hsp27 and Bcl-2 and increases p53 and MAPKs, which include JNK and p38, increasing apoptosis and decreasing NF-κB, inhibiting the cell cycle and cell proliferation. Metformin and resveratrol both inhibit the cell cycle halting cell proliferation. Metformin inhibits the electron transport chain by increasing lactate production, decreasing oxygen levels fed into the electron transport chain, and inhibiting complex 1. Metformin and resveratrol decrease ATP levels, decreasing cell proliferation. Resveratrol and metformin increase ROS levels, which cause mitochondrial apoptosis associated with mitochondrial membrane depolarization. Resveratrol increases caspase-3 levels, leading to apoptosis. Generated using BioRender.
Cancers 15 03368 g006
Table 1. Metformin and resveratrol on the PI3K/Akt pathway in glioblastoma cells.
Table 1. Metformin and resveratrol on the PI3K/Akt pathway in glioblastoma cells.
Cell LineIncubation ConcentrationResultsReferences
Metformin
SF2682.5 mM for 24 h phosphorylation of Akt[17]
cellular invasion
migration
GBM1–4GBM1: 9.2 mM
GBM2: 4.9 mM
GBM3: 9.0 mM
GBM4: 9.4 mM
for 48 h
phosphorylation of Akt[16]
cell survival
proliferation
U87, LN18, U251, SF7671, 5, 10 mM for 6 days Akt phosphorylation[18]
PI3K pathway
cell proliferation
G1 phase
cells in G0 phase
Resveratrol
U251100 mM for 48 h LRIG1[19]
EGFR
apoptosis
cell proliferation
GIC 400, 411, 41220 μM for 48 h Akt phosphorylation[20]
NF-κB
cell invasion
GSC (44-GSC)
U87
0, 5, 25, 50, 100 μM
for 4–48 h
AKT protein activation[21]
expression of p53
cell proliferation
cell migration
apoptosis
U87N/A PI3K/AKT[22]
NF-κB
SIRT1- dependent apoptosis
cell proliferation
U87, U138, U25130 µM or 100 µM for 48 h PI3K class III[23]
number of cells undergoing autophagy
number of mature autophagosomes formed per cell
S-G2/M cell cycle arrest
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 2. Metformin and resveratrol on the mTOR pathway in glioblastoma cells.
Table 2. Metformin and resveratrol on the mTOR pathway in glioblastoma cells.
Cell LineIncubation ConcentrationResultsReferences
Metformin
A1720, 0.1, 1, 10 mM for 24–72 h apoptosis[31]
AMPK and pAMPK
proliferation
mTOR/Bcl-2
invasion
migration
U87
U251
A172
5, 10, 20 mM for 24–72 h mTOR phosphorylation[33]
AMPK phosphorylation
proliferation
apoptosis
U87
LN18
U251
SF767
10 mM for 48 h mTOR phosphorylation[18]
Redd1
proliferation
apoptosis
autophagy
U87
U251
10 mM for 0–48 h
0–20 mM for 48 h
Akt/mTOR pathway[32]
phosphorylated mTOR
proliferation
apoptosis
Resveratrol
SHG4410 µM for 72 h ROS production[34]
AMPK
mTOR
Bcl-2
apoptosis
proliferation
U251100 μM for 24 h phosphorylated Akt[35]
phosphorylated mTOR
caspase-3
apoptosis
U8710 or 15 μM for 48 h mTOR[22]
HSF1
Hsp27 expression
proliferation
apoptosis
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 3. Metformin and resveratrol on the MAPK pathway in glioblastoma cells.
Table 3. Metformin and resveratrol on the MAPK pathway in glioblastoma cells.
Cell LineIncubation ConcentrationResultsReferences
Metformin
GBM tissue samples5 mM, 10 mM, 20 mM, 50 mM RAF/RAS/MAPK/MEK/ERK[36]
Bcl-2
viability
proliferation
apoptosis
GSCN/A MAPK[37]
autophagy
apoptosis
Resveratrol
A172100 µΜ ROS-induced activation of MAPK subfamily[38]
apoptosis
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 4. Metformin and resveratrol on the AMPK pathway in glioblastoma cells.
Table 4. Metformin and resveratrol on the AMPK pathway in glioblastoma cells.
Cell LineIncubation ConcentrationResultsReferences
Metformin
A1720, 0.1, 1, 10 mM for 24, 48, 72 h AMPK phosphorylation[31]
Bax expression
apoptosis
proliferation
U87
U251
A172
5, 10, 20 mM for 24, 48, 72 h AMPK phosphorylation[33]
U87
LN18
U251
SF767
10 mM for 48 h AMPK phosphorylation[18]
U87
U251
10 mM for 0–48 h
0–2 0 mM for 48 h
AMPK phosphorylation[32]
proliferation
apoptosis
GICs1 mM AMPK phosphorylation[41]
FOXO3 activation
Resveratrol
A172N/A AMPK and YAP transcription[42]
cell viability
apoptosis
SHG4410 µM for 72 h ROS production[34]
AMPK phosphorylation
mTOR
apoptosis
G2/M arrest
