Next Article in Journal
Design and Assembly of a Biofactory for (2S)-Naringenin Production in Escherichia coli: Effects of Oxygen Transfer on Yield and Gene Expression
Next Article in Special Issue
Systemic Photoprotection in Melanoma and Non-Melanoma Skin Cancer
Previous Article in Journal
Association between Serum Levels of Interleukin-25/Thymic Stromal Lymphopoietin and the Risk of Exacerbation of Chronic Obstructive Pulmonary Disease
Previous Article in Special Issue
Bicalutamide Anticancer Activity Enhancement by Formulation of Soluble Inclusion Complexes with Cyclodextrins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Treatment Options for Neuroblastoma with Polyphenols through Anti-Proliferative and Apoptotic Mechanisms

1
Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
2
Department of Anatomy, 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.
Biomolecules 2023, 13(3), 563; https://doi.org/10.3390/biom13030563
Submission received: 24 January 2023 / Revised: 11 March 2023 / Accepted: 16 March 2023 / Published: 20 March 2023

Abstract

:
Neuroblastoma (NB) is an extracranial tumor of the peripheral nervous system arising from neural crest cells. It is the most common malignancy in infants and the most common extracranial solid tumor in children. The current treatment for high-risk NB involves chemotherapy and surgical resection followed by high-dose chemotherapy with autologous stem-cell rescue and radiation treatment. However, those with high-risk NB are susceptible to relapse and the long-term side effects of standard chemotherapy. Polyphenols, including the sub-class of flavonoids, contain more than one aromatic ring with hydroxyl groups. The literature demonstrates their utility in inducing the apoptosis of neuroblastoma cells, mostly in vitro and some in vivo. This review explores the use of various polyphenols outlined in primary studies, underlines the pathways involved in apoptotic activity, and discusses the dosage and delivery of these polyphenols. Primary studies were obtained from multiple databases with search the terms “neuroblastoma”, “flavonoid”, and “apoptosis”. The in vitro studies showed that polyphenols exert an apoptotic effect on several NB cell lines. These polyphenols include apigenin, genistein, didymin, rutin, quercetin, curcumin, resveratrol, butein, bisphenols, and various plant extracts. The mechanisms of the therapeutic effects include calpain-dependent pathways, receptor-mediated apoptosis, and, notably, and most frequently, mitochondrial apoptosis pathways, including the mitochondrial proteins Bax and Bcl-2. Overall, polyphenols demonstrate potency in decreasing NB proliferation and inducing apoptosis, indicating significant potential for further in vivo research.

1. Introduction

Neuroblastoma (NB) is an embryonal extracranial tumor of the peripheral nervous system derived from the neural crest. This cancer mainly affects children, with the median age of diagnosis being 17–18 months. Moreover, NB is the most common malignancy diagnosed within the first year of life [1]. It is likely to relapse and develop acquired drug resistance, posing a challenge in treatment [2]. The current treatment for high-risk NB involves chemotherapy and surgical resection followed by high-dose chemotherapy with autologous stem-cell rescue and radiation treatment [3]. However, those with high-risk NB are susceptible to relapses and the long-term side effects of standard chemotherapy [4]. The threat of multidrug resistance is also not uncommon, especially with the increasing intensity of therapy, which increases the likelihood of chemotherapeutic treatment failure in high-risk NB [5]. This necessitates the study of novel therapeutic agents, which would be less likely to result in adverse side effects. Many of these agents fall in the realm of natural dietary compounds, which several previous studies have demonstrated as having promising anti-cancer effects [6,7,8,9,10,11,12,13].
Natural polyphenols are organic compounds in plants that contain one phenol ring or more and exhibit anti-cancer potential [10,11,12]. Although they have the general structure of containing phenolic groups, polyphenols are a large and diverse class of molecules with many ubiquitous subgroups in plants, including fruits, teas, and vegetables. The main classes of polyphenols, as per one classification system, are phenolic acids (e.g., curcumin), phenolic alcohols, flavonoids, stilbenes (e.g., resveratrol), and lignans [14]. Polyphenols exert their anti-cancer properties in many ways, including but not limited to cell-cycle inhibition, the inhibition of proliferative enzymes, and apoptosis activation [13]. In general, polyphenols have low bioavailability for several reasons, such as their low water solubility, trouble with targeted delivery, and rapid elimination. However, polyphenols demonstrate significant biological effects; thus, a “low bioavailability/high bioactivity paradox” is highlighted in many studies [15].
Long-term polyphenol ingestion has been demonstrated to have positive and negative effects on human health and drug delivery. In vitro studies of green tea containing many polyphenols, such as EGCG, showed anti-cancer effects in breast, prostrate, liver, head, and neck cancers [16]. Green tea has also been linked to a lower risk of cognitive deterioration [17]. Polyphenols can slow the incidence rate of coronary heart disease by reducing platelet aggregation. Quercetin, for example, disrupts atherosclerotic plaque formation by inhibiting MMP1, which is involved in inflammation, atherosclerosis, and plaque formation [18,19]. Polyphenols also have anti-inflammatory and antioxidant properties and can help manage and prevent type 2 diabetes by protecting beta-pancreatic cells from glucose toxicity [20]. Clinical studies investigating polyphenol interaction with cancer cells often find that polyphenols have no adverse effect on normal cells [21].
Flavonoids are naturally occurring chemicals and are members of the polyphenol family [22]. However, their structure distinguishes them from polyphenols; polyphenols have a more complicated structure than flavonoids. Almost all flavonoids have a C6–C3–C6 structure with two benzene rings, labeled A and B in Figure 1, connected by an oxygen-containing heterocyclic-pyrene ring (C), whereas polyphenols do not have a comparable structure [23].
There is firm support in the literature for the argument that flavonoids have a powerful anti-cancer effect. Flavonoids exert potent anti-neoplastic capacity via their modulation of reactive oxygen species (ROS) enzyme activity, support for cell-cycle arrest, induction of apoptosis, and lowering of proliferation [24]. Flavonoids are beneficial against a wide range of cancers, including NB. It is important to note that most studies on flavonoids, and polyphenols in general, are conducted with isolated compounds. However, complementary or synergistic interactions between natural compounds with similar anti-cancer mechanisms can enhance their overall curative and preventive properties and their bioavailability [10]. An understanding of how to apply flavonoids to NB in a way that has the best effect on the reduction in NB would be highly beneficial due to the various apoptotic and anti-proliferative pathways in NB cell lines that flavonoid treatments can exploit.

2. Study Methodology

The keywords “neuroblastoma”, “apoptosis” and “flavonoids” or “polyphenols” were used to obtain primary in vitro studies from multiple databases, including Google Scholar, MDPI, Springer, and PubMed. Studies were analyzed for relevant data, which included the identification of NB apoptosis followed by the application of compound, as well as insight into the apoptotic mechanisms. Studies concerning the neuroprotective, rather than the cytotoxic potential, of flavonoids were excluded. Twenty-six studies were included, from which four major apoptotic pathways were identified. Data were sorted into tables according to similarities in pathways, and shared apoptotic mechanisms were identified and mapped out in figures generated on BioRender.

3. Mechanisms of Apoptotic Action

3.1. Calpain-Dependent Apoptotic Pathway

The release of calcium (Ca2+) from the endoplasmic reticulum (ER) leads to the activation of calpains. Calpain is a Ca2+-activated endo-protease involved in apoptotic mechanisms [25]. Exposure to the compounds listed in Table 1 activates proteolytic pathways involving calpain, leading to NB-cell apoptosis. Specifically, apigenin, epigallocatechin (EGC), epigallocatechin gallate (EGCG), and genistein trigger ER stress, thus increasing intracellular free Ca2+ and causing calpain activation at the ER membrane [26,27], resulting in the degradation of cytoskeletal proteins and the destabilization of cellular integrity in SH-SY5Y cells. Caspase release is also stimulated, with caspase-3 activating caspase-activated DNase (CAD), contributing to DNA fragmentation [8,28]. Caspase-12 is also activated when apigenin and genistein are applied, reinforcing their apoptotic effects [27]. Several studies concerning other cancer types reinforced the calpain–caspase apoptotic pathway, illustrated in Figure 2 [25,26,27,28,29].

3.2. Anti-Proliferative Pathways

In vitro studies on the anti-cancer effects of flavonoids and non-flavonoid polyphenols in NB cell lines reveal remarkable anti-proliferative effects, as demonstrated in Figure 3 and Table 2. Quercetin exhibits anti-proliferative effects by increasing p27 mRNA expression, inhibiting the formation and activity of the cyclin–cyclin-dependent kinase (cyclin/CDK) complex, disrupting the cell cycle in NB. Quercetin also reduces B-cell lymphoma-w (Bcl-w) mRNA expression, which decreases tumor-gene expression and induces apoptosis in NB cells [30]. Quercetin can further inhibit NB-cell growth by blocking voltage-gated potassium (K+) channel activity [31]. Other flavonoids, such as apigenin and 2-(cis-1,2-dihydroxy-4-oxo-cyclohex-5-enyl)-5,7-dihydroxy-chromone (DEDC), increase p53 and p21 mRNA expression while decreasing cyclin-B1 expression in NB [32,33]. Didymin decreases the proliferation of NB cells via the downregulation of the phosphoinositide 3-kinase (PI3K) and Akt pathways, accompanied by reduced vimentin levels, indicating a decrease in cell motility. Furthermore, proto-oncogene N-Myc transcription was inhibited by didymin. Increased Raf-1 kinase inhibitor protein (RKIP) levels inhibit the mitogen-activated protein kinase (MAPK) pathway, also decreasing proliferation [34]. Isoliquiritigenin inhibits cell motility and increases the activation of extracellular regulated kinase 1/2 (pERK1/2), which inhibits NB-cell migration and proliferation while arresting the cell cycle in the S phase [35]. Isoliquiritigenin and rutin both enhance G2/M-phase arrest in NB [35,36].
Non-flavonoid polyphenols affect multiple cell lines through multiple pathways. Curcumin decreases CDC2 and cyclin-B1, resulting in NB-cell-cycle arrest in the G2/M phase [37]. Furthermore, it reduces NF-κB activator protein (AP-1) and STAT3 and STAT5 activation, suppressing gene transcription [38]. Honokiol inhibits NB-cell-cycle progression at the sub-G1 phase [39]. Resveratrol reduced Cyclin D1 levels in NB cells, causing cell-cycle arrest in the S phase [40]. Similarly, the treatment of NB cells with resveratrol resulted in a significant drop in pAkt, Cyclin D, E, A, and CDK2 levels and increased p53 and NF-κB, resulting in cell-cycle arrest in the S phase [41]. Resveratrol causes p21 levels to rise, which inhibits CDK levels and causes cell-cycle arrest in the G1, G2/M, and S phases in NB cell lines [42]. Prenyl hydroxy coumarin derivatives also have notable anti-proliferative effects on NB cell lines, inducing cell-cycle arrest in the sub-G1 phase, with no effect on normal lymphocytic cells [43].
In addition to isolated flavonoids and non-flavonoid polyphenolic compounds, recent research supports the potent anti-cancer effects of whole plants or plant extracts characterized by numerous phytochemicals acting synergistically or additively [44,45,46,47]. For example, a recent study by Morandi et al. (2021) demonstrated the capacity of olive-leaf extract (rich in phenolic compounds) to inhibit the proliferation of NB cells through cell arrest in the G0/G1 phase and the accumulation of cells in the sub-G0 phase, accompanied by the induction of apoptosis [48]. Numerous other recent studies highlight the anti-cancer potential of plant extracts; for example, the fruit extract of Kigelia Africana, a plant rich in flavonoids that are used in traditional African medicine, inhibited proliferation and other mechanisms associated with carcinogenesis in NB cells [49]. Interestingly, research results obtained by Roomi et al. (2013) suggested the therapeutic potential of a nutrient mixture of lysine, proline, ascorbic acid, and green-tea extracts for NB management through the inhibition of tumor growth and proliferation and the induction of apoptosis in neuroblastoma models in vitro and in vivo [50]. Indeed, green tea is rich in numerous phytochemicals, mainly catechins. Green-tea catechins, including ECG, EGCG, and EGC, are phytochemicals with strong anti-cancer effects [51,52].
Table 2. Results of in vitro studies of polyphenols’ anti-proliferative effects on NB cell lines *.
Table 2. Results of in vitro studies of polyphenols’ anti-proliferative effects on NB cell lines *.
CompoundCell LineIncubation PeriodConcentration(s)Biomarker Changes and EffectsReference
Flavonoids
ApigeninNUB-7 and LAN-524 h10, 50, 100, 150, 200 µM
IC50: 35 μM in NUB-7
IC50: 22 μM in LAN-5
p53
p21WAF−1/CIP−1
Proliferation[33]
DEDCSH-SY5Y24 h7.5 µg/mL p53 mRNA
p21 mRNA
Cyclin-B1
[32]
DidyminCHLA-90 and SK-N-BE2 (p53-mutant) l + SMS-KCNR and LAN-5 (p53 wild-type)24 h50 μmol/L P13K
Akt
Proliferation[34]
Vimentin Motility of tumor cells
N-Myc transcription
RKIP MAPK pathway
Proliferation
IsoliquiritigeninSH-SY5Y24 h10–100 µM
IC50: 25.4 µM
pERK1/2 Cell migration
Proliferation
S + G2/M-phase arrest
[35]
RutinLAN-524 h 0, 25, 50, 100 μM G2/M-phase arrest[36]
Quercetin Neuro2a (mouse cell line) 24 h 10, 20, 40, 80, 120 μM
IC50: 40 µM
p27 Cyclin–CDK complex binding[30]
Bcl-w Tumor-cell-gene expression
Quercetin Neuroblastoma X glioma NG 108-15 cells (mouse cell line) 48 h 10 µM, 20 µM
IC50: 10 µM
K+-channel activity Cell growth[31]
Non-Flavonoid Polyphenols
Curcumin SK-N-SH 24 h 8, 16, 32 µM CDC2
Cyclin B1
G2/M-phase arrest[37]
Curcumin GI-L-IN, HTLA-230, SH-SY5Y, LAN5, SK-NBE2c, and IMR-32 18–72 h 0.1–25 µM NFκβ activator protein (AP-1)
STAT3, STAT5 activation
Cell growth[38]
Curcumin NUB-7, LAN-5, IMR-32 and SK-N-BE(2) 2–8 days0–100 µM *
* Significantly inhibited proliferation in the range of 5–10 µM
p53 translocation from cytoplasm to nucleus
p21WAF−1/CIP−1
G1-, G2/M-, and S-phase arrest[42]
Honokiol Neuro-2a (mouse cell line) and NB41A3 72 h 2.5, 5, 10, 20,
30, 40, 50, 60,
80, 100 µM
LC50: 63.3 µM
Sub-G1-phase arrest[39]
Prenyl hydroxy-coumarins Neuro-2a (mouse cell line) 24, 48, 72 h 6.25–200 µg/mL Sub-G1-phase arrest[43]
Resveratrol B103 (rat cell line) 48 h 5–20 µM
IC50: 17.86 µM
Cyclin D1 G1-phase arrest[40]
Resveratrol B65 (rat dopaminergic cell line) 24 h 25, 50, 100 µM pAkt
Cyclin D, E, A
CDK2
p53
NFκβ
S-phase arrest[41]
Resveratrol NUB-7, LAN-5, IMR-32 and SK-N-BE(2) 2–8 days 25–160 µM p53 translocation from cytoplasm to nucleus
p21WAF−1/CIP−1
G1-, G2/M-, and S-phase arrest[53]
* denotes increase of biomarker, while denotes decrease.

