Next Article in Journal
Visceral Adipose Tissue Bioenergetics Varies According to Individuals’ Obesity Class
Next Article in Special Issue
Plumbagin Exhibits Genotoxicity and Induces G2/M Cell Cycle Arrest via ROS-Mediated Oxidative Stress and Activation of ATM-p53 Signaling Pathway in Hepatocellular Cells
Previous Article in Journal
Microbiome and Metabolome Variation as Indicator of Social Stress in Female Prairie Voles
Previous Article in Special Issue
Potential Focal Adhesion Kinase Inhibitors in Management of Cancer: Therapeutic Opportunities from Herbal Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy

by
Adele Chimento
1,*,
Arianna De Luca
1,
Maria D’Amico
1,2,
Francesca De Amicis
1,2,† and
Vincenzo Pezzi
1,†
1
Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
2
Health Center, University of Calabria, 87036 Rende, CS, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(2), 1680; https://doi.org/10.3390/ijms24021680
Submission received: 12 December 2022 / Revised: 9 January 2023 / Accepted: 11 January 2023 / Published: 14 January 2023
(This article belongs to the Special Issue Natural Compounds in Cancer Therapy and Prevention)

Abstract

:
Various literature data show how a diet rich in vegetables could reduce the incidence of several cancers due to the contribution of the natural polyphenols contained in them. Polyphenols are attributed multiple pharmacological actions such as anti-inflammatory, anti-oxidant, antibiotic, antiseptic, anti-allergic, cardioprotective and even anti-tumor properties. The multiple mechanisms involved in their anti-tumor action include signaling pathways modulation associated with cell proliferation, differentiation, migration, angiogenesis, metastasis and cell death. Since the dysregulation of death processes is involved in cancer etiopathology, the natural compounds able to kill cancer cells could be used as new anticancer agents. Apoptosis, a programmed form of cell death, is the most potent defense against cancer and the main mechanism used by both chemotherapy agents and polyphenols. The aim of this review is to provide an update of literature data on the apoptotic molecular mechanisms induced by some representative polyphenol family members in cancer cells. This aspect is particularly important because it may be useful in the design of new therapeutic strategies against cancer involving the polyphenols as adjuvants.

1. Introduction

Cancer is a complex pathological process mainly characterized by altered cell death and the uncontrolled proliferation of cells, which, through blood or lymphatic diffusion, can also give rise to distant metastases [1]. The continuing growth of global cancer incidence is rapidly outpacing the ability to control and block this disease [2]. In 2020, over 19 million new cases were diagnosed, and more than 10 million deaths were recorded [3]. Most cancers can have multiple causes of which the trigger is difficult to identify; generally, environmental [4], infectious [5] and hereditary factors [6] play a key role in cancer onset [7].
Cancer hallmarks include a set of functional capabilities acquired by human cells that allow the transition from a normal physiological state to a malignant neoplastic one [1]. The sustained proliferative signaling, evasion of growth suppressors and immune destruction, replicative immortality, angiogenesis, invasion, metastasis, as well as cell death resistance and reprogramming cellular metabolism, represent the main cancer hallmarks [1]. In particular, the loss of balance between cell proliferation and cell death can lead to cancer onset [8]. When death signals are missing, the uncontrolled cell proliferation occurs leading to different cancer types [9]. The main mechanism for cell death is represented by apoptosis, a complex process genetically controlled and evolutionarily conserved; it primarily consists of two main pathways, the extrinsic death-receptor (DR)-mediated one and the mitochondria-mediated one, both of which could lead to the same terminal execution pathway [10]. In addition, the inflammation, genome instability and mutations, as well as non-mutational epigenetic reprogramming and polymorphic variations in organ/tissue microbiomes, are involved in activating the above functional hallmark capabilities necessary for tumor growth and progression [1]. Genomic instability, resulting from increased mutation rate and DNA sequence alterations, is a major enabling cancer mechanism [11,12]. There is a close relationship between genomic integrity and cell death regulation [13,14]; genomic instability can lead to mutations or changes in the expression levels of cell death main regulators, while dysregulated apoptosis can promote genomic instability [13]. In fact, the telomere dysfunction [15], DNA double strand breaks [16], polyploidy [17] or abnormal mitoses [18] can directly trigger apoptosis through specific pathways; on the other hand, the inhibited apoptosis increases chromosomal instability risk and directs cells to survive for a long time. Therefore, apoptosis can prevent genetically damaged cells from surviving and thus can protect against genomic instability by limiting the viable mutants’ number. In this regard, the knowledge of dysregulation apoptosis and the signaling cascades triggering apoptosis in response to anticancer agents provides information on new effective therapeutic approaches based on molecular-targeted anticancer therapies.
Cancer therapy, indeed, has the main goal of blocking cancer cell survival and inducing death in all clonogenic cells. Current therapies such as chemotherapy [19] and radiotherapy [20] are capable of inducing cell death through various molecular mechanisms; however, they have disadvantages, including chemo- and radio-resistance [21] and various systemic side effects. The drug resistance mechanisms by cancer cells attenuate the therapeutic response, leading to tumor recurrence [21]. In order, therefore, to overcome cancer drug resistance, it is necessary to use alternative therapeutic approaches; a drug combination with natural molecules represents a great strategy not only effective in overcoming cancer drug resistance but useful for increasing anti-tumor effects and reducing systemic toxicity [22].
In this regard, polyphenols, a natural compounds of plant origin, represent promising approaches for either single or combined anti-tumor therapies, displaying greater efficacy and less toxicity [23,24]. In vitro and in vivo reports, as well as human studies, corroborate polyphenols’ protective effects against neurodegenerative and inflammatory chronic diseases, obesity, diabetes [25,26,27,28] and also tumor development [29,30,31]. However, human epidemiological studies are not having much success in evaluating polyphenols’ effects due to their poor bioavailability and high metabolism rate [32]. Accumulating data showed in vitro polyphenols anti-proliferative effects on several cancer types, such as prostate [33,34], colon [35,36], breast [37,38,39], lung [40,41], bladder [42,43], pancreatic [44,45], leukemia [46,47], osteosarcoma [48,49] and ovarian [50,51], through different mechanisms of action. They can influence important molecular events related to carcinogenesis; these include the modulation of cell cycle signaling, induction of detoxifying and antioxidant enzyme systems, alteration of the epigenome and metabolism, and changes in the expression of key proteins involved in signal transduction pathways (e.g., MAPK and PI3K) or in the activity of transcription factors (e.g., NFkB) [52,53,54]. Most of the well-studied polyphenols induce apoptosis [55]. Polyphenol-mediated apoptosis occurs in some cell lines at different concentrations by directly influencing different apoptotic pathways and/or the expression of regulatory proteins. Among the molecules that exert their regulatory effect in determining cell fate are the BCL2 family members [56], the caspases [57], and the transcription factor p53 [58], which represent the most important checkpoints that control the main apoptosis polyphenol-regulated steps [55].
In this paper, we reviewed the current knowledge regarding the molecular mechanisms by which most representative polyphenol family members induce apoptotic cell death in cancer cells. This aspect is very interesting because it could contribute to the design of new therapeutic strategies against tumor involving polyphenols as adjuvants.