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 5. Metformin and resveratrol on the mitochondrial pathway in glioblastoma cells.
Table 5. Metformin and resveratrol on the mitochondrial pathway in glioblastoma cells.
Cell LineIncubation ConcentrationResultsReferences
Metformin
U87
LN18
U251
SF767
10 mM for 48 h oxygen consumption[18]
mitochondrial dependent ATP production
glycolytic ATP production
lactate production
ETC1 activity
U87MG
LNZ308
LN229
0, 25, 50, 75, 100, 125 mM for 24 h PGC-1α[46]
mtTFA
ROS
mitochondrial biogenesis
mitochondrial membrane potential
U2514 mM for 24 h ROS production[48]
mitochondrial depolarization
apoptosis
U251
T98G
10 mM for 24, 48, 72 h glucose consumption[47]
lactate production
Resveratrol
DBTRG50 µM for 24 h Ca2+ influx[49]
mitochondrial apoptosis
caspase 3 activity
ROS production
cell sensitivity
U251150 µM for 6–72 h collapsed mitochondria membrane potential[50]
apoptosis
N/AN/A mitochondrial-dependent ATP production[45]
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 6. Metformin and resveratrol on xenograft mice models inoculated with glioblastoma cells.
Table 6. Metformin and resveratrol on xenograft mice models inoculated with glioblastoma cells.
Cell LineApplied ConcentrationResultsReferences
Metformin
Athymic nude mice inoculated with U87 cells2 mg/25 g/day for 4 weeks phosphorylated AMPK[33]
Fatty acid synthase (FASN)
tumor growth
survival in models
NU/NU athymic mice injected with U87 and LN18 cells200 mL of 300 mg/kg/day for 30 days active caspase-3[18]
Ki67
tumor growth
cell proliferation
cell death
Female nude mice injected with U251 or T98G cells250 mg/kg/day for 21 days tumor volume only when combined with (400 mg/kg) TMZ[47]
tumor growth when combined with (400 mg/kg) TMZ
Resveratrol
BALB/cA nude mice injected with SHG44 cellsOral administration 40 mg/kg tumor volume when combined with (68 mg/kg) TMZ[34]
Ki-67 staining index when combined with (68 mg/kg) TMZ
BALB/cA nude mice injected with U87 cells0.1 mg/mL or 50 mg/kg or 5 injections of 200 mL of 5 mg over 2 weeks tumor volume[21]
tumor growth
BALB/cA nude mice injected with SU-2 cells150 mg/kg tumor growth[51]
Bcl-2
apoptosis
autophagy
Rat models with C6 gliomaOral administration RES 8 mg/kg/day survival in models[24]
tumor growth
number of apoptotic cells
EGFR, NF-κB, COX-2 and VEGF
Note: denotes increase of biomarker, denotes decrease of biomarker.
Table 7. Effect of elevated glucose levels on glioblastoma cell lines in in vitro studies.
Table 7. Effect of elevated glucose levels on glioblastoma cell lines in in vitro studies.
Cell LineIncubation ConcentrationResultsReferences
U8725 mM glucose for 24, 48, 72 h cell proliferation[11]
cell survival
tumorigenesis
Bcl-2
Mcl-1
NF-κB phosphorylation
FPR1
EGFR
VEGF
T98G
HROG02
HROG17
4.5 g/L glucose for 48 h cell viability[54]
GBM cell division
Dispersal
U87
U251
T98G
5, 10, 40 mg/mL glycolytic activity[52]
expression of PDK1, PDK3, ECH, and HADH
N/AN/A ERK[53]
STAT3
EGF
EGFR
ROS production
NF-ĸB
cell proliferation
anti-apoptosis
VEGF
Warburg effect
impaired mitochondrial function
Note: denotes increase of biomarker, denotes decrease of biomarker.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ibrahim, R.S.; Ibrahim, S.S.; El-Naas, A.; Koklesová, L.; Kubatka, P.; Büsselberg, D. Could Metformin and Resveratrol Support Glioblastoma Treatment? A Mechanistic View at the Cellular Level. Cancers 2023, 15, 3368. https://doi.org/10.3390/cancers15133368

AMA Style

Ibrahim RS, Ibrahim SS, El-Naas A, Koklesová L, Kubatka P, Büsselberg D. Could Metformin and Resveratrol Support Glioblastoma Treatment? A Mechanistic View at the Cellular Level. Cancers. 2023; 15(13):3368. https://doi.org/10.3390/cancers15133368

Chicago/Turabian Style

Ibrahim, Raghad Sabaawi, Shahad Sabaawi Ibrahim, Ahmed El-Naas, Lenka Koklesová, Peter Kubatka, and Dietrich Büsselberg. 2023. "Could Metformin and Resveratrol Support Glioblastoma Treatment? A Mechanistic View at the Cellular Level" Cancers 15, no. 13: 3368. https://doi.org/10.3390/cancers15133368

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