3.3. Mitochondrial and ER-Stress-Related Apoptotic Pathways

Results of several studies have also identified Mitochondrial and ER-Stress-Related Apoptotic Pathways, as demonstrated in Figure 4 and Table 3.

3.3.1. Bcl-2-Family Proteins

The relative amounts of mitochondrial protein subfamilies of Bcl-2 (including antiapoptotic Bcl-xL) and pro-apoptotic Bcl-2 associated X-protein (Bax) act as apoptosis triggers. Specifically, a high Bax/Bcl-2 ratio leads to cytochrome c release by the mitochondria [54]. In turn, this causes the activation of apoptotic caspases-9 and -3. Moreover, an increased Bax/Bcl-2 ratio leads to poly (ADP-ribose) polymerase (PARP) cleavage. Flavonoids apigenin, butein, DEDC, genistein, luteolin, EGC, EGCG, and quercetin increase the amount of Bax protein in NB [27,30,32,33,55,56,57,58]. Similar effects on Bax are attributed to the non-flavonoid polyphenols bisphenol-A (BPA), bisphenol-B (BPB), bisphenol-S (BPS), honokiol, and resveratrol [39,42,59]. The flavonoids apigenin, butein, carnosic acid (CA), DEDC, genistein, and rutin and the non-flavonoid polyphenols BPA, BPB, BPS, curcumin, and resveratrol decrease the amount of both Bcl-2 and Bcl-xL, contributing to both cytochrome c release and PARP cleavage in NB [27,32,36,37,39,40,55,56,59,60,61]. The findings in another study indicate that oleacein demonstrates tumor-suppressive effects against SH-SY5Y neuroblastoma cells. Oleacein blocked the S phase of the cell cycle, induced apoptosis via the upregulation of both Bax/Bcl-2 ratio and p53 expressions, and increased STAT3 phosphorylation [62]. Another antiapoptotic protein in the Bcl family IS Mcl-1, whose expression in SH-SY5Y cells is notably reduced by Nordentatin, a coumarin derivative isolated from Clausena harmandiana. A reduction in Mcl-1 concentration results in Bax activation. This leads to caspase-3 cleavage and the inhibition of the migratory protein MMP-9 via the glycogen synthase kinase-3 (GSK-3) pathway, ultimately inhibiting the proliferation and migration of NB cells [63].

3.3.2. PARP Cleavage

The proteolytic cleavage of the enzyme PARP is upregulated by caspase-3 and Bax, reducing the inhibition of DNase activity and increasing DNA fragmentation during apoptosis [64,65]. A significant impact on numerous signaling molecules was observed after the administration of isoflavone lupiwighteone in NB cells. Specifically, lupiwighteone inhibited NB-cell growth, induced G2/M phase arrest, and increased apoptosis via the caspase-dependent mitochondrial pathway; these effects were accompanied by decreased cyclin B1/D1 and CDK 1/2/4/6, the effects on MMP, increased ROS, Bax, cytochrome c, cleaved caspase-9 and -3, cleaved PARP, decreased Bcl-2, and activated Nrf2/ARE signaling [66]. Moreover, a potent anti-cancer capacity has recently been attributed to dichloromethane extract from Scrophularia orientalis L., a traditional Chinese medicinal plant, which induced cell death. At the same time, these effects were associated with aberrant calcium signaling, leading to mitochondria-related apoptosis, as demonstrated by intracellular Ca2+ release, the opening of the mitochondrial permeability transition pore, increased caspase-3, and PARP cleavage [67].

3.3.3. MMP and Cytochrome C

The mitochondrial membrane potential (MMP) generated by proton pumps on the mitochondrial membrane drives the proton gradient exploited in ATP synthesis during oxidative phosphorylation [68]. The loss of MMP imposes a loss of cell viability. Studies involving luteolin, BPS, BPA, BPS, curcumin, and honokiol found the downregulation of MMP to significantly contribute to apoptosis in NB by enhancing cytochrome C release and the release of apoptosis-inducing factor (AIF), an apoptotic protease [39,57,58,59,69]. As with ECG and EGCG, activated caspase-3 can also indirectly cleave BH3-interacting-domain-death agonist (Bid) via the activation of caspase-8, inducing Bid cleavage into truncated-Bid (tBid) and, subsequently, cytochrome c release [37,38,70]. Other studies suggest the role of tBid in the pathway, as it mobilizes cytochrome c across the outer mitochondrial membrane [71]. In NB cells treated with resveratrol, the loss of MMP associated with cytochrome c release induced Smac/Diablo release from the mitochondria, promoting caspase-9 release [61].

3.3.4. Oxidative and ER Stress

The release of cytochrome C by the mitochondria upregulates the generation of reactive oxygen species (ROS), which results in oxidative stress [59]. Oxidative stress contributes to ER stress and vice versa. In addition, ER stress inhibits protein synthesis, eventually leading to apoptosis. The flavonoids butein, CA, isoliquiritigenin, and luteolin and the non-flavonoid polyphenols BPA, BPB, and BPS directly increase the generation of ROS [26,57,59,60,72]. In the luteolin-specific pathway, ROS-associated ER stress leads to eukaryotic Initiation Factor 2α (eIF2α) phosphorylation and, subsequently, activating transcription factor 6α (ATF6α) cleavage. The ATF6α cleavage is correlated with the expression of the ER-stress-related proteins CHOP, GRP94, and GRP78 [57]. Neuroblastoma cells treated with BPA, BPB, and BPS showed elevated malondialdehyde (MDA) levels, reflecting severe oxidative damage [59]. In another study, carnosic acid (CA) showed cytotoxic potential against SH-SY5Y neuroblastoma cells. The CA suppressed methylglyoxal-induced nitrosative and oxidative stress by triggering the PI3K/Akt/Nrf2 signaling pathway. Furthermore, the CA stimulated antioxidant enzymes in neuroblastoma cells through Nrf2-transcription-factor activation [73]. Using the same cells, 3,4-dihydroxybenzalacetone or caffeic acid phenethyl ester induced oxidized-protein-mediated ER stress. In addition, both compounds increased the expression of LC3-II, an autophagy marker, and decreased 4-phenylbutyric acid, a chaperone that reduces ER stress [74]. Graham et al. showed that resveratrol elevated ER stress and the cytotoxic effects of glycolytic inhibition in neuroblastoma cell lines. Analyzing the mechanism of action, the authors described the downregulation of Akt (by increasing PP1α activity) in signaling pathways independent of SIRT1. Moreover, resveratrol initiated both caspase-3- and calpain-mediated apoptosis [75].

3.3.5. p53 and p38

The ROS activates the p38 pathway, as observed in NB cell lines treated by CA [60]. Consequently, p38 increases apoptotic signals, eventually leading to apoptosis. As observed in NB cells, quercetin and resveratrol increase p53 translocation from the cytosol to the nucleus [29]. Furthermore, p53 also played a role in honokiol-triggered autophagic apoptosis via an intrinsic mitochondrion-dependent mechanism [76]. A study investigating curcumin-mediated Bex-gene induction found a molecular association between p53 activation and Bex-gene induction. Reintroducing all the endogenous anti-cancer Bex genes directly contributes to NB-cell death via the activation of the intrinsic apoptotic pathway [65].
Table 3. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via mitochondrial apoptotic pathways and/or endoplasmic reticulum stress *.
Table 3. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via mitochondrial apoptotic pathways and/or endoplasmic reticulum stress *.
CompoundCell LineIncubation PeriodConcentration(s)Biomarker ChangesReference
Flavonoids
ApigeninSH-SY5Y24 h50 µM Bax
Bcl-2
Cytochrome c release
Caspase-3, -9
[27]
ApigeninNUB-7 and LAN-524 h10, 50, 100, 150,
200 µM
IC50: 35 μM in NUB-7
IC50: 22 μM in LAN-5
Bax
PARP cleavage
Caspase-3
[33]
Butein Neuro2a (mouse cell line)24 and 48 h6.25, 12.5, 25, 50, 100 μM
IC50: 6.25 µM, 24 h
Bax
Bcl-2
Caspase-3
PARP cleavage
ROS
[55]
CA (carnosic acid, rosemary phenolic compound)IMR-3224 h5, 10, 20, 30, 40 µM
IC50: 30 µM
Caspase-3, -9
PARP cleavage
Bcl-2
ROS
p38 activation
[60]
CASH-SY5Y1 h0.2–2 μM Nitrosative and oxidative stress
PI3K/Akt/Nrf2 signaling
Nrf2 expression
[73]
DEDCSH-SY5Y24 h7.5 µg/mL Bax
Bcl-2
[32]
3,4-dihydroxybenzalacetone and caffeic acid phenethyl esterSH-SY5Y4 and 8 h10 and 20 μM LC3-II
4-Phenylbutyric acid (chaperone)
Autophagy
[74]
EGCSH-SY5Y24 h50 µM Cytochrome c
Caspase-9
[27]
ECGCSH-SY5Y24 h50 µM Cytochrome c
Caspase-9
[27]
Genistein SK-N-DZ 24 h 10 µM Caspase-3, -9
Bax/Bcl-2 ratio
Bcl-2 with only genistein
Complete Bcl-2 knockdown with combination treatment (with Bcl-2 siRNA plasmid vector)
[56]
Genistein SH-SY5Y 24 h 100 µM Bax
Bcl-2
Cytochrome c
Caspase-3, -9
[27]
Isoliquiritigenin IMR-32 and SK-N-BE(2) 24 h 5–200 μM ROS level[72]
Luteolin Neuro-2a (mouse cell line) 24 h 1–50 µM
IC50: 10 µM
ER: CHOP, GRP94, GRP78
ATF6α cleavage
eIF2α phosphorylation
ROS
MMP
Bax
cytochrome c
[57]
Quercetin Neuro2a (mouse cell line) 24 h10, 20, 40, 80, 120 μM
IC50: 40 µM
Caspase-3, -9
p53 mRNA
Bax
Cytochrome c
[30]
Rutin LAN-5 24 h 0, 25, 50, 100 μM Bcl-2 expression
Bax/Bcl-2 ratio
[36]
Non-Flavonoid Polyphenols
Bisphenol A (BPA)
Bisphenol B (BPB)
Bisphenol S (BPS)
IMR-32 (male) and SK-N-SH (female) 24 h BPA and xBPS:
IMR-32:
1, 10, 100 nM
IMR-32 and SK-N-SH:
1, 10, 100 μM
BPB:
IMR-32:
1, 10 nM
IMR-32 and SK-N-SH:
100 nM
1, 10, 100 μM
Caspase-3
Bak1
Bax
Cytochrome c
Bcl-2
MMP
ROS
MDA
[59]
Curcumin NUB-7, LAN-5, IMR-32 and SK-N-BE(2) 2–8 days 25–160 µM p53 translocation
Bax
[42]
Curcumin GI-L-IN, HTLA-230, SH-SY5Y, LAN5, SK-NBE2c, and IMR-32 18–72 h 0.1–25 µM Cytochrome C[38]
Curcumin SK-N-SH 24 h 8, 16, 32 µM Caspase-3
ROS
PARP cleavage
p53
Bcl-2
MMP
Cytochrome C
[37]
Curcumin LAN-5 3, 5, 24 h 5, 10, 15, 20 µM Hsp60
HK-II
[77]
Curcumin Neuro-2a (mouse cell line) 24 h 10, 25, 50 µM Caspase-3, -9
ROS
PARP cleavage
p53
Bex genes
[65]
Honokiol Neuro-2a (mouse cell line) and NB41A3 72 h 2.5, 5, 10, 20, 30,
40, 50, 60, 80,
and 100 µM
LC50: 63.3 µM
Bax
Cytochrome-C
MMP
Caspase-3, -6, -9
[39]
HonokiolNeuro-2a (mouse cell line) and NB41A324, 48, 72 h50 µM p53
Cytochrome-C
Autophagy
Caspase-3
LC3-II
[76]
NordentatinSH-SY5Y24, 48, 72 h1, 10, 100 µM GSK-3 phosphorylation
Mcl-1
MMP-9
Caspase-3
[63]
OleaceinSH-SY5Y6 and 24 h10 and
25 μM
Bax/Bcl-2 ratio
p53
STAT phosphorylation
[62]
ResveratrolSK-N-AS, NGP, and SH-SY5Y48 hIC50:
SK-N-A: 70 µM/L
NGP: 120 µM/L
SH-SY5Y: 100 µM/L
Caspase-3, -9
Cytochrome-C
Smac/Diablo
Bcl-2
[61]
Resveratrol B103 (rat cell line) 48 h 5–20 µM
IC50: 17.86 µM
Bcl-2
Bcl-xL
Mcl-1
Caspase-3, -9
[40]
Resveratrol NUB-7, LAN-5, IMR-32 and SK-N-BE(2) 2–8 days 25–160 µM Bax
p53 translocation
[42]
ResveratrolK-N-SH, SH-SY5Y, SK-N-Be2, SMS-KCNR, and NB16918 h10–100 μM Cell death
Caspase-3
ER stress
Akt
PP1α
[75]
Plant extracts
Kaffir lime leaf
(contains alkaloid, flavonoid, terpenoid, tannin, and saponin compounds)
UKF-NB3, IMR-5 and SK-N-AS IC50:
UKF-NB3: 18.9 µg/mL
IMR-5: 6.4 µg/mL
SK-N-AS: 9.4 µg/mL
[78]
Juniperus communis L. Berry (contains 13 flavonoid glycosides and 2 phenolic acids) SH-SY5Y 12, 36, 48 h 10 µg/mL p53[79]
* denotes increase of biomarker, while denotes decrease.