2. Chemical Structure, Sources and Pharmacokinetics of Dietary Polyphenols

Polyphenols are water-soluble organic compounds; chemically, these compounds possess an aromatic ring and a benzene ring with one or more hydroxyl substituents [59]. Due to their great heterogeneity, they can be classified in two large groups, flavonoids and non-flavonoids, in turn divided into many subgroups based on the number of phenolic units within their molecular structure, substituent groups and/or the linkage type between phenolic units [59]. Isoflavones, flavonols, flavones, flavanones, flavanols and anthocyanins make up the majority of the flavonoid group [59]. Non-flavonoid compounds include phenolic acids (benzoic acid and cinnamic acid derivatives), lignans, stilbenes, and others [59] (Figure 1). Flavonoids present a C6-C3-C6 structure containing two benzene rings, A and B, connected by a heterocycle pyrene ring (C) that contains oxygen; most of them have a structure in which the C2 of the C ring is attached with a B ring, but C3 and C4 attachments are also found. Non-flavonoids, unlike flavonoids, contain only one phenolic ring [59]. It has been reported that specific chemical structural features are involved in the polyphenols’ biological actions. The structure of the catechol in the B ring is the main determinant of the radical scavenging capacity of the flavonoids; in the absence of catechol, the C2-C3 double bond, along with C3-OH and C4 carbonyl, is determinant for antioxidant action [60,61]. Moreover, the C ring chemical variations and the type and number of substituents, as well as the position of the substituents, can modify their antitumor activity. In fact, the presence of the C2-C3 double bond, the oxo group in C4, the open C ring and the position of the B ring (in C2 or C3) are the structural variations that not only differentiate flavones from flavanones, catechins, and isoflavones but may also affect their anticancer properties. Furthermore, both the sugar fractions and the polyhydroxylation reduce the hydrophobicity of the flavonoids by hindering their entry into the cells. In addition, the substituents’ modifications at different positions, such as at the 3’ and 4’ in the flavonoid structure, can modify their efficacy [60]. Structural modifications can be performed for the increased biological activity of the non-flavonoids. Aromatic ring modifications, changes in the hydroxyl groups position and number, and the addition of an electron-withdrawing group, as well as carboxyl group modifications, are critical structural features for the antioxidant activity of hydroxycinnamic acid [61]. Furthermore, several structures were developed from the stilbene parent molecule by hydroxyl, alkoxy, and glycoside group substitution in order to obtain derivatives with improved biological activity [61].
Flavonoids are the most widely distributed and studied polyphenols. In fact, dietary polyphenol consumption consists principally of 80% flavanols, 8% flavonols, 6% flavanones, 5% anthocyanins and less than 1% isoflavones and flavones [62].
The greatest dietary sources of flavonoids are tea, onions, apples, citrus fruits, berries, soybeans, legumes and red wine [63]. Specifically, isoflavones are very abundant compounds in legumes; in plants, they can be found mainly as inactive glycosides (e.g., daidzin and genistin) which are converted to the active corresponding aglycones (e.g., genistein, daidzein) after ingestion [64]. Flavonols (e.g., quercetin, myricetin, kampferol), although present in most edible plants (red wine, onions, blueberries and broccoli), are less concentrated than other flavonoids [65]. They mainly exist in the glycosylated form with glucose or rhamnose but can also be conjugated to galactose, xylose or arabinose [65]. Flavones, common in spices and some yellow- or orange-colored fruits and vegetables, include apigenin, luteolin, tangeritin and chrysin [66], while flavanones (e.g., hesperetin, hesperidin, naringenin) [67], generally glycosylated from a disaccharide, are the most abundant flavonoids in citrus fruits and in grapes and the medicinal herbs of Rutaceae, Rosaceae and Leguminosae. Flavanols, such as epicatechin, catechin, epicatechin gallate, epigallocatechin and epigallocatechin gallate (EGCG), are widespread in fruit and also in medicinal herbs and dietary plants (e.g., tea, apples, berries, cocoa and catechu) [68]; the oxidation of their monomeric form present in black tea following heating leads to the formation of complex molecules, such as theaflavins and thearubigins, which are dimeric and polymeric in nature, respectively [69]. Anthocyanins (e.g., cyanidin, malvidin, petunidin) are typical pigments found in red, blue or purple flowers and fruits; in particular, berries, currants, grapes and some tropical fruits have high anthocyanin content [70].
Compounds with smaller and simpler chemical structures than flavonoids belong to the non-flavonoids group, even if some of them are structurally more complex and have a high molar mass [59]. Phenolic acids, lignans, stilbenes and miscellaneous subgroups (e.g., curcuminoids) make up this group (Figure 1).
The phenolic acids include two subgroups: hydroxycinnamic and hydroxybenzoic acids. The first derive from cinnamic acid and are often present in foods as simple esters of quinic acid or glucose; the most abundant of them is the bound type, the chlorogenic acid, a combined form of caffeic acid and quinic acid, while the most common free forms are ferulic, caffeic, p-coumaric and sinapic acids [71]. Compared to the former group, the hydroxybenzoic acids (e.g., gallic, ellagic, p-hydroxybenzoic, protocatechuic, vanillic and syringic acids) are generally found in low concentrations in red fruits, black radish, mango, tea and onions [71]. Lignans (e.g., secoisolariciresinol) have 2, 3-dibenzylbutane as their basic structure, which is formed when cinnamic acid residues form a dimer [72]. Legumes, cereals in particular, wheat, pears, plums, garlic, carrots and asparagus are rich in lignans, but the main source of secoisolariciresinol are flax seeds. Stilbenes are found in small quantities in the human diet [73]; the major representative compound of this family are pterostilbene and resveratrol, which have been found in various edible natural products, such as grapes, peanuts, berries and rhubarb [73]. Curcuminoids are natural non-flavonoid polyphenol compounds derived from turmeric (Curcuma longa), the traditional Indian spice that is a member of the ginger family (Zingiberaceae); they include curcumin and related compounds that are bright yellow pigments constituting a group of lipophilic diketones [74].
The chemical structure, the degree of glycosylation/acylation and polymerization, the molecular size and also the solubility [75] influence polyphenol absorption and metabolism. Initially, polyphenols are conjugated with sugar moieties within plants (glycosides) and must be hydrolyzed to separate the sugar group (glycon) from the polyphenol (aglycone) prior to absorption. This process occurs in the epithelial cells of the small intestine by lactase-phlorizin hydrolase (LPH) [76]. Aglycones and simple monomeric polyphenols can be absorbed through the intestinal mucosa into the circulation, but undergo glucuronidation, sulfonoridation and methylation both within the epithelial cells and liver [77]. Since mammalians lack appropriate β-glycosidases, the glycosides cannot be absorbed; some of them can be partially absorbed due to the presence of enzyme present in the gastrointestinal microbiota. In particular, a minor dietary polyphenol amount (5–10% of total intake) can be easily absorbed using deconjugation reactions (e.g., deglycosylation) in the small intestine; these polyphenols undergo to extensive phase I (e.g., oxidation, reduction, hydrolysis, etc.) and phase II (e.g., conjugation reactions) biotransformations in enterocytes, originating water-soluble metabolites (e.g., methyl, glucuronide, sulfate derivatives, etc.) that can be rapidly released into systemic circulation for subsequent organ distribution, including to the liver [77]. Here, polyphenols undergo a further rapid phase I and II metabolism. The formation of anionic derivatives by glucuronidation and sulfatation facilitates their urinary and biliary excretion and then their rapid elimination. In the bile, some of them can be deconjugated and reabsorbed several times (enterohepatic cycle) [77]. Furthermore, 90–95% of ingested polyphenols (most flavonoids) reach the colon, where are metabolized by gut microbiota producing several metabolites of phase II (e.g., methyl, glucuronide, sulfate derivatives) or dehydroxylated, deglycosylated and demetylated products destined for faecal excretion [77].
Therefore, factors such as low water solubility, poor absorption, low tissue distribution, high rate of metabolism, inactivity of metabolic products and/or rapid elimination contribute to the low bioavailability of polyphenols, which limits their clinical applications. Several strategies have been developed over the years in order to improve the polyphenols’ pharmacokinetic profile. These include the preparation of new analogs and drug delivery systems such as emulsions and liposomes that could improve their solubility and prolong their residence time in plasma [78,79,80].

3. Polyphenol-Mediated Apoptosis as Anticancer Mechanism

Polyphenols are known for their beneficial properties against various pathological conditions, such as diabetes, cardiovascular and neurodegenerative diseases, as well as cancer [26,63,81]. Polyphenols’ anticancer activity is mainly attributed to their antioxidant property being strong radical scavengers, metal chelators, and modifiers of endogenous defense mechanisms, such as SOD, CAT, GPx, GSH and regulators of several proteins and transcription factors, such as Nrf2 [25,82]. Furthermore, they exhibit the ability to decrease tumor cells growth through the inhibition of polyamine biosynthesis and signal transduction enzymes like PTK, PKC and PI3K and through the expression modulation of proteins involved in cell cycle arrest, cell migration, metastasis and cell death [23,63,81,83,84,85].
Polyphenols’ anticancer effects largely depend on their ability to trigger apoptotic mechanisms of death [83]. At a cellular level, apoptosis can occur in mammalian cells by extrinsic or intrinsic pathways that are mainly carried out by caspases, a family of cysteine-dependent aspartate-directed proteases [86]. The extrinsic pathway is activated when a specific ligand binds (e.g., TNFα, FasL, TRAIL) its related cell surface death receptor (e.g., TNF-R, Fas, TRAIL-R); this event is followed by receptor trimerization, the recruitment of adaptors proteins (e.g., FADD) and the activation of an initiator caspase (e.g., procaspase-8) [87]. The intrinsic or mitochondrial pathway is activated by several agents, such as oxidants, toxins, drugs or ionizing radiation, which induce ROS overproduction and then oxidative stress. It involves the mitochondria and mitochondrial proteins [87], such as those belonging to the BCL2 family, which include anti-apoptotic proteins (bcl-2, bcl-xL, bcl-w, mcl-1, Bfl-1/A1), pro-apoptotic pore-formers (bax, bad, bak, bok) and (3) pro-apoptotic BH3-only proteins (bid, bik, bim, bmf, hrk, noxa, puma, etc.) [88]. The intrinsic pathway is characterized by cytochrome c translocation from the mitochondrial intermembrane space into the cytoplasm. Cytochrome c, by binding apaf-1 and procaspase-9, forms the molecular apoptosome complex, which activates caspase-9 through autocatalysis. The two apoptotic pathways can be linked by the caspase-mediated activation of some proteins (e.g., Bid) that can directly affect the mitochondrial intrinsic pathway [88,89] (Figure 2). Both extrinsic and intrinsic pathways lead to the activation of effector caspases (e.g., caspase-3) which, in turn, by targeting specific targets, cause DNA fragmentation, apoptotic bodies’ formation and finally cell death. In case of DNA damage, the activation of pro-apoptotic genes or tumor suppressor genes such as p53 can trigger the apoptosis initiation; p53 is able to modulate key control points in both intrinsic and extrinsic pathways [90] by transcriptionally upregulating apoptosis-related proteins (e.g., puma, noxa, bid and bax) expression and physically interacting with and neutralizing the anti-apoptotic activity of bcl-2 and bcl-xL; moreover, it can transactivate the death receptor genes and/or induce those (e.g., PTEN) that inhibit the antiapoptotic pathway, such as the of PI3K/AKT survival signaling [90]. In addition, massive damage to mitochondria also plays a key role in mitochondrial apoptosis [91]. Cancer cells with absent tumor suppressors’ activity may develop some survival mechanisms to prevent apoptosis progression. Tumors with loss of p53 and PTEN show high expressions of anti-apoptotic genes such as PI3K, NF-κB and BCL2. On the other hand, a reduction in the expression of pro-apoptotic genes can be expected for most tumor cells [92]. In particular, cancer cells can escape apoptosis by: (i) upregulating or downregulating the anti- or pro-apoptotic genes’ expression, respectively; (ii) modulating anti- or pro-apoptotic proteins functions through post-translational modifications (e.g., phosphorylation); or (iii) decreasing caspases function [9]. Therefore, a promising anti-tumor strategy could consist of the restoration of one or both apoptotic pathways through the inhibition of antiapoptotic factors and/or the stimulation of proapoptotic ones [10].
Several data reported that polyphenols can trigger both the extrinsic and intrinsic apoptotic pathways by inhibiting antiapoptotic factors and/or activating proapoptotic molecules; these molecular events lead to morphological and biochemical changes culminating in cancer cells’ apoptosis (Figure 2).
The main apoptotic mechanisms induced by some representative flavonoid and non-flavonoid family members in cancer cells are summarized in Table 1.