3.4. Receptor-Mediated Apoptotic Pathway

Receptor-mediated pathways primarily trigger apoptosis and inhibit cell-cycle progression through various mechanisms, which are outlined in Figure 5 and Table 4. The mechanisms of flavonoid and non-flavonoid polyphenols’ actions match those of the previously discussed pathways.
The flavonoid DEDC decreases the expression of phospho-STAT3 in a ROS-mediated manner in NB cells [32]. The downregulation of phospho-STAT3 leads to the upregulation of p53 and p21, downregulating Cyclin B1, resulting in cell-cycle arrest and apoptosis in the G2/M phase [32].
The flavonoids genistein and rutin have similar mechanisms, leading to apoptotic effects. Rutin increases NB’s necrosis factor α (TNF-α) secretion [36]. Similarly, genistein increased TNF-α and Fas ligand (FasL), TRADD, and FADD in NB [56]. The upregulation of TNF and FasL lead to upregulated TNF receptor type 1-associated DEATH domain protein (TRADD), and Fas-associated DEATH domain protein (FADD) activates caspase-8, which cleaves Bid to tBid. Increased levels of tBid encourage conformational changes in Bax, forming oligomer channels, which promote cytochrome c release. The release of tBid activates caspase-3, leading to nuclear-DNA fragmentation and apoptosis. In addition, both studies found a decrease in antiapoptotic Bcl-2, an increase in pro-apoptotic Bax and, therefore, an overall increase in the Bax:Bcl-2 ratio, which triggers the mitochondrial pathway of apoptosis by releasing cytochrome c in NB. This mechanism is also present when EGC and EGCG increase the activation of caspase-8 and the proteolytic cleavage of Bid to tBid, and Bax oligomerization leads to apoptosis [27].
Similarly, EGC and/or EGCG showed significant effects through the induction of both mitochondria-mediated (as evidenced by increased Bax and decreased Bcl-2) and receptor-mediated apoptotic pathways (demonstrated via the activation of caspase-8 and the cleavage of Bid into tBid) in two NB-cell lines. However, the effects of the EGC and EGCG on apoptosis were accompanied by decreased oncogenic miRNAs and increased tumor-suppressor miRNAs. Furthermore, the authors concluded that the overexpression of tumor-suppressor miRNA-7-1 increases the efficacy of EGCG in apoptosis induction, suggesting the potential of this combination therapy in NB [52].
The non-flavonoid polyphenol curcumin decreases heat shock protein-60 (Hsp60) and hexokinase II (HK-II) levels and increases Bcl-2 associated agonist of cell death (Bad), phosphatase and tensin homolog (PTEN), and ROS in NB cells [77]. The mitochondrial protein Bad increases mitochondrial membrane permeability, promoting cytochrome c release and apoptosis via an intrinsic apoptotic pathway [77]. Phosphatase and tensin homolog is associated with the (PI3K)/Akt pathway—a “highly oncogenic pro-survival” signaling pathway. The PTEN antagonizes the (PI3K)/Akt pathway, leading to ROS-mediated apoptosis [77]. Honokiol, another non-flavonoid polyphenol, demonstrated upregulation of ROS, resulting in increased receptor-interacting protein kinase 3 (RIP3), a critical regulator of programmed necrosis [80].
Resveratrol showed its capacity to increase caspase-3, resulting in an inhibited cell cycle in NB cells [41]. Activated caspase-3 plays a role in cell maintenance and survival through the cleaving of multiple structural and regulatory proteins. Additionally, activated caspase-3 contributes to mitochondrial apoptotic processes, such as releasing cytochrome c and Bax translocation [81].

4. Overview of Findings

4.1. Potent Findings and Treatment Mechanisms

Currently, the approaches that utilize natural compounds, including flavonoids, in treating NB cell lines, can be characterized into four primary mechanisms of treatment: the induction of apoptosis via mitochondrial ER pathways, receptor-mediated apoptosis, calpain-driven apoptosis, and anti-proliferative mechanisms.
Calpain-dependent apoptosis is one mechanism that can be exploited in NB treatment. The flavonoids apigenin, EGC, and EGCG exert apoptotic effects at the lowest concentration (50 µM). However, the finding of calpain-driven apoptosis in NB cell lines via the application of flavonoids was presented only by Das et al. (2006) [27]. Apigenin is rich in dried parsley, with a concentration of 45,035 μg/g, while EGC and EGCG, which are both types of catechin, can be found in green tea [82]. In addition, EGC was found at a concentration of 1160.6 μg/mL, while the concentration of EGCG was 542.6 μg/mL, when green tea was brewed for 80 min at 50 °C, suggesting green tea as a potent source of flavonoids [83]. Green tea is mainly consumed in Asian countries, such as China, Japan, Korea, and India; this indicates a relationship between green-tea consumption and the relatively low frequency of NB in all of Asia, although this proposition requires further research [84,85].
In the studies reviewed here, curcumin, quercetin, and resveratrol are the most heavily researched polyphenol compounds in relation to NB treatment, and exert the most potent effects. In two studies, curcumin decreased cell growth and induced G1-, G2/M-, and S-phase arrest at concentrations of 0.1–25 µM and 5–10 µM, respectively [38,42]. Pure turmeric powder has one of the highest concentrations of curcumin, at around 3.14% by weight; thus, it is a potentially significant source of curcumin in cancer-treatment supplementation [86]. Quercetin decreased levels of Bcl-w and Cyclin-CDK at an IC50 of 40 µM; in another study, quercetin inhibited cell growth at an IC50 of 10 µM [30,31]. However, despite these promising IC50 values, which suggest high potency, the studies were conducted using mouse cell lines rather than human-NB-cell lines. Regarding natural sources, multiple berries are rich in quercetin, such as cranberries, with concentrations of 83–121 mg/kg. Furthermore boiling blueberries (1 g/10 mL) for 4 min yields a quercetin concentration of 1138.4 mg/mL [87,88]. Isoliquiritigenin, another flavonoid, is mainly found in licorice (Glycyrrhiza uralensis), at a concentration of 0.45 mg/g [89]. The anti-proliferative effect of isoliquiritigenin is also promising, with an IC50 of 25.4 µM [35]. In other studies, isoliquiritigenin had apoptotic effects on prostate cancer cells, melanoma, leukemia, ovarian cancer, lung cancer, and colon cancer [90].
Another notable anti-NB mechanism is the regulation of p53 expression. Most of the papers reviewed showed an increase in the tumor-suppressor gene, resulting in cell death. However, a study conducted by Ye et al. (2021) demonstrated that treatment with curcumin decreased p53 levels but increased NB apoptosis [37]. This suggests an apoptosis mechanism that is not p53-dependent, or the possibility of p53 exerting dual effects [91].
The most studied form of anti-NB mechanism was the induction of apoptosis via ER and/or mitochondria-related apoptotic mechanisms, including, but not limited to, cytochrome c release, Bax/Bcl-2, ROS generation, and p53 expression. Butein induced apoptosis via increased Bax/Bcl-2 ratio, PARP cleavage, and ROS at a low IC50 of 6.5 µM [55]. Luteolin also demonstrated notable apoptotic effects, at an IC50 of 10 µM; it increased cytochrome c release, Bax/Bcl-2, ER-stress proteins, ATF6α cleavage, and eIF2α phosphorylation [57]. Onion leaves are among the many naturally occurring sources of luteolin, with a measured content level of 391.0 mg/kg, making them excellent candidates for supplementary NB therapies [92]. Other naturally occurring sources abundant in luteolin include carrots, parsley, apple skin, and broccoli [93].
Many other flavonoids also induced apoptosis, although the studies on butein and luteolin reported the lowest IC50 values for flavonoids exerting apoptotic effects via mitochondrial/ER pathways. One of the more widely studied compounds is curcumin: two studies reported curcumin’s involvement in mitochondrial/ER pathways to induce apoptosis [37,38]. Three studies using resveratrol to treat NB cell lines reported apoptosis via mitochondrial/ER pathways [40,42,61]. Rahman et al. (2012) reported the lowest IC50 (17.86 µM) of these three studies. Notably, this result was obtained by treating a rat cell line rather than a human cell line [40].
Honokiol, a constituent of the traditional Chinese medicinal plant, Magnolia officinalis, has been shown to pass through the blood–brain barrier (BBB) without affecting normal brain cells [39,94]. Studies have demonstrated its induction of autophagic apoptosis in NB cells via an intrinsic mitochondrion-dependent pathway and its involvement in the ROS-mediated upregulation of RIP3 protein [39,76,80]. With a growing body of research to support its potential, honokiol is a significant candidate for use as therapy for high-risk NB.
Apoptosis through receptor-mediated pathways involves specific receptors to induce an apoptotic cascade. Of the studies found, none mentioned a specific IC50 value. Curcumin, EGC, and EGCG are polyphenol compounds demonstrating notable apoptotic effects via receptor-mediated pathways. The main apoptotic mechanisms reported were increased TNF-α, caspase-8 activation, tBid generation, adaptor protein FADD, and Bax oligomerization [27,77]. As in the studies reporting apoptosis via mitochondrial/ER pathways, the Bax/Bcl-2 ratio is also prominent in receptor-mediated apoptosis. Many receptor-mediated apoptotic pathways depend on mitochondrial apoptotic pathways, further underlining the vital role of the mitochondria in the apoptosis of treated NB lines. Moreover, the prevalence of Bax and Bcl-2 as biomarkers was expected, as the pro-apoptotic protein Bax commits the cell to a mitochondrial suicide pathway and is considered one of the main targets of anti-cancer interventions [95].

4.2. Plant Extracts

Plant extracts often contain polyphenols and are investigated for their anti-cancer properties. A paper by Tunjung et al. (2015) investigated Kaffir lime leaf (which contains alkaloid, flavonoid, terpenoid, tannin, and saponin compounds) against three NB cell lines. The IC50 values were as follows: UKF-NB3 cell line, 18.9 µg/mL; IMR-5 cell lines, 6.4 µg/mL; and SK-N-AS cell lines, 9.4 µg/mL [78]. Another paper, by Lantto et al. (2016), investigated the extract of Juniperus communis L. Berry (containing 13 flavonoid glycosides, 2 phenolic acids) by treating the NB cell line SH-SY5Y with 10 µg/mL. The extract increased the expression of the tumor-suppressor gene p53 [79]. More studies are needed to determine whether combinations of polyphenols (as seen in plant extracts) work synergistically against NB, or whether these summations have no significant effects.

4.3. In Vivo Studies Involving NB

Although the IC50 values of polyphenols demonstrate considerable potency, a vehicle for effective flavonoid delivery remains unclear. Firstly, it is not feasible to translate in vitro concentrations into dosages for clinical use. Compared to in vitro studies, fewer in vivo studies demonstrate polyphenols’ anti-NB abilities. However, researchers investigating compounds in live mouse models have demonstrated noteworthy results (Table 5). These studies consider three alternate flavonoid-delivery methods: oral, intraperitoneal, or peritumor injection. Resveratrol is the most frequently studied polyphenol, with three papers supporting its in vivo efficacy [61,96,97]. All three of the proposed delivery methods proved effective; one study, which utilized oral delivery, found an 80% decrease in tumor volume, despite the low bioavailability of resveratrol [61]. The five studies outlined in Table 5 highlight an area that needs future study; data from mice models can aid the clinical implementation of natural compounds in the treatment of NB.

5. Key Considerations and Challenges

5.1. Limitations of Polyphenol Use

Polyphenols have the potential to exert adverse effects in certain circumstances. Polyphenols’ toxicity in normal cells involves their pro-oxidant behavior in the presence of redox-active metals, which is unusual given their antioxidant activity. Pro-oxidant polyphenols have a negative effect on DNA, proteins, and lipids. Toxicity can also form tumors in normal cells, where a few phenolic acids, which are examples of polyphenols, have tumor-inducing properties [98].
The ingestion of large doses of polyphenols is associated with carcinogenesis, thyroid toxicity, and the disruption of the bioavailability of pharmaceuticals. Increased polyphenol levels have also been linked to increased kidney tumors in mice and rats [99]. Notably, the inability to mimic in vivo conditions in in vitro studies limits our understanding of polyphenol toxicity in human health [20].

5.2. Insights into Flavonoid-Delivery Mechanisms

A physical mechanism of polyphenol delivery is difficult to achieve. Specifically, the delivery of flavonoids is associated with several limitations due to their low water solubility and low bioavailability [100]. Therefore, many delivery methods have been used and tested to increase flavonoids’ bioavailability when they are consumed orally. Two prominent strategies explored in the literature are nanotechnology and encapsulation methods, as they have shown the ability to overcome barriers to flavonoid delivery. Nanotechnological strategies encompass a wide variety of therapeutic delivery methods. Much of the research into nano-based delivery explores ways of increasing herbal drugs’ bioavailability and protection against gastrointestinal (GI) deterioration. Nano-structural (such as protein, carbohydrate and/or lipid-based carriers) and nanoparticle delivery systems for flavonoids have presented increased bioavailability due to their ability to prevent GI degradation, increasing flavonoids’ absorption rate into the bloodstream [101,102].
Additionally, nanocrystal methods are used, as the small size of the particle or compound increases its dissolution rate (effects vary based on the surface area available for interactions with their target), increasing the bioavailability of compounds upon delivery [101]. The nano-micellar delivery of flavonoids has also been studied for its enhanced bioavailability properties. These micelles can be developed via the self-assembly of amphiphilic molecules in an appropriate liquid medium to improve and maintain targeted delivery.
Encapsulation methods involve trapping a compound of interest (in this paper, flavonoids) in sealed miniature nano-capsules. They have been studied in the literature for their ability to improve flavonoid distribution, their solubility, and their stability in the body—as with nanotechnological strategies—by protecting the compound from oxidation and degradation upon oral consumption [101].
The presence of the BBB also affects the bioavailability of flavonoids. It plays an essential role in protecting the brain through its selective permeability of substances entering the brain—such as essential nutrients and hormones—and elimination of toxins [103].
The current literature on flavonoids and the BBB suggests that the permeability of a flavonoid depends on the compound’s properties and how they interact with the properties of the BBB. Lipid solubility is one of the primary properties of flavonoids that affect their permeability into the BBB [104,105]. For the compound to cross the BBB, there are two primary transport mechanisms: passive diffusion (as studies have found that lipophilic flavonoids demonstrate increased transportation rates with increased concentrations) and efflux transporters [105]. Efflux transporters, such as P-glycoprotein, play a role in controlling the transport of xenobiotic agents. Research suggests that efflux transporters may play a role in the permeability rates of flavonoids by limiting their bioavailability and distribution across the BBB [106].
One study evaluating the transport of flavonoids across the BBB found that the flavonoids genistein and isoliquiritigenin had higher permeation than rutin and quercetin [105]. Notably, it also found that combining a flavonoid with weaker permeability, such as rutin, with verapamil (a P-glycoprotein inhibitor) can help to enhance its permeability across the BBB.
Zhenzhu et al. (2022) underline the potential for extracellular vesicles (EVs) to penetrate brain tissue and the BBB, specifically for treating neurodegenerative (ND) diseases. Extracellular vesicles are naturally produced by many cell types and include subtypes such as exosomes, microvesicles, apoptotic bodies, and oncosomes, all ranging in size and application [107]. Extracellular vesicles have been studied in the delivery of polyphenols to treat neurodegenerative diseases. The flavonoids studied include curcumin, quercetin, and resveratrol [108,109,110].

5.3. Clinical Trials Investigating Therapeutic Use of Polyphenols

While conducting searches for clinical-trial data across databases such as ClinicalTrials.gov and the Cochrane Central Register of Controlled Trials (CENTRAL), relatively very few clinical studies on neuroblastoma treatments were found, and none of these investigate polyphenols as potential treatments, despite promising in vitro research, as well as clinical trials on other types of cancer. The majority of the completed clinical studies regarding interventional treatments for neuroblastoma feature limitations in their characteristics, as well as presenting serious and non-serious adverse effects on their participants [111,112,113,114,115]. This demonstrates the need to explore and investigate other treatment options, among which polyphenols have been proposed based on current in vitro research, as well as clinical studies on the effects of polyphenols on other types of cancer.
Upon expanding the search from “neuroblastoma” to “cancer” and polyphenols on clinical-trial databases, a handful of clinical trials with published data were found. The most common cancers studied were prostate, breast, oral, and colon cancers. Of the polyphenols discussed in this paper, curcumin, EGCG, resveratrol, quercetin, and apigenin have been submitted to 71, 39, 17, 14, 1, and 0 clinical trials, respectively [21]. For this section, 13 completed and published clinical studies were selected from the previously mentioned clinical databases.
Overall, many of these clinical trials presented promising results that suggested the potential of polyphenols as cancer treatments, with the outcomes outlined in Table 6. Safety and tolerability were demonstrated across these studies [21], with most reporting no adverse effects of the polyphenols tested on their participants. The trial results were found to be positively correlated with the results of in vitro studies—however, the results should be interpreted with caution due to the limited statistically significant results and study characteristics.
Research regarding childhood cancer and polyphenols is rarely performed. One dissertation featured a pilot study, entitled “Purple Grape Juice in Improving Vascular Health in Childhood Cancer Survivors”, with 24 participants, to evaluate whether purple grape juice can reduce oxidative stress and improve the vascular health of survivors of childhood cancer who are in early stages of cardiovascular disease [128]. The results showed no improvements in endothelial function and no significant improvements in oxidative stress and inflammation biomarkers.
Across the clinical studies discussed, many limitations arose from the study characteristics and methodologies, which included:
  • Non-blinded studies [116,117,122].
  • The effects of physiological parameters on the results [117,121], including varying rates of disease progression in sample populations [126].
  • Small sample sizes [117,119,120,121,122,123,124,125,126,127,128].
  • Potential measurement errors [123], or invalidated assays [119].
  • Low bioavailability due to the poor absorption of polyphenols [125,127].
  • The short duration of trials [118,121,122,123,124,126,128].
  • Reliance on a few select endpoints without other indicators of disease progression [118].
Despite the limitations in the currently available data, polyphenols are still under consideration as candidates for cancer treatment due to their potential as less toxic treatments [21]. Of the clinical trials discussed, a few of those that presented significant results involved trials with strong study characteristics, primarily randomization, placebo control, and double masking [118,121], suggesting potential for future studies and clinical applications. Therefore, the future of cancer and polyphenol research will need to focus on improving study characteristics, testing different methods of delivering polyphenols, and expanding to other types of cancer to properly assess polyphenols’ anti-cancer properties and how to best deliver them to patients. Furthermore, these clinical studies, alongside in vitro studies, support the investigation of the application of polyphenols as treatments for neuroblastoma.

6. Conclusions

Polyphenols are natural compounds synthesized by plants, and they can be divided into flavonoids and non-flavonoids. Polyphenols have shown promising results in both in vitro and in vivo studies of their use against neuroblastoma, a cancer that predominantly affects children. The anti-proliferative effects of polyphenols include arresting the cell cycle, decreasing bcl-w and cyclin-CDK levels, and increasing p53 levels. Polyphenols can also affect apoptotic mechanisms, including apoptosis, via mitochondrial ER pathways, through which polyphenols can promote cytochrome c release and increase the Bax/Bcl-2 ratio and ROS levels. Another apoptotic mechanism that polyphenols utilize is receptor-mediated apoptosis, which is dependent on the mitochondrial apoptotic pathway and involves TNF-α and caspase-8 activation and Bax oligomerization. Calpain-driven apoptosis was only reported by a single paper, making it a potential topic for future research. The use of flavonoid delivery, including a solution to the question of how to increase its bioavailability orally and across the blood–brain barrier, as the physical mechanism of delivery, is difficult to achieve. There is inadequate in vivo evidence to suggest that in vitro success would translate into clinical studies, and all the apoptotic mechanisms should be researched further, despite the encouraging results so far.

Author Contributions

Conceptualization, A.K. and Z.S.; resources, D.B.; writing—original draft preparation, A.K., Z.S., R.S.I. and R.A.-J.; writing—review and editing, D.B., A.K., A.M., P.K. and Z.S.; visualization, A.K. and Z.S.; 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.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The publication costs of this article were funded by Qatar Foundation through the Weill Cornell Medicine-Qatar Biomedical Research Program.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AktSerine–threonine kinase
AIFApoptosis-inducing factor
ATFActivating transcription factor
BaxBcl-2 associated X-protein
BadBcl-2 associated agonist of cell death
BBBBlood–brain barrier
BclB-cell lymphoma
BidBH3 interacting-domain death agonist
BPBisphenol
Ca2+Calcium ion
CADCaspase-activated DNase
CDKCyclin-dependent kinase
CHOPC/EBP homologous protein
DEDC2-(cis-1,2-dihydroxy-4-oxo-cyclohex-5-enyl)-5,7-dihydroxy-chromone
EGCGEpigallocatechin gallate
EGCEpigallocatechin
eIFEukaryotic initiation factor
EREndoplasmic reticulum
ERKExtracellular signal-regulated kinase
FADDFas-associated DEATH domain protein
FasLFas ligand
GIGastrointestinal
GRPGlucose-regulated protein
GSKGlycogen synthase kinase
HSPHeat-shock protein
JNKc-Jun N-terminal kinase
MAPKMitogen activated protein kinase
MDAMalondialdehyde
MMPMitochondrial membrane potential
NBNeuroblastoma
NF-κBNuclear Factor κappa of B cells
PARPPoly (ADP-Ribose) polymerase
PI3KPhosphoinositide 3-kinases
PTENPhosphatase and tensin homolog
RIPReceptor-interacting protein
RKIPRaf-1 kinase inhibitor protein
ROSReactive oxygen species
tBidTruncated Bid
TNFTumor necrosis factor
TRADDTumor necrosis factor receptor type 1-associated DEATH domain