3.1. Isoflavones and Apoptosis

Epidemiological studies reported a lower incidence of cancer in Asian subjects who consume soy products [93]. In fact, multiple anti-cancer actions have been attributed to isoflavone genistein, the main soy product [94]. In isoflavones, the presence of –OH between C4 and C7 makes them similar to the estradiol structure. Given the structural similarity of genistein to estrogen, it may exhibit estrogenic activity by acting on estrogen receptor (ER) alpha and beta, mainly through the classical genomic mechanism [95]. However, it differs from estrogens in its preference for ERβ, showing a 20-fold higher affinity for this over ERα [96]. Evidence suggests that the metabolism and biological effects of genistein may vary with dosage or exposure levels, even depending on the cell type involved. In fact, there is a duplicity of genistein action as it appears to be a possible beneficial or therapeutic agent in some cases and an endocrine disruptor in others. In vitro studies on tumor cells highlighted very variable and contrasting biological effects, especially in relation to cell proliferation and apoptosis depending on the used dose: low doses (≤1 μg/mL) promote proliferation and inhibit apoptosis, while higher exposure levels (≥10 μg/mL) produce growth inhibitory effects and induce apoptotic mechanisms [95].
Genistein induced apoptosis in MCF-7 breast cancer cells via regulating ERα expression and altering the bax/bcl-2 ratio [97]. Bcl-2 and bcl-xL downregulation was observed in MDA-MB-231 breast cancer cells together to endogenous copper ions’ mobilization and ROS generation [98]. Recently, ROS-dependent apoptosis mediation by genistein was demonstrated in A549 and 95D non-small cell lung cancer (NSCLC) [99]. In these cell models, genistein elevated intracellular ROS generation, decreased MMP, upregulated bax expression and cytochrome c release from mitochondria and decreased bcl-2 protein levels. Further examinations revealed that the expression level of FOXO3a and puma in NSCLC was significantly increased by genistein [99]. Genistein-mediated cell apoptosis through endoplasmic reticulum stress- and mitochondria-dependent pathways was demonstrated in HL-60 leukemia cells [100]. In these cells, it promoted ROS and Ca2+ accumulation; decreased MMP, bcl-2 and bid protein levels; and increased the ATF-6α, GRP78, bax, bad, bak, parp-1 cleavage, caspase-9 and -3 expression [100]. Similarly, in T24 human bladder cancer cells, genistein determined apoptosis through ROS-dependent PI3K/AKT signal transduction pathway inhibition [101]. In particular, it caused mitochondrial dysfunction, which was associated with bcl-2/bax expression ratio decrease; parp-1 cleavage; cytochrome c release into cytosol; and caspase-3, -8 and -9 activation [101]. Genistein prompted the generation of significant ROS amounts favoring cell death in the HepG2 liver cancer cell line [102]. In this cell model, mitochondrial apoptosis was associated with cytosolic cytochrome c, bax, cleaved caspase-3 and -9 expression increase and bcl-2 decrease [102]. Moreover, genistein-mediated apoptosis was demonstrated in HT-29 colon cancer cells where it activated caspase-3 [103]; the same effects on caspase-3 activity together with a bax expression increase was observed in HCT-116 and LoVo colon cancer cells [104]. Similarly, bcl-2, bcl-xL, c-IAP1, survivin and NF-κB downregulation after genistein use was found in A2780 and C200 ovarian cancer cells [105]. On the contrary, in LNCaP tumor prostate cells, genistein induced apoptosis via the TRAIL-mediated disruption of MMP [106]. In the same tumor cell model and C4-2B bone metastatic LNCaP-derivative prostate cancer cells, genistein reduced cell proliferation and induced apoptosis, as confirmed by elevated DNA fragmentation; moreover, the combination of low dose of genistein and daidzein displayed synergistic preventive effects on prostate cancer cells when compared with single soy isoflavone [107].

3.2. Flavonols and Apoptosis

Quercetin, the most predominant flavonol in plants, possesses phytoestrogenic action, as it is structurally similar to estradiol. Several studies demonstrated its anti-cancer actions involving growth inhibition and apoptosis induction through the modulation of multiple signaling pathways including those of p53, NF-κB, MAPK, JAK/STAT, PI3K/AKT, and Wnt/β-catenin; in addition, quercetin controls the activity of oncogenic and tumor suppressor ncRNAs [108,109]. In particular, quercetin induced intrinsic apoptotic mechanism via bax increase, bcl-2 decrease [110,111], MMP decrease, AIF release and caspase-6, -9 activation in MCF-7 breast cancer cells [111]. Quercetin-mediated mitochondrial apoptotic effects were also observed in HL-60 leukemia cells through COX2 inhibition, caspase-3 activation, bax and bad increase, bcl-2 decrease, cytochrome c release and parp-1 cleavage [112]. Moreover, quercetin showed an inhibitory effect on A549 lung cancer cell proliferation, by inducing apoptosis through BAX and BCL2 gene expression modulation and caspase-3 activation [113]. Zhou and coworkers demonstrated that quercetin-mediated apoptosis in SKOV-3 and A2780 human ovarian cancer cell lines was dependent on miRNA regulation; quercetin, by stimulating miR-145 expression, induced caspase-3 expression and then apoptosis [114]. The combinatory treatment of quercetin and irradiation caused DNA damage and apoptosis induction in OV2008 and A2780 ovarian cancer cells; in particular, the cell death occurred by bax and p21 increase and bcl-2 decrease in protein expression, in a p53 dependent manner [115]. On the contrary, quercetin triggered a caspase-dependent extrinsic apoptosis by upregulating caspase-8 and -3 and parp-1 cleaved forms in BT474 human breast cancer cells [116]. A study performed in Hela cervical cancer cells demonstrated that the genes involved in the apoptotic extrinsic pathway are upregulated by quercetin [117]. In particular, the expression of TRAIL, FasL, TNF and their receptors (Fas, TNFSF10, TNFRSF10A, TNFRSF10B, TNFRSF1A, TNFRSF1B, TNFRSF21, TNFRSF25), as well as those of TRADD, CRADD and DEDD, was elevated after quercetin treatment. Moreover, the enhanced expression of caspase-8, -10, -3 and -7 confirmed the role of the extrinsic pathway in quercetin-mediated apoptosis [117].