References

  1. Johnsen, J.I.; Dyberg, C.; Wickström, M. Neuroblastoma—A Neural Crest Derived Embryonal Malignancy. Front. Mol. Neurosci. 2019, 12, 9. [Google Scholar] [CrossRef]
  2. Finklestein, J.Z.; Gilchrist, G.S. Recent Advances in Neuroblastoma. N. Engl. J. Med. 2010, 362, 116–127. [Google Scholar] [CrossRef] [Green Version]
  3. Smith, V.; Foster, J. High-Risk Neuroblastoma Treatment Review. Children 2018, 5, 114. [Google Scholar] [CrossRef] [Green Version]
  4. Ducassou, A.; Gambart, M.; Munzer, C.; Padovani, L.; Carrie, C.; Haas-Kogan, D.; Bernier-Chastagner, V.; Demoor, C.; Claude, L.; Helfre, S.; et al. Long-Term Side Effects of Radiotherapy for Pediatric Localized Neuroblastoma: Results from Clinical Trials NB90 and NB94. Strahlenther. Onkol. 2015, 191, 604–612. [Google Scholar] [CrossRef] [PubMed]
  5. Keshelava, N.; Seeger, R.C.; Reynolds, C.P. Drug Resistance in Human Neuroblastoma Cell Lines Correlates with Clinical Therapy. Eur. J. Cancer 1997, 33, 2002–2006. [Google Scholar] [CrossRef]
  6. Liskova, A.; Koklesova, L.; Samec, M.; Smejkal, K.; Samuel, S.M.; Varghese, E.; Abotaleb, M.; Biringer, K.; Kudela, E.; Danko, J.; et al. Flavonoids in Cancer Metastasis. Cancers 2020, 12, 1498. [Google Scholar] [CrossRef] [PubMed]
  7. Zhai, K.; Mazurakova, A.; Koklesova, L.; Kubatka, P.; Büsselberg, D. Flavonoids Synergistically Enhance the Anti-Glioblastoma Effects of Chemotherapeutic Drugs. Biomolecules 2021, 11, 1841. [Google Scholar] [CrossRef] [PubMed]
  8. Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef] [Green Version]
  9. Liskova, A.; Stefanicka, P.; Samec, M.; Smejkal, K.; Zubor, P.; Bielik, T.; Biskupska-Bodova, K.; Kwon, T.K.; Danko, J.; Büsselberg, D.; et al. Dietary Phytochemicals as the Potential Protectors against Carcinogenesis and Their Role in Cancer Chemoprevention. Clin. Exp. Med. 2020, 20, 173–190. [Google Scholar] [CrossRef]
  10. Niedzwiecki, A.; Roomi, M.W.; Kalinovsky, T.; Rath, M. Anti-cancer Efficacy of Polyphenols and Their Combinations. Nutrients 2016, 8, 552. [Google Scholar] [CrossRef] [Green Version]
  11. Fantini, M.; Benvenuto, M.; Masuelli, L.; Frajese, G.V.; Tresoldi, I.; Modesti, A.; Bei, R. In Vitro and in Vivo Antitumoral Effects of Combinations of Polyphenols, or Polyphenols and Anti-cancer Drugs: Perspectives on Cancer Treatment. Int. J. Mol. Sci. 2015, 16, 9236–9282. [Google Scholar] [CrossRef] [Green Version]
  12. Fresco, P.; Borges, F.; Diniz, C.; Marques, M.P.M. New Insights on the Anti-cancer Properties of Dietary Polyphenols. Med. Res. Rev. 2006, 26, 747–766. [Google Scholar] [CrossRef] [Green Version]
  13. Bhosale, P.B.; Ha, S.E.; Vetrivel, P.; Kim, H.H.; Kim, S.M.; Kim, G.S. Functions of Polyphenols and Its Anti-cancer Properties in Biomedical Research: A Narrative Review. Transl. Cancer Res. 2020, 9, 7619–7631. [Google Scholar] [CrossRef]
  14. Abbas, M.; Saeed, F.; Anjum, F.M.; Afzaal, M.; Tufail, T.; Bashir, M.S.; Ishtiaq, A.; Hussain, S.; Suleria, H.A.R. Natural Polyphenols: An Overview. Int. J. Food Prop. 2016, 20, 1689–1699. [Google Scholar] [CrossRef] [Green Version]
  15. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of Dietary Polyphenols: The Role of Metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef] [PubMed]
  16. Fujiki, H.; Sueoka, E.; Watanabe, T.; Suganuma, M. Primary Cancer Prevention by Green Tea, and Tertiary Cancer Prevention by the Combination of Green Tea Catechins and Anti-cancer Compounds. J. Cancer Prev. 2015, 20, 1–4. [Google Scholar] [CrossRef] [PubMed]
  17. Noguchi-Shinohara, M.; Yuki, S.; Dohmoto, C.; Ikeda, Y.; Samuraki, M.; Iwasa, K.; Yokogawa, M.; Asai, K.; Komai, K.; Nakamura, H.; et al. Consumption of Green Tea, but Not Black Tea or Coffee, Is Associated with Reduced Risk of Cognitive Decline. PLoS ONE 2014, 9, e96013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. García-Lafuente, A.; Guillamón, E.; Villares, A.; Rostagno, M.A.; Martínez, J.A. Flavonoids as Anti-Inflammatory Agents: Implications in Cancer and Cardiovascular Disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef]
  19. Watanabe, N.; Ikeda, U. Matrix Metalloproteinases and Atherosclerosis. Curr. Atheroscler. Rep. 2004, 6, 112–120. [Google Scholar] [CrossRef]
  20. Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front. Nutr. 2018, 5, 87. [Google Scholar] [CrossRef] [Green Version]
  21. Cháirez-Ramírez, M.H.; de la Cruz-López, K.G.; García-Carrancá, A. Polyphenols as Antitumor Agents Targeting Key Players in Cancer-Driving Signaling Pathways. Front. Pharm. 2021, 12, 710304. [Google Scholar] [CrossRef] [PubMed]
  22. González, R.; Ballester, I.; López-Posadas, R.; Suárez, M.D.; Zarzuelo, A.; Martínez-Augustin, O.; Sánchez de Medina, F. Effects of Flavonoids and Other Polyphenols on Inflammation. Crit. Rev. Food Sci. Nutr. 2011, 51, 331–362. [Google Scholar] [CrossRef] [PubMed]
  23. Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S.; Giovinazzo, G.; Gerardi, C.; Mosca, L. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  24. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anti-cancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [Green Version]
  25. Momeni, H.R. Role of Calpain in Apoptosis. Cell J. (Yakhteh) 2011, 13, 65. [Google Scholar]
  26. Martinez, J.A.; Zhang, Z.; Svetlov, S.I.; Hayes, R.L.; Wang, K.K.; Larner, S.F. Calpain and Caspase Processing of Caspase-12 Contribute to the ER Stress-Induced Cell Death Pathway in Differentiated PC12 Cells. Apoptosis 2010, 15, 1480–1493. [Google Scholar] [CrossRef] [PubMed]
  27. Das, A.; Banik, N.L.; Ray, S.K. Mechanism of Apoptosis with the Involvement of Calpain and Caspase Cascades in Human Malignant Neuroblastoma SH-SY5Y Cells Exposed to Flavonoids. Int. J. Cancer 2006, 119, 2575–2585. [Google Scholar] [CrossRef]
  28. Ray, S.K.; Fidan, M.; Nowak, M.W.; Wilford, G.G.; Hogan, E.L.; Banik, N.L. Oxidative Stress and Ca2+ Influx Upregulate Calpain and Induce Apoptosis in PC12 Cells. Brain Res. 2000, 852, 326–334. [Google Scholar] [CrossRef]
  29. Sergeev, I.N. Genistein Induces Ca2+-Mediated, Calpain/Caspase-12-Dependent Apoptosis in Breast Cancer Cells. Biochem. Biophys. Res. Commun. 2004, 321, 462–467. [Google Scholar] [CrossRef]
  30. Sugantha Priya, E.; Selvakumar, K.; Bavithra, S.; Elumalai, P.; Arunkumar, R.; Raja Singh, P.; Brindha Mercy, A.; Arunakaran, J. Anti-Cancer Activity of Quercetin in Neuroblastoma: An in Vitro Approach. Neurol. Sci. 2014, 35, 163–170. [Google Scholar] [CrossRef]
  31. Rouzaire-Dubois, B.; Gérard, V.; Dubois, J.M. Involvement of K+ Channels in the Quercetin-Induced Inhibition of Neuroblastoma Cell Growth. Pflügers Archiv. 1993, 423, 202–205. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, H.; Jiang, C.; Xiong, C.; Ruan, J. DEDC, a New Flavonoid Induces Apoptosis via a ROS-Dependent Mechanism in Human Neuroblastoma SH-SY5Y Cells. Toxicol. Vitr. 2012, 26, 16–23. [Google Scholar] [CrossRef]
  33. Torkin, R.; Lavoie, J.F.; Kaplan, D.R.; Yeger, H. Induction of Caspase-Dependent, P53-Mediated Apoptosis by Apigenin in Human Neuroblastoma. Mol. Cancer 2005, 4, 1–11. [Google Scholar] [CrossRef]
  34. Singhal, J.; Nagaprashantha, L.D.; Vatsyayan, R.; Ashutosh; Awasthi, S.; Singhal, S.S. Didymin Induces Apoptosis by Inhibiting N-Myc and Upregulating RKIP in Neuroblastoma. Cancer Prev. Res. 2012, 5, 473–483. [Google Scholar] [CrossRef] [Green Version]
  35. Escobar, S.J.d.M.; Fong, G.M.; Winnischofer, S.M.B.; Simone, M.; Munoz, L.; Dennis, J.M.; Rocha, M.E.M.; Witting, P.K. Anti-Proliferative and Cytotoxic Activities of the Flavonoid Isoliquiritigenin in the Human Neuroblastoma Cell Line SH-SY5Y. Chem. Biol. Interact. 2019, 299, 77–87. [Google Scholar] [CrossRef]
  36. Chen, H.; Miao, Q.; Geng, M.; Liu, J.; Hu, Y.; Tian, L.; Pan, J.; Yang, Y. Anti-Tumor Effect of Rutin on Human Neuroblastoma Cell Lines through Inducing G2/M Cell Cycle Arrest and Promoting Apoptosis. Sci. World J. 2013, 2013, 269165. [Google Scholar] [CrossRef] [Green Version]
  37. Ye, Z.; Chen, D.; Zheng, R.; Chen, H.; Xu, T.; Wang, C.; Zhu, S.; Gao, X.; Zhang, J.; Li, D.; et al. Curcumin Induced G2/M Cycle Arrest in SK-N-SH Neuroblastoma Cells through the ROS-Mediated P53 Signaling Pathway. J. Food Biochem. 2021, 45, e13888. [Google Scholar] [CrossRef]
  38. Pisano, M.; Pagnan, G.; Dettori, M.A.; Cossu, S.; Caffa, I.; Sassu, I.; Emionite, L.; Fabbri, D.; Cilli, M.; Pastorino, F.; et al. Enhanced Anti-Tumor Activity of a New Curcumin-Related Compound against Melanoma and Neuroblastoma Cells. Mol. Cancer 2010, 9, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Lin, J.W.; Chen, J.T.; Hong, C.Y.; Lin, Y.L.; Wang, K.T.; Yao, C.J.; Lai, G.M.; Chen, R.M. Honokiol Traverses the Blood-Brain Barrier and Induces Apoptosis of Neuroblastoma Cells via an Intrinsic Bax-Mitochondrion-Cytochrome c-Caspase Protease Pathway. Neuro Oncol. 2012, 14, 302–314. [Google Scholar] [CrossRef] [Green Version]
  40. Rahman, M.A.; Kim, N.H.; Kim, S.H.; Oh, S.M.; Huh, S.O. Anti-proliferative and Cytotoxic Effects of Resveratrol in Mitochondria-Mediated Apoptosis in Rat B103 Neuroblastoma Cells. Korean J. Physiol. Pharm. 2012, 16, 321–326. [Google Scholar] [CrossRef] [PubMed]
  41. Pizarro, J.G.; Verdaguer, E.; Ancrenaz, V.; Junyent, F.; Sureda, F.; Pallàs, M.; Folch, J.; Camins, A. Resveratrol Inhibits Proliferation and Promotes Apoptosis of Neuroblastoma Cells: Role of Sirtuin 1. Neurochem. Res. 2011, 36, 187–194. [Google Scholar] [CrossRef] [PubMed]
  42. Liontas, A.; Yeger, H. Curcumin and Resveratrol Induce Apoptosis and Nuclear Translocation and Activation of P53 in Human Neuroblastoma. Anticancer Res. 2004, 24, 987. [Google Scholar]
  43. Sargolzaei, J.; Sadeghian, H.; Golahmadi, S.; Soukhtanloo, M. Cytotoxic Effects of Hydroxy Coumarin Derivations to Neuroblastoma N2a Cells. Iran. J. Pharm. Sci. 2020, 16, 95–106. [Google Scholar] [CrossRef]
  44. Kubatka, P.; Kapinová, A.; Kello, M.; Kruzliak, P.; Kajo, K.; Výbohová, D.; Mahmood, S.; Murin, R.; Viera, T.; Mojžiš, J.; et al. Fruit Peel Polyphenols Demonstrate Substantial Anti-Tumour Effects in the Model of Breast Cancer. Eur. J. Nutr. 2016, 55, 955–965. [Google Scholar] [CrossRef]
  45. Kapinova, A.; Stefanicka, P.; Kubatka, P.; Zubor, P.; Uramova, S.; Kello, M.; Mojzis, J.; Blahutova, D.; Qaradakhi, T.; Zulli, A.; et al. Are Plant-Based Functional Foods Better Choice against Cancer than Single Phytochemicals? A Critical Review of Current Breast Cancer Research. Biomed. Pharmacother. 2017, 96, 1465–1477. [Google Scholar] [CrossRef]
  46. Hosseini, A.; Ghorbani, A. Cancer Therapy with Phytochemicals: Evidence from Clinical Studies. Avicenna J. Phytomed. 2015, 5, 84. [Google Scholar]
  47. Kubatka, P.; Kello, M.; Kajo, K.; Samec, M.; Liskova, A.; Jasek, K.; Koklesova, L.; Kuruc, T.; Adamkov, M.; Smejkal, K.; et al. Rhus Coriaria L. (Sumac) Demonstrates Oncostatic Activity in the Therapeutic and Preventive Model of Breast Carcinoma. Int. J. Mol. Sci. 2021, 22, 183. [Google Scholar] [CrossRef] [PubMed]
  48. Morandi, F.; Bensa, V.; Calarco, E.; Pastorino, F.; Perri, P.; Corrias, M.V.; Ponzoni, M.; Brignole, C. The Olive Leaves Extract Has Anti-Tumor Effects against Neuroblastoma through Inhibition of Cell Proliferation and Induction of Apoptosis. Nutrients 2021, 13, 2178. [Google Scholar] [CrossRef]
  49. Ahrens, S.; Appl, B.; Trochimiuk, M.; Dücker, C.; Feixas Serra, G.; Oliver Grau, A.; Reinshagen, K.; Pagerols Raluy, L. Kigelia Africana Inhibits Proliferation and Induces Cell Death in Stage 4 Neuroblastoma Cell Lines. Biomed. Pharmacother. 2022, 154, 113584. [Google Scholar] [CrossRef]
  50. Roomi, M.W.; Kalinovsky, T.; Roomi, N.W.; Niedzwiecki, A.; Rath, M. Inhibition of the SK-N-MC Human Neuroblastoma Cell Line in Vivo and in Vitro by a Novel Nutrient Mixture. Oncol. Rep. 2013, 29, 1714–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Mazurakova, A.; Samec, M.; Koklesova, L.; Biringer, K.; Kudela, E.; Al-Ishaq, R.K.; Pec, M.; Giordano, F.A.; Büsselberg, D.; Kubatka, P.; et al. Anti-Prostate Cancer Protection and Therapy in the Framework of Predictive, Preventive and Personalised Medicine—Comprehensive Effects of Phytochemicals in Primary, Secondary and Tertiary Care. EPMA J. 2022, 13, 461–486. [Google Scholar] [CrossRef]
  52. Chakrabarti, M.; Ai, W.; Banik, N.L.; Ray, S.K. Overexpression of MiR-7-1 Increases Efficacy of Green Tea Polyphenols for Induction of Apoptosis in Human Malignant Neuroblastoma SH-SY5Y and SK-N-DZ Cells. Neurochem. Res. 2013, 38, 420–432. [Google Scholar] [CrossRef]
  53. ISSN 1791-7530 (Online)|Anti-cancer Research|The ISSN Portal. Available online: https://portal.issn.org/resource/ISSN/1791-7530 (accessed on 9 November 2022).
  54. Wang, Q.; Zhang, L.; Yuan, X.; Ou, Y.; Zhu, X.; Cheng, Z.; Zhang, P.; Wu, X.; Meng, Y.; Zhang, L. The Relationship between the Bcl-2/Bax Proteins and the Mitochondria-Mediated Apoptosis Pathway in the Differentiation of Adipose-Derived Stromal Cells into Neurons. PLoS ONE 2016, 11, e0163327. [Google Scholar] [CrossRef] [Green Version]
  55. Chen, Y.H.; Yeh, C.W.; Lo, H.C.; Su, S.L.; Hseu, Y.C.; Hsu, L.S. Generation of Reactive Oxygen Species Mediates Butein-Induced Apoptosis in Neuroblastoma Cells. Oncol. Rep. 2012, 27, 1233–1237. [Google Scholar] [CrossRef] [Green Version]
  56. George, J.; Banik, N.L.; Ray, S.K. Genistein Induces Receptor and Mitochondrial Pathways and Increases Apoptosis during BCL-2 Knockdown in Human Malignant Neuroblastoma SK-N-DZ Cells. J. Neurosci. Res. 2010, 88, 877–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Choi, A.Y.; Choi, J.H.; Yoon, H.; Hwang, K.Y.; Noh, M.H.; Choe, W.; Yoon, K.S.; Ha, J.; Yeo, E.J.; Kang, I. Luteolin Induces Apoptosis through Endoplasmic Reticulum Stress and Mitochondrial Dysfunction in Neuro-2a Mouse Neuroblastoma Cells. Eur. J. Pharm. 2011, 668, 115–126. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, F.; Gao, F.; Pan, S.; Zhao, S.; Xue, Y. Luteolin Induces Apoptosis, G0/G1 Cell Cycle Growth Arrest and Mitochondrial Membrane Potential Loss in Neuroblastoma Brain Tumor Cells. Drug Res. 2015, 65, 91–95. [Google Scholar] [CrossRef]
  59. Wang, C.; He, J.; Xu, T.; Han, H.; Zhu, Z.; Meng, L.; Pang, Q.; Fan, R. Bisphenol A(BPA), BPS and BPB-Induced Oxidative Stress and Apoptosis Mediated by Mitochondria in Human Neuroblastoma Cell Lines. Ecotoxicol. Environ. Saf. 2021, 207, 111299. [Google Scholar] [CrossRef]
  60. Tsai, C.W.; Lin, C.Y.; Lin, H.H.; Chen, J.H. Carnosic Acid, a Rosemary Phenolic Compound, Induces Apoptosis through Reactive Oxygen Species-Mediated P38 Activation in Human Neuroblastoma IMR-32 Cells. Neurochem. Res. 2011, 36, 2442–2451. [Google Scholar] [CrossRef] [PubMed]
  61. Van Ginkel, P.R.; Sareen, D.; Subramanian, L.; Walker, Q.; Darjatmoko, S.R.; Lindstrom, M.J.; Kulkarni, A.; Albert, D.M.; Polans, A.S. Resveratrol Inhibits Tumor Growth of Human Neuroblastoma and Mediates Apoptosis by Directly Targeting Mitochondria. Clin. Cancer Res. 2007, 13, 5162–5169. [Google Scholar] [CrossRef] [Green Version]
  62. Cirmi, S.; Celano, M.; Lombardo, G.E.; Maggisano, V.; Procopio, A.; Russo, D.; Navarra, M. Oleacein Inhibits STAT3, Activates the Apoptotic Machinery, and Exerts Anti-Metastatic Effects in the SH-SY5Y Human Neuroblastoma Cells. Food Funct. 2020, 11, 3271–3279. [Google Scholar] [CrossRef] [PubMed]
  63. Boonyarat, C.; Boonput, P.; Tongloh, N.; Kaewamatawong, R.; Chaiwiwatrakul, S.; Yenjai, C.; Waiwut, P. Nordentatin Inhibits Neuroblastoma Cell Proliferation and Migration through Regulation of GSK-3 Pathway. Curr. Issues Mol. Biol. 2022, 44, 1062–1074. [Google Scholar] [CrossRef] [PubMed]
  64. Boulares, A.H.; Yakovlev, A.G.; Ivanova, V.; Stoica, B.A.; Wang, G.; Iyer, S.; Smulson, M. Role of Poly(ADP-Ribose) Polymerase (PARP) Cleavage in Apoptosis. J. Biol. Chem. 1999, 274, 22932–22940. [Google Scholar] [CrossRef] [Green Version]
  65. Sidhar, H.; Giri, R.K. Induction of Bex Genes by Curcumin Is Associated with Apoptosis and Activation of P53 in N2a Neuroblastoma Cells. Sci. Rep. 2017, 7, 41420. [Google Scholar] [CrossRef] [Green Version]
  66. Ren, J.; Yang, J.; Xu, Y.; Huang, Q.; Yang, M.; Hu, K. Lupiwighteone Induces Cell Cycle Arrest and Apoptosis and Activates the Nrf2/ARE Pathway in Human Neuroblastoma Cells. Biomed. Pharmacother. 2015, 69, 153–161. [Google Scholar] [CrossRef] [PubMed]
  67. Lange, I.; Moschny, J.; Tamanyan, K.; Khutsishvili, M.; Atha, D.; Borris, R.P.; Koomoa, D.L. Scrophularia Orientalis Extract Induces Calcium Signaling and Apoptosis in Neuroblastoma Cells. Int. J. Oncol. 2016, 48, 1608–1616. [Google Scholar] [CrossRef] [Green Version]
  68. Ly, J.D.; Grubb, D.R.; Lawen, A. The Mitochondrial Membrane Potential (Δψm) in Apoptosis; an Update. Apoptosis 2003, 8, 115–128. [Google Scholar] [CrossRef] [PubMed]
  69. Cai, J.; Yang, J.; Jones, D.P. Mitochondrial Control of Apoptosis: The Role of Cytochrome c. Biochim. Et Biophys. Acta (BBA)—Bioenerg. 1998, 1366, 139–149. [Google Scholar] [CrossRef] [Green Version]
  70. Huang, K.; Zhang, J.; O’Neill, K.L.; Gurumurthy, C.B.; Quadros, R.M.; Tu, Y.; Luo, X. Cleavage by Caspase 8 and Mitochondrial Membrane Association Activate the BH3-Only Protein Bid during TRAIL-Induced Apoptosis. J. Biol. Chem. 2016, 291, 11843. [Google Scholar] [CrossRef] [Green Version]
  71. Scorrano, L.; Ashiya, M.; Buttle, K.; Weiler, S.; Oakes, S.A.; Mannella, C.A.; Korsmeyer, S.J. A Distinct Pathway Remodels Mitochondrial Cristae and Mobilizes Cytochrome c during Apoptosis. Dev. Cell 2002, 2, 55–67. [Google Scholar] [CrossRef] [Green Version]
  72. Alshangiti, A.M.; Togher, K.L.; Hegarty, S.V.; Sullivan, A.M.; O’Keeffe, G.W. The Dietary Flavonoid Isoliquiritigenin Is a Potent Cytotoxin for Human Neuroblastoma Cells. Neuronal Signal. 2019, 3, NS20180201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. De Oliveira, M.R.; Ferreira, G.C.; Schuck, P.F.; Dal Bosco, S.M. Role for the PI3K/Akt/Nrf2 Signaling Pathway in the Protective Effects of Carnosic Acid against Methylglyoxal-Induced Neurotoxicity in SH-SY5Y Neuroblastoma Cells. Chem. Biol. Interact. 2015, 242, 396–406. [Google Scholar] [CrossRef]
  74. Tomiyama, R.; Takakura, K.; Takatou, S.; Le, T.M.; Nishiuchi, T.; Nakamura, Y.; Konishi, T.; Matsugo, S.; Hori, O. 3,4-Dihydroxybenzalacetone and Caffeic Acid Phenethyl Ester Induce Preconditioning ER Stress and Autophagy in SH-SY5Y Cells. J. Cell Physiol. 2018, 233, 1671–1684. [Google Scholar] [CrossRef] [PubMed]
  75. Graham, R.M.; Hernandez, F.; Puerta, N.; De Angulo, G.; Webster, K.A.; Vanni, S. Resveratrol Augments ER Stress and the Cytotoxic Effects of Glycolytic Inhibition in Neuroblastoma by Downregulating Akt in a Mechanism Independent of SIRT1. Exp. Mol. Med. 2016, 48, e210. [Google Scholar] [CrossRef] [Green Version]
  76. Lin, M.C.; Lee, Y.W.; Tseng, Y.Y.; Lin, Y.W.; Chen, J.T.; Liu, S.H.; Chen, R.M. Honokiol Induces Autophagic Apoptosis in Neuroblastoma Cells through a P53-Dependent Pathway. Am. J. Chin. Med. 2019, 47, 895–912. [Google Scholar] [CrossRef]
  77. Picone, P.; Nuzzo, D.; Caruana, L.; Messina, E.; Scafidi, V.; Di Carlo, M. Curcumin Induces Apoptosis in Human Neuroblastoma Cells via Inhibition of AKT and Foxo3a Nuclear Translocation. Free. Radic. Res. 2014, 48, 1397–1408. [Google Scholar] [CrossRef] [PubMed]
  78. Tunjung, W.A.S.; Cinatl, J.; Michaelis, M.; Smales, C.M. Anti-Cancer Effect of Kaffir Lime (Citrus Hystrix DC) Leaf Extract in Cervical Cancer and Neuroblastoma Cell Lines. Procedia Chem. 2015, 14, 465–468. [Google Scholar] [CrossRef] [Green Version]
  79. Lantto, T.A.; Laakso, I.; Dorman, H.J.D.; Mauriala, T.; Hiltunen, R.; Kõks, S.; Raasmaja, A. Cellular Stress and P53-Associated Apoptosis by Juniperus Communis L. Berry Extract Treatment in the Human SH-SY5Y Neuroblastoma Cells. Int. J. Mol. Sci. 2016, 17, 1113. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, J.; Liu, Q.; Shi, L.; Qin, P.; Wang, Q. Honokiol Triggers Receptor-Interacting Protein Kinase 3-Mediated Cell Death of Neuroblastoma Cells by Upregulating Reactive Oxygen Species. Mol. Med. Rep. 2017, 16, 8525–8529. [Google Scholar] [CrossRef] [Green Version]
  81. Seervi, M.; Xue, D. Mitochondrial Cell Death Pathways in Caenorhabiditis Elegans. Curr. Top. Dev. Biol. 2015, 114, 43–65. [Google Scholar] [CrossRef]
  82. Shankar, E.; Goel, A.; Gupta, K.; Gupta, S. Plant Flavone Apigenin: An Emerging Anti-cancer Agent. Curr. Pharmacol. Rep. 2017, 3, 423–446. [Google Scholar] [CrossRef] [PubMed]
  83. Labbé, D.; Tremblay, A.; Bazinet, L. Effect of Brewing Temperature and Duration on Green Tea Catechin Solubilization: Basis for Production of EGC and EGCG-Enriched Fractions. Sep. Purif. Technol. 2006, 49, 1–9. [Google Scholar] [CrossRef]
  84. Javad, M.; Bsc, F.-M.; Nabian Bsc, P.; Gholami Bsc, A.; Dehghanbaghi Bsc, N.; Azizipanah Bsc, M.; Jokar Bsc, K.; Eslami Bsc, M.; Kargarian Bsc, Z.; Tamehri Bsc, M.; et al. A Review of Neuroblastoma: Prevalence, Diagnosis, Related Genetic Factors, and Treatment. Iran. J. Ped. Hematol. Oncol. 2018, 8, 237–246. [Google Scholar]
  85. Katiyar, S.K.; Elmets, C.A. Green Tea Polyphenolic Antioxidants and Skin Photoprotection (Review). Int. J. Oncol. 2001, 18, 1307–1313. [Google Scholar] [CrossRef] [PubMed]