3.3. Flavones and Apoptosis

Apigenin, a member of the flavone family, is a nutraceutical that has been reported to be an anticancer agent in several experimental studies. It is able to reduce cell growth and induce apoptosis in different tumors such as skin, colon, prostate, breast, lung, liver, pancreas, cervical, oral and stomach [118,119]. Apigenin triggers both the intrinsic and extrinsic pathways of apoptosis. The apigenin-mediated intrinsic pathway occurs primarily via cytochrome c release, bax expression increase and caspase-3 activation, while the extrinsic pathway activation is confirmed by caspase-3, caspase-8 and TNF-α mRNA upregulation [118]. The pro-apoptotic properties of apigenin was demonstrated in PC-3 and DU145 prostate cancer cells. In these cell models, it triggered apoptosis by both bax protein expression and cytochrome c release increase, and XIAP, c-IAP1, c-IAP2, survivin, bcl-2 and bcl-xL decrease. In particular, the apigenin-mediated increase of bax was due to the dissociation of bax from Ku70, an important event for bax apoptotic activity [120]. The same effects on BCL2 family members was found in the T24 bladder cancer cell line, where it upregulated bax, bad and bak and downregulated bcl-2, bcl-xL and mcl-1 protein expression; these events were associated with caspase-9, -3, -7 activation, cytochrome c release and parp-1 cleavage [121]. Woo and coworkers demonstrated that apigenin was able to induce apoptosis by regulating AKT and MAPK pathways in A375SM human melanoma cells. In particular, in apigenin-treated cells, p-ERK and p-JNK increased, while p-p38 and p-AKT protein expression levels decreased; these molecular events correlated to downregulation of bcl-2, and upregulation of parp-1 and caspase-9 cleaved forms, as well as bax and p53, in a dose-dependent manner [122]. The apigenin-mediated extrinsic apoptotic mechanism was demonstrated in SCC25 and A431 HNSCC cells via TNF-R, TRAIL-R and bcl-2-mediated caspase-dependent cell death [123]. Additionally, apigenin-induced apoptosis was related to p53 expression, bax/bcl-2 ratio increase, caspases-9 and -8 activation and parp-1 cleavage in malignant mesothelioma cells [124].

3.4. Flavanones and Apoptosis

Among the flavanones, hesperetin showed great anti-tumor efficacy that primarily occurs via targeting apoptosis-mediated cell death pathways [125]. A study performed in both an in vitro and in vivo gastric cancer cell model demonstrated that hesperetin induced the intrinsic apoptotic mechanism by increasing ROS levels; in particular, it determined apaf-1 and bax expression upregulation, caspases-3 and -9 activation, MMP and bcl-2 expression decrease, cytochrome c release and DNA fragmentation [126]. The same ROS-dependent apoptotic mechanism was also observed in Eca109 esophageal [127], HT-29 colon [128] and MCF-7 breast [129] cancer cells. In Eca109 cells, apoptosis was confirmed by caspase-9 and -3 activation; apaf-1, SuFu and bax protein expression increase; and bcl-2 decrease [127]. In HT-29 cells, hesperetin-mediated apoptosis was associated with cytochrome c release, bax and cleaved caspase-3 expression increase and bcl-2 expression decrease, as well as with ROS increase and antioxidants enzymes such as SOD, CAT and GPx inhibition [128]. In MCF-7 cells, hesperetin treatment caused phosphatidyl-serine externalization, DNA fragmentation, caspase-9 and -7 activation, MMP loss, cytochrome c release, bax/bcl-2 ratio increase and parp-1 cleavage [129]. Conversely, in H522 lung cancer cells, hesperetin extrinsically determined apoptosis in a p53- and bax-independent manner through Fas and FADD overexpression and caspase-8 activation [130]. In non-small cell lung cancer, the A549 hesperetin-mediated suppression of Hsp70, a negative regulator of the mitochondrial apoptosis pathway, was associated with reduced cytosolic bax and increased mitochondrial bax levels, leading to mitochondrial apoptotic cascade activation [131]. Moreover, the hesperetin-mediated both intrinsic and extrinsic apoptotic pathway was found in SiHa cervical cancer cell lines [132].

3.5. Flavanols and Apoptosis

Epicatechin, a flavanol derived primarily from green tea and other plants, is found in most commonly consumed food beverages, unlike many phytochemicals. Several studies suggested that one of the possible mechanisms of epicathechin-mediated cancer inhibition is apoptosis induction [133]. The apoptotic effects of epicatechin were evaluated in breast cancer cells. In MDA-MB-231 cells, it upregulated death receptor (DR4/DR5), increased ROS production and mitochondrial permeability and modulated pro-apoptotic proteins (e.g., cytochrome c, Smac/Diablo, and HtrA2/Omi) [134]. In MCF-7 cells, it did not generate changes in the TRAIL receptor, but an increase in ROS and the upregulation of pro-apoptotic proteins (e.g., bad and bax) were observed [134]. In another cancer cell model, SW480 human colon cells, the epicathechin apoptotic effects were related to bax and p53 mRNA and protein expression increase, as well as to bcl-2 decrease and DNA fragmentation [135]. Moreover, a recent study demonstrated that epicatechin acts as an agonist towards the novel androgen receptor ZIP9 by miming testosterone-induced apoptosis through this receptor in prostate and breast cancer cells; in particular, it exerted androgenic actions inducing cell death through an intrinsic apoptotic pathway, as confirmed by bax expression increase, cytochrome c release and caspase-3 activation [136].

3.6. Anthocyanins and Apoptosis

Several studies indicated anthocyanins as nutraceutical for cancer prevention and management. Anthocyanins’ anti-tumor actions are due to their antioxidant, anti-inflammatory and anti-proliferative properties and their ability to regulate apoptosis-related mediators, including p53, bcl-2, bax, cytochrome c and caspase-3 in several cancer cell models [137]. In DU145 and LnCap prostate cell lines, cyanidin determined caspase-3 activation and DNA fragmentation [138], while in U87 glioblastoma cells, apoptosis occurred via bax and p53 mRNA expression increase and bcl-2 decrease [139]. Another recent in vitro study demonstrated that cyanidin induced apoptosis through the upregulation of PPARγ, p21 and bax mRNA expression in osteosarcoma cell lines [140].

3.7. Phenolic Acids and Apoptosis

Phenolic acids are a subgroup of non-flavonoid compounds that are associated with potent anticancer abilities demonstrated in several in vitro and in vivo studies. They act as anti-tumor agents by promoting apoptosis, reducing proliferation and differentiation and targeting other hallmark of cancer, such as angiogenesis and metastasis [141].
The hydroxycinnamic derivative ferulic acid was able to trigger apoptosis process. Ferulic acid-mediated apoptosis was evaluated in MCF-7 breast cancer cells where it caused both caspase-8 and -9 activation [142]. The same caspases’ activation was found in HepG2 hepatic cancer cells after ferulic acid treatment, although caspase-8 activation was more involved [142]. Furthermore, recently, it has been demonstrated that ferulic acid increased bax/bcl-2 ratio and displayed a synergistic activity with epirubicin on apoptosis induction in MDA-MB-231 breast cancer cells [143]. Additionally, ferulic acid treatment remarkably downregulated the PI3K/AKT, reduced bcl-2 and mcl-1 expression and increased that of bax in Caski cervical cancer cells [144].
Similarly, gallic acid was able to induce apoptosis through Fas, FasL and DR5 expression upregulation; caspase-8, -9 and -3 activation; bad and bak protein expression increase; and bcl-2 decrease in AGS gastric cancer cells [145]. In particular, in these cells, p53 was shown to be involved in gallic-acid-mediated apoptosis [145]. Additionally, in MDA-MB-231 breast cancer cells, it increased caspase-8 and -9 activity, decreased MMP and induced cytochrome c release [146]. The ability of gallic acid to trigger apoptosis and also potentiate the apoptotic effect of paclitaxel and carboplatin has also been demonstrated in MCF-7 breast cancer cells where it caused the overexpression of bax, caspase 3 and p53 and the downregulation of bcl-2 [147].

3.8. Lignans and Apoptosis

Many studies demonstrated that natural lignans possess great anti-tumor activities against a variety of human cancer cell lines by inhibiting all phases of carcinogenesis, including initiation, promotion and progression and by modulating different signaling pathways involved in cell proliferation and death control [148]. Dietary lignan secoisolariciresinol diglucoside can mainly induce the intrinsic apoptotic mechanism by targeting several molecules. Recently, its apoptotic effects were investigated in both 2D and 3D cultures of SW480 colon cancer cells. Results indicated a significant increase in AIF and caspase-3 staining and gene expression in lignan-treated cells compared to the control group in both monolayer and spheroid culture cells [149]. Moreover, a mechanistic study showed that the same compound reduced MCF-7 cell proliferation via downregulating ER and growth factor-mediated signaling, also triggering apoptosis. In particular, it significantly decreased the mRNA expression of bcl-2, cyclin D1, pS2, ERα, ERβ, EGFR and IGF-IR [150].