  86. Tayyem, R.F.; Heath, D.D.; Al-Delaimy, W.K.; Rock, C.L. Curcumin Content of Turmeric and Curry Powders. Nutr. Cancer 2009, 55, 126–131. [Google Scholar] [CrossRef]
  87. Faienza, M.F.; Corbo, F.; Carocci, A.; Catalano, A.; Clodoveo, M.L.; Grano, M.; Wang, D.Q.H.; D’Amato, G.; Muraglia, M.; Franchini, C.; et al. Novel Insights in Health-Promoting Properties of Sweet Cherries. J. Funct. Foods 2020, 69, 103945. [Google Scholar] [CrossRef]
  88. Häkkinen, S.H.; Kärenlampi, S.O.; Heinonen, I.M.; Mykkänen, H.M.; Törronen, A.R. Content of the Flavonols Quercetin, Myricetin, and Kaempferol in 25 Edible Berries. J. Agric. Food Chem. 1999, 47, 2274–2279. [Google Scholar] [CrossRef]
  89. Cao, Y.; Wang, Y.; Ji, C.; Ye, J. Determination of Liquiritigenin and Isoliquiritigenin in Glycyrrhiza Uralensis and Its Medicinal Preparations by Capillary Electrophoresis with Electrochemical Detection. J. Chromatogr. A 2004, 1042, 203–209. [Google Scholar] [CrossRef]
  90. Wang, K.L.; Yu, Y.C.; Hsia, S.M. Perspectives on the Role of Isoliquiritigenin in Cancer. Cancers 2021, 13, 115. [Google Scholar] [CrossRef]
  91. Sukumari-Ramesh, S.; Bentley, J.N.; Laird, M.D.; Singh, N.; Vender, J.R.; Dhandapani, K.M. Dietary phytochemicals induce p53- and caspase-independent cell death in human neuroblastoma cells. Int. J. Dev. Neurosci. 2011, 29, 701–710. [Google Scholar] [CrossRef] [Green Version]
  92. Miean, K.H.; Mohamed, S. Flavonoid (Myricetin, Quercetin, Kaempferol, Luteolin, and Apigenin) Content of Edible Tropical Plants. J. Agric. Food Chem. 2001, 49, 3106–3112. [Google Scholar] [CrossRef]
  93. Lin, Y.; Shi, R.; Wang, X.; Shen, H.-M. Luteolin, a Flavonoid with Potential for Cancer Prevention and Therapy. Curr. Cancer Drug Targets 2008, 8, 634–646. [Google Scholar] [CrossRef]
  94. Fried, L.E.; Arbiser, J.L. Honokiol, a Multifunctional Antiangiogenic and Antitumor Agent. Antioxid. Redox Signal. 2009, 11, 1139. [Google Scholar] [CrossRef] [PubMed]
  95. Edlich, F. BCL-2 Proteins and Apoptosis: Recent Insights and Unknowns. Biochem. Biophys. Res. Commun. 2018, 500, 26–34. [Google Scholar] [CrossRef]
  96. Chen, Y.; Tseng, S.H.; Lai, H.S.; Chen, W.J. Resveratrol-Induced Cellular Apoptosis and Cell Cycle Arrest in Neuroblastoma Cells and Antitumor Effects on Neuroblastoma in Mice. Surgery 2004, 136, 57–66. [Google Scholar] [CrossRef]
  97. Soto, B.L.; Hank, J.A.; Van De Voort, T.J.; Subramanian, L.; Polans, A.S.; Rakhmilevich, A.L.; Yang, R.K.; Seo, S.; Kim, K.; Reisfeld, R.A.; et al. The Anti-Tumor Effect of Resveratrol Alone or in Combination with Immunotherapy in a Neuroblastoma Model. Cancer Immunol. Immunother. 2011, 60, 731–738. [Google Scholar] [CrossRef] [Green Version]
  98. Anantharaju, P.G.; Gowda, P.C.; Vimalambike, M.G.; Madhunapantula, S.V. An Overview on the Role of Dietary Phenolics for the Treatment of Cancers. Nutr. J. 2016, 15, 1–16. [Google Scholar] [CrossRef] [Green Version]
  99. Mennen, L.I.; Walker, R.; Bennetau-Pelissero, C.; Scalbert, A. Risks and Safety of Polyphenol Consumption. Am. J. Clin. Nutr. 2005, 81, S326–S329. [Google Scholar] [CrossRef] [Green Version]
  100. Ninfali, P.; Antonelli, A.; Magnani, M.; Scarpa, E.S. Antiviral Properties of Flavonoids and Delivery Strategies. Nutrients 2020, 12, 2534. [Google Scholar] [CrossRef] [PubMed]
  101. Sindhu, R.K.; Verma, R.; Salgotra, T.; Rahman, M.H.; Shah, M.; Akter, R.; Murad, W.; Mubin, S.; Bibi, P.; Qusti, S.; et al. Impacting the Remedial Potential of Nano Delivery-Based Flavonoids for Breast Cancer Treatment. Molecules 2021, 26, 5163. [Google Scholar] [CrossRef] [PubMed]
  102. Vazhappilly, C.G.; Amararathna, M.; Cyril, A.C.; Linger, R.; Matar, R.; Merheb, M.; Ramadan, W.S.; Radhakrishnan, R.; Rupasinghe, H.P.V. Current Methodologies to Refine Bioavailability, Delivery, and Therapeutic Efficacy of Plant Flavonoids in Cancer Treatment. J. Nutr. Biochem. 2021, 94, 108623. [Google Scholar] [CrossRef] [PubMed]
  103. Faria, A.; Mateus, N.; Calhau, C. Flavonoid Transport across Blood-Brain Barrier: Implication for Their Direct Neuroprotective Actions. Nutr. Aging 2012, 1, 89–97. [Google Scholar] [CrossRef] [Green Version]
  104. Youdim, K.A.; Qaiser, M.Z.; Begley, D.J.; Rice-Evans, C.A.; Abbott, N.J. Flavonoid Permeability across an in Situ Model of the Blood–Brain Barrier. Free Radic. Biol. Med. 2004, 36, 592–604. [Google Scholar] [CrossRef]
  105. Yang, Y.; Bai, L.; Li, X.; Xiong, J.; Xu, P.; Guo, C.; Xue, M. Transport of Active Flavonoids, Based on Cytotoxicity and Lipophilicity: An Evaluation Using the Blood–Brain Barrier Cell and Caco-2 Cell Models. Toxicol. Vitr. 2014, 28, 388–396. [Google Scholar] [CrossRef] [PubMed]
  106. Faria, A.; Meireles, M.; Fernandes, I.; Santos-Buelga, C.; Gonzalez-Manzano, S.; Dueñas, M.; De Freitas, V.; Mateus, N.; Calhau, C. Flavonoid Metabolites Transport across a Human BBB Model. Food Chem. 2014, 149, 190–196. [Google Scholar] [CrossRef]
  107. Zhu, Z.; Liao, L.; Qiao, H. Extracellular Vesicle–Based Drug Delivery System Boosts Phytochemicals’ Therapeutic Effect for Neurodegenerative Diseases. Acupunct. Herb. Med. 2022, 2, 229–239. [Google Scholar] [CrossRef]
  108. Wang, H.; Sui, H.; Zheng, Y.; Jiang, Y.; Shi, Y.; Liang, J.; Zhao, L. Curcumin-Primed Exosomes Potently Ameliorate Cognitive Function in AD Mice by Inhibiting Hyperphosphorylation of the Tau Protein through the AKT/GSK-3β Pathway. Nanoscale 2019, 11, 7481–7496. [Google Scholar] [CrossRef]
  109. Fan, Y.; Li, Y.; Huang, S.; Xu, H.; Li, H.; Liu, B. Resveratrol-Primed Exosomes Strongly Promote the Recovery of Motor Function in SCI Rats by Activating Autophagy and Inhibiting Apoptosis via the PI3K Signaling Pathway. Neurosci. Lett. 2020, 736, 135262. [Google Scholar] [CrossRef]
  110. Peng, H.; Li, Y.; Ji, W.; Zhao, R.; Lu, Z.; Shen, J.; Wu, Y.; Wang, J.; Hao, Q.; Wang, J.; et al. Intranasal Administration of Self-Oriented Nanocarriers Based on Therapeutic Exosomes for Synergistic Treatment of Parkinson’s Disease. ACS Nano 2022, 16, 869–884. [Google Scholar] [CrossRef]
  111. Sholler, G.L.S.; Gerner, E.W.; Bergendahl, G.; MacArthur, R.B.; VanderWerff, A.; Ashikaga, T.; Bond, J.P.; Ferguson, W.; Roberts, W.; Wada, R.K.; et al. A Phase I Trial of DFMO Targeting Polyamine Addiction in Patients with Relapsed/Refractory Neuroblastoma. PLoS ONE 2015, 10, e0127246. [Google Scholar] [CrossRef] [Green Version]
  112. Study of Nifurtimox to Treat Refractory or Relapsed Neuroblastoma or Medulloblastoma—Study Results—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/results/NCT00601003?recrs=eghim&rslt=With&type=Intr&cond=Neuroblastoma&draw=2&rank=31&view=results (accessed on 8 March 2023).
  113. Modak, S.; Kushner, B.H.; Basu, E.; Roberts, S.S.; Cheung, N.K.V. Combination of Bevacizumab, Irinotecan, and Temozolomide for Refractory or Relapsed Neuroblastoma: Results of a Phase II Study. Pediatr. Blood Cancer 2017, 64. [Google Scholar] [CrossRef] [PubMed]
  114. Thalidomide and Temozolomide in Relapsed or Progressive CNS Disease or Neuroblastoma—Study Results—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/results/NCT00098865?recrs=eghim&rslt=With&type=Intr&cond=Neuroblastoma&draw=2&rank=40#evnt (accessed on 8 March 2023).
  115. Irinotecan Hydrochloride and Temozolomide With Temsirolimus or Dinutuximab in Treating Younger Patients with Refractory or Relapsed Neuroblastoma—Study Results—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/results/NCT01767194?recrs=eghim&rslt=With&type=Intr&cond=Neuroblastoma&draw=2&rank=33#evnt (accessed on 8 March 2023).
  116. Henning, S.M.; Wang, P.; Said, J.W.; Huang, M.; Grogan, T.; Elashoff, D.; Carpenter, C.L.; Heber, D.; Aronson, W.J. Randomized Clinical Trial of Brewed Green and Black Tea in Men with Prostate Cancer Prior to Prostatectomy. Prostate 2015, 75, 550–559. [Google Scholar] [CrossRef] [Green Version]
  117. McLarty, J.; Bigelow, R.L.H.; Smith, M.; Elmajian, D.; Ankem, M.; Cardelli, J.A. Tea Polyphenols Decrease Serum Levels of Prostate-Specific Antigen, Hepatocyte Growth Factor, and Vascular Endothelial Growth Factor in Prostate Cancer Patients and Inhibit Production of Hepatocyte Growth Factor and Vascular Endothelial Growth Factor in Vitro. Cancer Prev. Res. 2009, 2, 673–682. [Google Scholar] [CrossRef]
  118. Thomas, R.; Williams, M.; Sharma, H.; Chaudry, A.; Bellamy, P. A Double-Blind, Placebo-Controlled Randomised Trial Evaluating the Effect of a Polyphenol-Rich Whole Food Supplement on PSA Progression in Men with Prostate Cancer—The UK NCRN Pomi-T Study. Prostate Cancer Prostatic Dis. 2014, 17, 180. [Google Scholar] [CrossRef] [Green Version]
  119. Pantuck, A.J.; Leppert, J.T.; Zomorodian, N.; Aronson, W.; Hong, J.; Barnard, R.J.; Seeram, N.; Liker, H.; Wang, H.; Elashoff, R.; et al. Phase II Study of Pomegranate Juice for Men with Rising Prostate-Specific Antigen Following Surgery or Radiation for Prostate Cancer. Clin. Cancer Res. 2006, 12, 4018–4026. [Google Scholar] [CrossRef] [Green Version]
  120. Nguyen, M.M.; Ahmann, F.R.; Nagle, R.B.; Hsu, C.H.; Tangrea, J.A.; Parnes, H.L.; Sokoloff, M.H.; Gretzer, M.B.; Chow, H.H.S. Randomized, Double-Blind, Placebo Controlled Trial of Polyphenon E in Prostate Cancer Patients before Prostatectomy: Evaluation of Potential Chemopreventive Activities. Cancer Prev. Res. (Phila.) 2012, 5, 290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Crew, K.D.; Ho, K.A.; Brown, P.; Greenlee, H.; Bevers, T.B.; Arun, B.; Sneige, N.; Hudis, C.; Mcarthur, H.L.; Chang, J.; et al. Effects of a Green Tea Extract, Polyphenon E, on Systemic Biomarkers of Growth Factor Signalling in Women with Hormone Receptor-Negative Breast Cancer. J. Hum. Nutr. Diet. 2015, 28, 272. [Google Scholar] [CrossRef] [Green Version]
  122. Campbell, J.L.; Cardelli, J.; McLarty, J.; Hershman, D.; Taback, B.; Refice, S.; Wang, S.; Bigelow, R.; Crew, K. Abstract PR-05: Effects of Presurgical Administration of Tea Polyphenols in Women with Operable Breast Cancer. Cancer Prev. Res. 2010, 3, PR-05. [Google Scholar] [CrossRef]
  123. Kapoor, R.; Ronnenberg, A.; Puleo, E.; Chatterton, R.T.; Dorgan, J.F.; Seeram, N.P.; Sturgeon, S.R. Effects of Pomegranate Juice on Hormonal Biomarkers of Breast Cancer Risk. Nutr. Cancer 2015, 67, 1113–1119. [Google Scholar] [CrossRef]
  124. Tsao, A.S.; Liu, D.; Martin, J.; Tang, X.M.; Lee, J.J.; El-Naggar, A.K.; Wistuba, I.; Culotta, K.S.; Mao, L.; Gillenwater, A.; et al. Phase II Randomized, Placebo-Controlled Trial of Green Tea Extract in Patients with High-Risk Oral Premalignant Lesions. Cancer Prev. Res. (Phila.) 2009, 2, 931. [Google Scholar] [CrossRef] [Green Version]
  125. Basak, S.K.; Bera, A.; Yoon, A.J.; Morselli, M.; Jeong, C.; Tosevska, A.; Dong, T.S.; Eklund, M.; Russ, E.; Nasser, H.; et al. A Randomized, Phase 1, Placebo-Controlled Trial of APG-157 in Oral Cancer Demonstrates Systemic Absorption and an Inhibitory Effect on Cytokines and Tumor-Associated Microbes. Cancer 2020, 126, 1668–1682. [Google Scholar] [CrossRef] [PubMed]
  126. Sinicrope, F.A.; Viggiano, T.R.; Buttar, N.S.; Wong Kee Song, L.M.; Schroeder, K.W.; Kraichely, R.E.; Larson, M.V.; Sedlack, R.E.; Kisiel, J.B.; Gostout, C.J.; et al. Randomized Phase II Trial of Polyphenon E versus Placebo in Patients at High Risk of Recurrent Colonic Neoplasia. Cancer Prev. Res. (Phila.) 2021, 14, 573. [Google Scholar] [CrossRef] [PubMed]
  127. Study Record|Beta ClinicalTrials.Gov. Available online: https://beta.clinicaltrials.gov/study/NCT01916239 (accessed on 7 March 2023).
  128. Blair, C.K.; Ross, J.A. A Pilot Study to Evaluate the Potential Cardioprotective Effects of Grape Juice in Survivors of Childhood Cancer. Ph.D. Dissertation, University of Minnesota, Minneapolis, MN, USA, 2011. [Google Scholar]
Figure 1. Basic skeletal structure of flavonoids. Created with MolView.com.
Figure 1. Basic skeletal structure of flavonoids. Created with MolView.com.
Biomolecules 13 00563 g001
Figure 2. Schematic diagram demonstrating the apoptotic effects of flavonoids on NB cell lines via a calpain-dependent pathway. The induction of ER stress induces Ca2+ release at the ER membrane, triggering caspase and CAD release and degradation of cytoskeletal proteins. Specific biomarkers can be viewed in Table 1. Created with BioRender.com.
Figure 2. Schematic diagram demonstrating the apoptotic effects of flavonoids on NB cell lines via a calpain-dependent pathway. The induction of ER stress induces Ca2+ release at the ER membrane, triggering caspase and CAD release and degradation of cytoskeletal proteins. Specific biomarkers can be viewed in Table 1. Created with BioRender.com.
Biomolecules 13 00563 g002
Figure 3. Schematic diagram demonstrating the anti-proliferative effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines. The inhibition of cell-cycle progression, cell motility, and gene expression limits NB-cell proliferation. Compounds caused cell-cycle arrest in the S phase, sub-G1 phase, G1 phase, and G2/M phase. Specific biomarkers can be viewed in Table 2. Created with BioRender.com.
Figure 3. Schematic diagram demonstrating the anti-proliferative effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines. The inhibition of cell-cycle progression, cell motility, and gene expression limits NB-cell proliferation. Compounds caused cell-cycle arrest in the S phase, sub-G1 phase, G1 phase, and G2/M phase. Specific biomarkers can be viewed in Table 2. Created with BioRender.com.
Biomolecules 13 00563 g003
Figure 4. Schematic diagram demonstrating the apoptotic effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines via mitochondrial or ER/oxidative-stress-related pathways. Elevated Bax/Bcl-2 ratio, increased PARP cleavage, loss of MMP and cytochrome C release, and ROS generation all contribute to apoptotic cell death. Specific biomarkers can be viewed in Table 3. Created with BioRender.com.
Figure 4. Schematic diagram demonstrating the apoptotic effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines via mitochondrial or ER/oxidative-stress-related pathways. Elevated Bax/Bcl-2 ratio, increased PARP cleavage, loss of MMP and cytochrome C release, and ROS generation all contribute to apoptotic cell death. Specific biomarkers can be viewed in Table 3. Created with BioRender.com.
Biomolecules 13 00563 g004
Figure 5. Schematic diagram demonstrating the apoptotic effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines via receptor-mediated pathways. Intrinsic apoptotic pathways were induced via mitochondrial dysfunction (influenced by Bax-protein levels) and pro-apoptotic complexes (due to Bad-protein increase). Specific biomarkers can be viewed in Table 4. Created with BioRender.com.
Figure 5. Schematic diagram demonstrating the apoptotic effects of flavonoids (blue) and non-flavonoid polyphenols (orange) on NB cell lines via receptor-mediated pathways. Intrinsic apoptotic pathways were induced via mitochondrial dysfunction (influenced by Bax-protein levels) and pro-apoptotic complexes (due to Bad-protein increase). Specific biomarkers can be viewed in Table 4. Created with BioRender.com.
Biomolecules 13 00563 g005
Table 1. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via a calpain-dependent pathway *.
Table 1. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via a calpain-dependent pathway *.
CompoundCell LineIncubation PeriodConcentration(s)Biomarker ChangesReference
Flavonoids
ApigeninSH-SY5Y24 h50 µM Intracellular free [Ca2+]
Calpain activation
Caspase-12, -3
CAD
[27]
EGCSH-SY5Y24 h50 µM Intracellular free [Ca2+]
Calpain activation
Cytoskeletal protein degradation
Caspase-3
CAD
[27]
EGCGSH-SY5Y24 h50 µM Intracellular free [Ca2+]
Calpain activation
Cytoskeletal protein degradation
Caspase-3
CAD
[27]
GenisteinSH-SY5Y24 h100 µM Intracellular free [Ca2+]
Calpain activation
Caspase-12
[27]
* denotes increase of biomarker, while denotes decrease.
Table 4. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via receptor-mediated pathways *.
Table 4. Results of in vitro studies of polyphenols’ apoptotic effects on NB cell lines via receptor-mediated pathways *.
CompoundCell LineIncubation PeriodConcentration(s)Biomarker ChangesReference
Flavonoids
DEDCSH-SY5Y24 h 7.5 µg/mL Phosphor-STAT3 expression (ROS mediated) [27]
Genistein SK-N-DZ24 h 10 µM TNF-α
FasL
TRADD
FADD
[27]
EGCSH-SY5Y24 h50 µM Caspase-8 activation
Proteolytic cleavage of Bid to tBid
Bax oligomerization
[27]
EGCGSH-SY5Y24 h100 µM Caspase-8 activation
Proteolytic cleavage of Bid to tBid
Bax oligomerization
[27]
Rutin LAN-5 24 h 25, 50, 100 μM TNF-α secretion[36]
Non-Flavonoid Polyphenols
Curcumin LAN-5 3, 5, 24 h 5, 10, 15, 20 µM Bad
PTEN
ROS
[77]
Honokiol Neuro-2a (mouse cell line) 30, 60, 120 µM 24, 48, 72 h RIP3
ROS
[80]
* denotes increase of biomarker, while denotes decrease.
Table 5. Results of in vivo studies of polyphenols’ apoptotic effects on NB cell lines *.
Table 5. Results of in vivo studies of polyphenols’ apoptotic effects on NB cell lines *.
CompoundCell LineIncubation PeriodDose(s)Effect(s)Reference
Flavonoids
DidyminSubcutaneous injection of 2 × 106 SMS-KCNR NB-cell suspensions into athymic nude mice8 weeks2 mg/kg
(oral gavage on alternate
days)
CD31 (angiogenesis marker)
ki67 (proliferation marker)
N-Myc (NB oncogenic marker)
Tumor mass
[34]
ApigeninScapular injection of 5 × 106 NUB-7 NB-cell suspensions into nonobese diabetic/severe combined immunodeficient mice1 week25 mg/kg
(intraperitoneal injection
daily)
Tumor mass (~50% less)
Apoptotic fraction
[33]
Non-Flavonoid Polyphenols
CurcuminOrthotopic injection of 1.5 × 106 GI-LI-N NB-cell suspensions into athymic nude mice4 weeks17.5 mg/kg
(intravenous injection
twice a week)
Tumor-growth rate[38]
ResveratrolSubcutaneous injection of 3 × 106 SK-N-AS NB-cell suspension into athymic nude mice5 weeks
OR
16 days
50 mg/kg
(oral gavage daily)
OR
20 mg (peritumor injection
5 times)
Tumor volume (~80% less)[61]
ResveratrolSubcutaneous injection of 1 × 106 Neuro-2a-cell suspension into A/J mice4 weeks40 mg/kg
(intraperitoneal injection
daily)
Tumor-growth rate
Long-term survival rate (~70%)
[96]
ResveratrolSubcutaneous injection of 2 × 106 NXS2 NB-cell suspension injected into A/J mice2 weeks20 mg
(peritumor injection
twice a week)
Tumor-growth rate
Long-term survival rate (~61%)
[97]
* denotes increase, while denotes decrease.
Table 6. Primary details from selected clinical studies on cancer and polyphenols.
Table 6. Primary details from selected clinical studies on cancer and polyphenols.
Cancer Type
(No. of Selected Studies)
Source of PolyphenolsNoted Outcomes *
Prostate Cancer (5)Green Tea and PomegranateFour studies produced statistically significant results, including decreased nuclear NFκB staining [116], decreased prostate-specific antigen (PSA) levels [117,118], and prolongation of PSA doubling time [119]. Another study also presented decreased PSA in study participants undertaking a trial polyphenol treatment, but this did not reach statistical significance [120].
Breast Cancer (3)Green Tea and PomegranateStatistically significant results, such as a decrease in serum hepatocyte growth factor (HGF) [121]. Non-statistically significant results included decreased serum vascular endothelial growth factor (VEGF) [121], as well as decreased serum HGF, in a different study [122].
Another study analyzed the effects of pomegranate juice on hormonal biomarkers of breast cancer risk [123]. Although the results presented statistically significant reductions in serum estrone and testosterone levels in women of normal weight, these results were not observed in overweight/obese women.
Oral CancerGreen Tea and PomegranateOne study investigated the effects of green tea, and the results presented a non-statistically significant downregulation of angiogenic stromal VEGF [124].
Another study investigated the effects of curcumin, with the results presenting a statistically significant reduction in inflammatory-cytokine concentrations in salivary cells [125].
Colorectal CancersGreen Tea and PomegranateOne study investigated green tea, which produced a non-statistically-significant reduction in percentage change in rectal aberrant crypt foci (ACF) number (compared to baseline measurements). These ACFs are generally seen as precursors of colorectal cancers [126].
Another study’s results showed no correlation between the levels of metabolites and the degree of differentiation of adenocarcinomas when investigating pomegranate extract [127].
* Only some results were selected from across the studies to provide the primary outcome(s). For the complete results of each study, please refer to their citations in the references.
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

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. https://doi.org/10.3390/biom13030563

AMA Style

Kafoud A, Salahuddin Z, Ibrahim RS, 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(3):563. https://doi.org/10.3390/biom13030563

Chicago/Turabian Style

Kafoud, Aisha, Zoya Salahuddin, Raghad Sabaawi Ibrahim, Reem Al-Janahi, Alena Mazurakova, Peter Kubatka, and Dietrich Büsselberg. 2023. "Potential Treatment Options for Neuroblastoma with Polyphenols through Anti-Proliferative and Apoptotic Mechanisms" Biomolecules 13, no. 3: 563. https://doi.org/10.3390/biom13030563

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