3.9. Stilbenes and Apoptosis

Stilbenes displayed an extraordinary potential for the prevent and treatment of different diseases, including cancer, due to their antioxidant and anti-inflammatory properties and ability to induce cell death [151]. Pterorostilbene is an active apoptotic agent in different in vitro cell models. Liu et al. reported its ability to inhibit osteosarcoma cell growth via JAK2/STAT3 signaling pathway downregulation. This led to a decrease in the protein expression of antiapoptotic factors (e.g., bcl-xL and mcl-1) and an increase in that of proapoptotic proteins (e.g., bax, bak, cytochrome c, caspase-3) and cyclin-dependent kinase inhibitors (e.g., p21 and p27) [152]. Pterostilbene treatment inhibited MCF-7 and MDA-MB-231 breast cancer growth in vitro through caspase-3, -7-dependent apoptosis; indeed, it decreased MMP and increased superoxide anions, activating downstream effector caspases [153]. In a recent work, by examining pterostilbene effects in the mutant p53-breast cancer cell lines MDA-MB-231 and T-47D, it has been demonstrated that it decreased mutant p53 protein expression while increasing the pro-apoptotic bax protein expression [154]. The same effect on bax expression was observed in MDA-MB-468 breast cancer cells, where it inhibited AKT and mTOR phosphorylation [155]. Another investigation demonstrated that this compound activated caspase-3 and decreased the expression of cell survival proteins bcl-2 and bcl-xL in HEC-1A and ECC-1 endometrial cells. Interestingly, it increased the anti-tumor activity of megestrol acetate, a commonly used drug in endometrial cancer patients [156]. The pro-apoptotic effects of pterostilbene were demonstrated in HeLa cervical cancer cells through the ROS-mediated activation of caspase-3 and caspase-9, as well as the downregulation of the antiapoptotic proteins bcl-2 and bcl-xL [157]. The proposed mechanism for ROS-dependent pterostilbene anticancer activity was assessed using HepG2 liver cancer cells; in these cells, it increased p53 expression, which, in turn, decreased SOD2 expression and induced ROS generation, leading to apoptosis [158].

3.10. Curcuminoids and Apoptosis

Polyphenolic curcuminoids, including the curcumin (or curcumin I), demethoxy curcumin (or curcumin II) and bis-demethoxycurcumin (curcumin III), represent 95% of the standardized extract from the underground root (rhizome) of Curcuma longa L. [159]. Curcumin, the most abundant curcumonoid (~77%), displays anticancer effects that occur by interfering with various cellular pathways and inducing/inhibiting the expression of several proteins related to proliferation, angiogenesis, invasion, migration, metastasis and apoptosis [78,160]. Curcumin-mediated apoptosis in cancer cells occurs by modulating both extrinsic and intrinsic apoptotic pathways mediators. Extrinsic apoptosis has been demonstrated through increased ROS production in leukemia [161], melanoma [162], breast [163] and renal cancer cells [164]. Furthermore, the upregulation of TRAIL, in glioblastoma multiforme [165], renal [164], breast [163], prostate cancer cells [166] and FasL in hepatocellular carcinoma [167] plays a key role in curcumin-mediated extrinsic apoptosis. In a recent paper, Obaidi and co-workers demonstrated that curcumin sensitized cancerous kidney cells to TRAIL-induced apoptosis via the Let-7C mediated deregulation of cell cycle proteins and cellular metabolism [168]. On the other hand, changes in mitochondria function, as well as the p53-dependent bax upregulation in breast [169] and colon cancer cells [36] and the bcl-2 down-regulation in laryngeal [170], breast [171] and prostate [172] cancer cells, are the most crucial changes that occur in curcumin-mediated intrinsic apoptosis. Furthermore, the ability of curcumin to activate caspase-8 in melanoma [173], and caspase-3 in hepatocellular carcinoma [167], neuroblastoma [174], linfoma [175] and lung cancer cells [176] confirmed its pro-apoptotic role. In RT4 schwannoma cells curcumin reduced bcl-2 expression, enhanced that of bax, and activated caspase-3, caspase-9, and parp-1 via miRNA 344a-3p upregulation [177]. Recently, apoptotic cell death by curcumin was observed in BCPAP and TPC-1 papillary thyroid cancer cells and derived thyroid cancer stem-like cells (thyrospheres) by targeting JAK/STAT3 signaling pathway. In particular, it suppressed the constitutive JAK/STAT3 signaling pathway and upregulated the ROS level to induce apoptosis. Indeed, protein expression analysis showed that curcumin-mediated caspase-3 activation, bcl-2 and bcl-xL decrease and bax increase were markedly reversed by NAC, a known ROS scavenger; these results confirmed the role of oxidative stress in curcumin-induced apoptosis [178].
Table 1. Apoptotic mechanisms induced by some representative flavonoid and non-flavonoid family members in cancer. ↓: decrease, ↑: increase; NSCL: non-small cell lung cancer; HNSCC: head and neck squamous cell carcinoma.
Table 1. Apoptotic mechanisms induced by some representative flavonoid and non-flavonoid family members in cancer. ↓: decrease, ↑: increase; NSCL: non-small cell lung cancer; HNSCC: head and neck squamous cell carcinoma.
FamilyPhytochemical CompoundsTypes or Names of CancerMechanism Leading to ApoptosisReferences
Flavonoids
Isoflavonesgenisteinbreastbax↑, bcl-2↓[97]
bcl-2↓, bcl-xL↓, ROS↑[98]
NSCLROS↑, MMP↓, cytochrome c↑, bcl-2↓, bax↑, FOXO3A↑, PUMA↑[99]
leukemiaROS↑, MMP↓, bcl-2↓, bid↓, ATF-6α↑, GRP78↑, bax↑, bad↑, bak↑, cleaved parp-1↑, caspase-9↑, caspase-3↑[100]
bladderbcl-2/bax ratio↓, caspase-3, -8 and -9 activation, cytochrome c↑, cleaved parp-1↑[101]
liverROS↑, cytochrome c↑, Bax↑, cleaved caspase-3↑, cleaved caspase-9↑, bcl-2↓[102]
coloncaspase-3 activation[103]
caspase-3 activation, bax↑[104]
ovarianbcl-2↓, bcl-xL↓, c-IAP↓, survivin↓[105]
prostateTRIAL↑, MMP↓[106]
DNA fragmentation↑[107]
Flavonolsquercetinbreastbax↑, bcl-2↓[110,111]
AIF↑, MMP↓, caspase-6, -9 activation[111]
caspase-8, -3 activation, cleaved parp-1↑ [116]
leukemiabax↑, bad↑, bcl-2↓, cytochrome c↑, caspase-3 activation, cleaved parp-1↑[112]
lungbax↑, bcl-2↑, caspase-3 activation[113]
ovariancaspase-3 activation [114]
bax↑, p21↑, bcl-2↓, p53↑[115]
cervicalTRAIL↑, FasL↑, TNF↑, Fas↑, TNFSF10↑, TNFRSF10A↑, TNFRSF10B↑, TNFRSF1A↑, TNFRSF1B↑, TNFRSF21↑, TNFRSF25↑, TRADD↑, CRADD↑, DEDD↑, caspase-8↑, -10↑, -3↑, -7↑[117]
Flavones apigeninprostatebax↑, bcl-2↓, bcl-xL↓, c-IAP1↓, c-IAP2↓, XIAP↓, survivin↓, cytochrome c↑[120]
bladderbax↑, bad↑, bak↑, bcl-2↓, bcl-xL↓, mcl-1↓, caspase-9, -3, -7 activation, cytochrome c↑, cleaved parp-1↑ [121]
melanomabcl-2↓, bax↑, cleaved caspase-9 ↑, cleaved parp-1↑, p53 ↑[122]
HNSCCTNF-R↑, TRAIL-R↑, bcl-2↓, caspase activation[123]
mesotheliomabax/bcl-2 ratio↑, p53↑, caspase-8, -9 activation, cleaved parp-1↑[124]
Flavanoneshesperetingastricbax↑, apaf-1↑, bcl-2↓, ROS↑, caspase-9, -3 activation, MMP↓, cytochrome c↑, DNA fragmentation[126]
esophagealROS↑, bax↑, apaf-1↑, SuFu↑, bcl-2↓, caspase-9, -3 activation[127]
colonROS↑, bax↑, bcl-2↓, cytochrome c↑, cleaved caspase-3 ↑, SOD↓, CAT↓, GPx↓[128]
breastROS↑, bax/bcl-2 ratio↑, phosphatydilserine externalization, caspase-9, -7 activation, MMP↓, cytochrome c↑,cleaved parp-1↑, DNA fragmentation[129]
lungFas↑, FADD↑, caspase-8 activation [130]
cytosolic bax↓, mitochondrial bax↑[131]
cervicalextrinsic and intrinsic apoptotic pathways[132]
Flavanols epicatechinbreast
DR4↑, DR5↑, ROS↑, MMP↓, cytochrome c↑, Smac/Diablo↑, HtrA2/Omi↑, TRAIL↑, bad↑, bax ↑[134]
bax↑, cytochrome c↑, caspase-3 activation[136]
colonbcl-2↓, bax↑, p53↑, DNA fragmentation[135]
prostatebax↑, cytochrome c↑, caspase-3 activation[136]
Anthocyaninscyanidinprostatecaspase-3 activation, DNA fragmentation[138]
glioblastomabax↑, p53↑, bcl-2↓[139]
osteosarcomabax↑, PPARγ↑, p21↑[140]
Non Flavonoids
Hydroxybenzoic acidsferulic acidbreastcaspase-8, -9 activation[142]
bax/bcl-2 ratio↑ [143]
hepaticcaspase-8, -9 activation[142]
cervicalbax↑, bcl-2↓, mcl-1↓[144]
Hydroxycinnamic acidsgallic acidgastricFas↑, FasL↑, DR5↑, caspase-8, -9, -3 activation, bad↑, bak↑, bcl-2↓, p53↑[145]
breastMMP↓, caspase-8, -9 activation, cytochrome c↑[146]
bax↑, bcl-2↓, caspase-3 ↑, p53↑[147]
LignanssecoisolariciresinolcolonAIF↑, caspase-3↑[149]
breastbcl-2↓[150]
Stilbenes pterostilbeneosteosarcomabax↑, bak↑, bcl-xL↓, mcl-1↓, caspase-3 ↑, cytochrome c↑[152]
breastsuperoxide anion↑, MMP↓, caspase-3, -7 activation[153]
bax↑, p53 mutant↓ [154]
bax↑[155]
endometrialbcl-2↓, bcl-xL↓, caspase-3 activation[156]
cervicalROS↑, bcl-2↓, bcl-xL↓, caspase-9, -3 activation[157]
liverROS↑, SOD2↓, p53↑[158]
CurcuminoidscurcuminleukemiaROS↑[161]
melanomaROS↑[162]
caspase-8 activation[173]
breastROS↑, TRAIL↑[163]
p53↑, bax↑[169]
bcl-2↓[171]
renalROS↑, TRAIL↑[164]
TRAIL↑[168]
glioblastomaTRAIL↑[165]
prostateTRAIL↑[166]
bcl-2↓[172]
hepatocellular carcinomaFasL↑
caspase-3 activation
[167]
laryngealbcl-2↓[170]
colonp53↑, bax↑[36]
neuroblastomacaspase-3 activation[174]
linfomacaspase-3 activation[175]
lungcaspase-3 activation[176]
schwannomabcl-2↓, bax↑, parp-1↑, caspase-3↑, caspase-9↑[177]
thyroidROS↑, caspase-3 activation, bcl-2↓, bcl-xL↓, bax↑[178]

4. Dietary Polyphenols and Drug Synergism in Cancer Therapy

In order to increase the chemopreventive effects of polyphenols, they are often combined with conventional chemotherapy drugs or even with other dietary polyphenols [179,180]. These associations enhance the efficacy of several chemotherapy agents or potentiate the anticancer activity of individual combined natural compounds [179,180] (Table 2). In particular, the improved anti-tumor activity deriving from pharmacological synergy with cytotoxic agents is associated with a lower chemotherapy toxicity in normal tissues, which could be due to an improvement of its pharmacokinetic and metabolic characteristics [179,180].
Genistein enhances the anticancer effect of cisplatin in CaSki cervical cancer cells [181]. The combination of genistein and cisplatin in these cells inhibited ERK1/2 phosphorylation, decreased bcl-2 protein expression levels and increased those of p53 and cleaved caspase-3 [181]. Moreover, it synergistically improved the anti-tumor action of antineoplastic centchroman (CC) in MCF-7 and MDA-MB-231 breast cancer cells [182]; this occurred by cell cycle arrest at the G2/M phase that culminated in ROS-dependent apoptosis. In particular, their combination determined bax and bcl-2 ratio dysregulation and mitochondrial dysfunction, together with caspase-3, -7 and -9 activation and parp-1 cleavage. Further, genistein and CC significantly inhibited PI3K/AKT/NF-κB phosphorylation, thus enhancing apoptosis [182].
Cisplatin cytotoxicity can be also enhanced by quercetin in HeLa and SiHa human cervical cancer cells [183]. Quercetin inhibited HeLa and SiHa cell viability in a dose- and time-dependent manner, and its combination with cisplatin had stronger cytotoxic effects than their individual effects. Besides, quercetin combined with cisplatin group induced more cell apoptosis in contrast to single treatment [183]. A similar drug synergism was also responsible for enhanced methotrexate chemotherapeutic efficacy against Saos-2 osteosarcoma [184]. The analysis of mRNA expression outcomes indicated that the combination therapy significantly upregulated p53, CBX7 and CYLD tumor suppressor genes and reduced anti-apoptotic genes bcl-2 and miR-223, which can lead to proliferation inhibition and apoptosis induction [184]. Moreover, a recent study demonstrated that quercetin, in combination with tamoxifen, controlled several apoptosis-related gene expression, including those of p53, bax, p21 and bcl-2 in MCF-7 breast cancer cells, leading to apoptosis regulation [185].
Studies performed on HONE1 and CNE2 human nasopharyngeal carcinoma cell lines revealed that apigenin significantly enhanced anti-tumor capacity of cetuximab by suppressing EGFR signaling in vivo and in vitro [186]. Results demonstrated that their combination could better suppress the viability and induce pro-apoptotic effects. In fact, the elevated bax/bcl-2 ratio and caspase-3 expression, as well as the decreased p-EGFR, p-AKT, p-STAT3 and cyclin D1 protein levels mediated by chemotherapy agent, were remarkably enhanced by apigenin in both cell lines [186]. Another work showed that apigenin, when used with cisplatin, reduced the proliferation of A549 lung, MCF-7 breast, HCT 116 colorectal, and HeLa cervical cancer cells; this effect was associated with increased cisplatin-induced DNA damage and apoptosis in a p53-dependent manner [187]. The combinatory cytotoxic effects of apigenin and paclitaxel was evaluated in HeLa cervical, A549 lung, Hep3B negroid hepatocyte and HEK293A embryonic kidney cancer cell lines [188]. Results indicated that combined treatment significantly enhanced paclitaxel cytotoxicity in all human cancer cells tested. Moreover, the ROS-mediated activation of caspase-2 and MMP decrease were essential for apigenin-/paclitaxel-induced apoptosis in HeLa cells [188]. In Jurkat lymphoid and THP-1 myeloid leukaemia cell lines, apigenin, in combination with etoposide or cyclophosphamide, induced apoptosis via the mitochondrial pathway by increasing cytochrome c, Smac/Diablo and HtrA2/Omi expression and by caspase-9 and -3 activation [189]. Moreover, the effects of several polyphenols (apigenin, quercetin, emodin and rhein) alone and in combination with three alkylating agents (cisplatin, cyclophosphamide and chlorambucil) were evaluated in Jurkat and CCRF-CEM lymphoid and THP1 and KG1a myeloid leukaemia cells lines [190]. In lymphoid leukaemia cell lines, a synergistic reduction in ATP and glutathione levels, a cell cycle arrest, DNA damage and apoptosis induction were observed by using quercetin, apigenin, emodin and rhein in combination with cisplatin and cyclophosphamide; the same effects are produced using apigenin and rhein with chlorambucil [190]. In myeloid leukemia cells, the combined use of apigenin, quercetin and emodin showed similar synergistic effects with all alkylating agents [190]. He and co-workers demonstrated that hesperetin was able to potentiate cisplatin-mediated apoptosis in HGC-27, SGC-7901 and MGC-803 human gastric cell lines [191]. In particular, this drug combination remarkably increased the expression of PTEN, cytosolic cytochrome c, bax, cleaved caspase-9 and -3 and AIF; moreover, p-AKT and bcl-2 protein expression levels were reduced [191]. The apoptosis- and oxidative-stress-related effects of cyanidin-3-O-glucoside, cisplatin and their combination were investigated on HeLa cervical cancer cells [192]. Results showed that cyanidin-3-O-glucoside and cisplatin increased oxidative stress by downregulating Nrf2 and consequently HO-1 and NQO1 expression. Furthermore, this combination promoted cell apoptosis by increasing the mRNA expression ratio of bax/bcl-2 [192]. The anti-tumor effects of cisplatin were also increased by gallic acid in A549 human NSCLC [193]. They were dependent on JAK/STAT3 signaling pathway modulation and changes in the expression of downstream apoptotic molecules, such as bax upregulation and bcl-2 downregulation [193].
Other studies determined the potential of pterostilbene to enhance gemcitabine tumor cytotoxicity and chemosensitivity in MIA PaCa-2 and MIA PaCa-2 GEMR (gemcitabine-resistant) pancreatic ductal adenocarcinoma cells [194]. In these cells, pterostilbene induced S-phase cell cycle arrest, apoptosis and autophagic cell death and inhibited multidrug resistance protein 1 (MDR1) expression by downregulating RAGE/PI3K/AKT signaling in both cell lines [194]. Another combination of pterostilbene and EGCG showed additive antiproliferative effects in MIA PaCa-2 and PANC-1 pancreatic cancer cell lines [195]. This combination induced apoptosis by mitochondrial depolarization and cytochrome c release increase in MIA PaCa-2 but not in PANC-1 cells. Moreover, while EGCG alone increased caspase-3/7 activity in MIA PaCa-2 cells, the same association did not significantly cause this effect in either cell line [195].
Several reports demonstrated the synergistic effects of several chemotherapeutic drugs in combination with curcumin. Curcumin combined with 5-fluorouracil (5-FU) or doxorubicin (DOX) chemosensitized the NT8e HNSCC cells towards apoptosis and promoted cell proliferation inhibition [196]. In particular, these combinations reduced cell proliferation through EGFR/ERK1/2 signaling downregulation and caused cell cycle arrest at the G1/S phase; moreover, apoptosis occurred by inhibiting bcl-2, increasing bax and cleaved parp-1 expression and activating caspase-3 [196]. Therefore, in MCF-7 breast cancer cells, the simultaneous treatment of paclitaxel with curcumin exhibited synergistic growth inhibition through EGFR signaling modulation and induced significant apoptosis as demonstrated by bax increase and bcl-2 protein expression decrease [197]. Cell death induced by curcumin and paclitaxel was also evaluated in both MCF-7 and MDA-MB-231 human breast cancer cell lines [198]. Results confirmed that this combination resulted in apoptosis induction as evidenced by caspase-3 and cleaved parp-1 expression increase; these effects were more evident when curcumin was used in combination than when used alone [198]. The ability of curcumin to improve the paclitaxel-induced apoptosis was also demonstrated in CaSki and HeLa human cervical cancer cell lines wherein the anti-tumor effects were associated with the NF-κB signaling inhibition together with p53 and caspase-3 expression increase [199]. Recently, a synergistic induction of apoptosis by quercetin and curcumin was observed in K562 chronic myeloid leukemia cells [200]. Gene expression analysis revealed that their combination was effective on genes that were particularly related to p53, NF-κB, AKT1, FasL and TGF-α pathways; in particular CDKN1B, AKT1 and IFN-γ are downregulated, while BTG2, p21, p27, CDKN1A, Fas and FasL are upregulated in this cell model [200].
Table 2. Anti-cancer effects of the dietary polyphenol and/or chemotherapy drug combination. ↑: increase, ↓: decrease.
Table 2. Anti-cancer effects of the dietary polyphenol and/or chemotherapy drug combination. ↑: increase, ↓: decrease.
Phytochemical Compounds/
Chemotherapy Drugs
Cell LinesOutcomesReferences
genistein+cisplatinCaSki cervical cancerERK1/2 phosphorylation↓, bcl-2↓, p53↑, cleaved caspase-3↑[181]
genistein+centchromanMCF-7, MDA-MB-231 breast cancerPI3K/AKT/NF-κB phosphorylation↓, cell cycle arrest at G2/M phase, ROS↑, bax↑, bcl-2↓, caspase-3, -7, -9↑, cleaved parp-1↑[182]
quercetin+cisplatinHeLa, SiHa cervical cancercell viability↓, apoptosis↑[183]
quercetin+methotrexateSaos-2 osteosarcomacell viability↓, apoptosis↑, p53↑, CBX7↑, CYLD ↑, bcl-2↓, miR-223↓[184]
quercetin+tamoxifenMCF-7 breast cancerapoptosis↑, p53↑, bax↑, p21↑, and bcl-2↓[185]
apigenin+cetuximabHONE1, CNE2 nasopharyngeal carcinoma
cell viability↓, apoptosis↑, bax/bcl-2 ratio↑, caspase-3↑, p-EGFR↓, p-AKT↓, p-STAT3↓, cyclin D1↓[186]
apigenin+cisplatinA549 lung, MCF-7 breast, HCT 116 colorectal, HeLa cervical cancerscell viability↓, apoptosis↑, p53↑, DNA damage↑[187]
apigenin+paclitaxelHeLa cervical, A549 lung, Hep3B negroid hepatocyte, HEK293A embryonic kidney cancercell viability↓[188]
HeLa cervical cancer apoptosis↑, ROS↑, caspase-2↑, MMP ↓ [188]
apigenin+etoposide apigenin+cyclophosphamideJurkat lymphoid and THP-1 myeloid leukaemiaapoptosis↑, cytochrome c↑, Smac/Diablo↑, HtrA2/Omi ↑, caspase-9, -3 activation[189]
apigenin+quercetin+emodin+rhein+
cisplatin and cyclophosphamide
apigenin+rhein+ chlorambucil
Jurkat and CCRF-CEM lymphoid leukaemiaapoptosis↑, ATP↓, glutathione↓, cell cycle arrest, DNA damage↑ [190]
apigenin+quercetin,+emodin+cisplatin, cyclophosphamide and chlorambucilTHP1 and KG1a myeloid leukaemiaapoptosis↑, ATP↓, glutathione↓, cell cycle arrest, DNA damage↑[190]
hesperetin+cisplatinHGC-27, SGC-7901, and MGC-803 gastricPTEN↑, cytosolic cytochrome c↑, bax↑, cleaved caspase-9 and -3↑, AIF↑, p-AKT↓, bcl-2↓[191]
cyanidin-3-O-glucoside+ cisplatinHeLa cervical cancer Nrf2↓, HO-1↓, NQO1↓, bax/bcl-2 ratio↑[192]
gallic acid+cisplatinA549 non-small cell lung cancer JAK/STAT3↓, bax↑, bcl-2↓[193]
pterostilbene+gemcitabineMIA PaCa-2, MIA PaCa-2 GEMR (gemcitabine-resistant) pancreatic ductal adenocarcinomaS-phase cell cycle arrest, apoptosis↑, MDR1↓, RAGE/PI3K/AKT signaling↓[194]
pterostilbene+epigallocatechin gallateMIA PaCa-2 pancreatic cancercell proliferation↓, mitochondrial depolarization, cytochrome c release↑[195]
PANC-1 pancreatic cancercell proliferation↓[195]
curcumin+5-fluorouracil or doxorubicinNT8e head and neck squamous cell carcinomacell proliferation↓, EGFR/ERK1/2 signaling↓, G1/S cell cycle arrest, apoptosis↑, bcl-2↓, bax↑, caspase-3 activation, cleaved parp-1↑[196]
curcumin+paclitaxelMCF-7 breast cancercell proliferation↓, EGFR signaling↓, apoptosis↑, bcl-2↓, bax↑[197]
curcumin+paclitaxelMCF-7, MDA-MB-231 breast cancerapoptosis↑, caspase-3↑, cleaved parp-1↑[198]
curcumin+paclitaxelCaSki, HeLa cervical cancerNF-κB signaling↓, p53↑, caspase-3↑[199]
curcumin+quercetinK562 chronic myeloid leukemiaCDKN1B↓, AKT1↓, IFN-γ↓, BTG2↑, p21↑, p27↑, CDKN1A↑, Fas↑, FasL↑[200]

5. Conclusions

Apoptosis is a protective mechanism against neoplastic transformation, as it is able to eliminate genetically damaged cells. It has been reported that its dysregulation is a biological event involved in the cancer etiopathology. In this regard, the discovery of natural compounds able to induce this death mechanism in tumor cells could be of great importance for new anticancer therapies. In recent decades, dietary polyphenols have attracted much interest due to their ability to act as both effective chemopreventive and chemotherapeutic agents. However, the anti-tumor effects observed in vitro are very often different from those obtained in vivo due to their poor bioavailability. From in vitro studies analysis, it emerged that polyphenols are able to interfere with all stages of carcinogenesis process by modulating different cellular pathways and inducing and/or inhibiting the expression of several proteins related to proliferation, migration, metastasis and apoptosis. Multifaceted actions of dietary flavonoid and non-flavonoid compounds on cell death signals modulation that involves both extrinsic and intrinsic apoptotic pathways were reported. Moreover, the polyphenols’ chemopreventive effects greatly depend on the natural compound concentration and specific tumor cell type. Interestingly, their beneficial actions increased when combined with conventional chemotherapy drugs or other dietary polyphenols. In conclusion, we believe that the increasingly detailed knowledge of the molecular apoptotic mechanisms induced by polyphenols will be very important to design new therapeutic strategies useful for cancer treatment involving polyphenols as adjuvants.

Author Contributions

Conceptualization, original draft preparation, writing, search and identification of articles: A.C.; figures and tables preparation: A.C., A.D.L. and M.D.; editing: A.C., A.D.L., M.D., F.D.A. and V.P.; supervision: A.C., F.D.A. and V.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by MIUR ex 60% (A.C., F.D.A., V.P.) and by AIRC (Associazione Italiana per la Ricerca sul Cancro), project n. IG20122.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AGSgastric adenocarcinoma
AKTprotein-chinasi B
APAF-1apoptotic protease activating factor-1
ATF6activating transcription factor 6
ATPadenosine triphosphate
BADBCL-2 associated agonist of cell death
BAKBCL-2 antagonist/killer
BAXBCL-2-associated X protein
BCL-2B-cell lymphoma-2
BCL-wB-cell lymphoma-w
BCL-XLB-cell lymphoma-extra large
BFL-1/A1Bcl-2-related protein A1
BICbone-inducing protein complex
BIMBCL-2-interacting mediator of cell death
BMFBCL-2 modifying factor
BOCcell adhesion associated, oncogene regulated
BTG2BTG family member 2
c-XIAP1cellular inhibitor of apoptosis protein 1
c-XIAP2cellular inhibitor of apoptosis protein 2
CASPASEcysteine-dependent aspartate-directed protease
CATcatalase
CBX7chromobox protein homolog 7
CDKN1Acyclin dependent kinase inhibitor 1A
CDKN1Bcyclin-dependent kinase inhibitor 1B
c-IAP1cellular inhibitor of apoptosis protein 1
COX2cyclooxygenase 2
CRADDCASP2 and RIPK1 domain containing adaptor with death domain
CYLDubiquitin carboxyl-terminal hydrolase
DEDDdeath effector domain containing protein
DNAdeoxyribonucleic acid
DOXdoxorubicin
DRdeath receptor
DR-5death receptor 5
EGCGepigallocatechin gallate
EGFRepidermal growth factor receptor
ERestrogen receptor
ERKextracellular signal-regulated kinase
ERK1/2extracellular signal-regulated kinase 1
ERαestrogen receptor alpha
ERβestrogen receptor beta
FADDfas-associated death domain
FASfas cell surface death receptor
FASLfas ligand
FOXO3aforkhead box O3a
GPxglutathione peroxidase
GRP78glucose-regulated protein 78
GSHreduced glutathione
HNSCChead and neck squamous cell carcinoma
HO-1heme oxygenase-1
HONE1epithelial tumor cell lines 1
HRKharakiri, BCL-2 interacting protein
HTRA2high-temperature requirement protein A2
IFN-γinterferon gamma
IGF-IRtype 1 insulin-like growth factor receptor
JAKjanus kinase
JNKc-Jun N-terminal kinase
LPHphloridzine hydrolase
MAPKmitogen-activated protein kinase
MCL-1myeloid cell leukemia-1
MDR1multidrug resistance protein 1
MMPmitochondrial membrane potential
mTORmammalian target of rapamycin
NACN-acetylcysteine
ncRNAsnon-coding RNAs
NF-κBnuclear factor kappa B
NOXAphorbol-12-myristate-13-acetate-induced protein 1
NQO1NAD(P)H quinone oxidoreductase
NRF2nuclear factor erythroid 2–related factor 2
NSCLCnon-small cell lung cancer
PARP-1poly [ADP-ribose] polymerase 1
PI3Kphoshatidylinositol-3 kinase
PKCprotein kinase C
PPARγperoxisome proliferator-activated receptor gamma
PTCpapillary thyroid cancer
PTENphosphatase and tensin homolog
PTKprotein tyrosine kinase
PUMAp53-up-regulated modulator of apoptosis
RAGEreceptor for advanced glycation end products
ROSreactive oxygen species
SMAC/DIABLOsecond mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low isoelectric point
SODsuperoxide dismutase
STATsignal transducer and activator of transcription
STAT3signal transducer and activator of transcription 3
TNFtumor necrosis factor
TNF-αtumor necrosis factor alpha
TNF-Rtumor necrosis factor receptor
TNFRSF10Atumor necrosis factor receptor superfamily 10A
TNFRSF10Btumor necrosis factor receptor superfamily 10B
TNFRSF1Btumor necrosis factor receptor superfamily member 1B
TNFRSF21tumor necrosis factor receptor superfamily member 21
TNFRSF25tumor necrosis factor receptor superfamily member 25
TNFSF10tumor necrosis factor (ligand) superfamily member 10
TRADDtumor necrosis factor receptor type 1-associated death domain protein
TRAILTNF-related apoptosis-inducing ligand
TRAIL-RTNF-related apoptosis-inducing ligand receptor
XIAPX-linked inhibitor of apoptosis
ZIP9Zrt- and Irt-like protein 9

References

  1. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  2. Global Burden of Disease 2019 Cancer Collaboration; Kocarnik, J.M.; Compton, K.; Dean, F.E.; Fu, W.; Gaw, B.L.; Harvey, J.D.; Henrikson, H.J.; Lu, D.; Pennini, A.; et al. Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life Years for 29 Cancer Groups From 2010 to 2019: A Systematic Analysis for the Global Burden of Disease Study 2019. JAMA Oncol. 2022, 8, 420–444. [Google Scholar] [CrossRef] [PubMed]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  4. Ansari, K.K.; Jha, A. Causes of Cancer in the World: Comparative Risk Assessment of Nine Behavioral and Environmental Risk Factors. Cureus 2022, 14, e28875. [Google Scholar] [CrossRef]
  5. Emanuele Liardo, R.L.; Borzi, A.M.; Spatola, C.; Martino, B.; Privitera, G.; Basile, F.; Biondi, A.; Vacante, M. Effects of infections on the pathogenesis of cancer. Indian J. Med. Res. 2021, 153, 431–445. [Google Scholar] [CrossRef] [PubMed]
  6. Neben, C.L.; Zimmer, A.D.; Stedden, W.; van den Akker, J.; O’Connor, R.; Chan, R.C.; Chen, E.; Tan, Z.; Leon, A.; Ji, J.; et al. Multi-Gene Panel Testing of 23,179 Individuals for Hereditary Cancer Risk Identifies Pathogenic Variant Carriers Missed by Current Genetic Testing Guidelines. J. Mol. Diagn. 2019, 21, 646–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. GBD 2019 Cancer Risk Factors Collaborators. The global burden of cancer attributable to risk factors, 2010–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2022, 400, 563–591. [Google Scholar] [CrossRef]
  8. Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef] [Green Version]
  9. Fernald, K.; Kurokawa, M. Evading apoptosis in cancer. Trends Cell Biol. 2013, 23, 620–633. [Google Scholar] [CrossRef] [Green Version]
  10. Jan, R.; Chaudhry, G.E.S. Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics. Adv. Pharm. Bull. 2019, 9, 205–218. [Google Scholar] [CrossRef]
  11. Vali-Pour, M.; Lehner, B.; Supek, F. The impact of rare germline variants on human somatic mutation processes. Nat. Commun. 2022, 13, 3724. [Google Scholar] [CrossRef] [PubMed]
  12. Levatic, J.; Salvadores, M.; Fuster-Tormo, F.; Supek, F. Mutational signatures are markers of drug sensitivity of cancer cells. Nat. Commun. 2022, 13, 2926. [Google Scholar] [CrossRef] [PubMed]
  13. Zhivotovsky, B.; Kroemer, G. Apoptosis and genomic instability. Nat. Rev. Mol. Cell Biol. 2004, 5, 752–762. [Google Scholar] [CrossRef] [PubMed]
  14. Bonora, M.; Missiroli, S.; Perrone, M.; Fiorica, F.; Pinton, P.; Giorgi, C. Mitochondrial Control of Genomic Instability in Cancer. Cancers 2021, 13, 1914. [Google Scholar] [CrossRef] [PubMed]
  15. DePinho, R.A. The age of cancer. Nature 2000, 408, 248–254. [Google Scholar] [CrossRef]
  16. Konishi, A.; Shimizu, S.; Hirota, J.; Takao, T.; Fan, Y.; Matsuoka, Y.; Zhang, L.; Yoneda, Y.; Fujii, Y.; Skoultchi, A.I.; et al. Involvement of histone H1.2 in apoptosis induced by DNA double-strand breaks. Cell 2003, 114, 673–688. [Google Scholar] [CrossRef] [Green Version]
  17. Storchova, Z.; Pellman, D. From polyploidy to aneuploidy, genome instability and cancer. Nat. Rev. Mol. Cell Biol. 2004, 5, 45–54. [Google Scholar] [CrossRef]
  18. Chen, J.G.; Yang, C.P.; Cammer, M.; Horwitz, S.B. Gene expression and mitotic exit induced by microtubule-stabilizing drugs. Cancer Res. 2003, 63, 7891–7899. [Google Scholar]
  19. Rallis, K.S.; Lai Yau, T.H.; Sideris, M. Chemoradiotherapy in Cancer Treatment: Rationale and Clinical Applications. Anticancer Res. 2021, 41, 1–7. [Google Scholar] [CrossRef]
  20. Kocakavuk, E.; Anderson, K.J.; Varn, F.S.; Johnson, K.C.; Amin, S.B.; Sulman, E.P.; Lolkema, M.P.; Barthel, F.P.; Verhaak, R.G.W. Radiotherapy is associated with a deletion signature that contributes to poor outcomes in patients with cancer. Nat. Genet. 2021, 53, 1088–1096. [Google Scholar] [CrossRef]
  21. Liu, Y.P.; Zheng, C.C.; Huang, Y.N.; He, M.L.; Xu, W.W.; Li, B. Molecular mechanisms of chemo- and radiotherapy resistance and the potential implications for cancer treatment. Med. Comm. 2021, 2, 315–340. [Google Scholar] [CrossRef] [PubMed]
  22. Maleki Dana, P.; Sadoughi, F.; Asemi, Z.; Yousefi, B. The role of polyphenols in overcoming cancer drug resistance: A comprehensive review. Cell. Mol. Biol. Lett. 2022, 27, 1. [Google Scholar] [CrossRef] [PubMed]
  23. Chairez-Ramirez, M.H.; de la Cruz-Lopez, K.G.; Garcia-Carranca, A. Polyphenols as Antitumor Agents Targeting Key Players in Cancer-Driving Signaling Pathways. Front. Pharmacol. 2021, 12, 710304. [Google Scholar] [CrossRef] [PubMed]
  24. Montane, X.; Kowalczyk, O.; Reig-Vano, B.; Bajek, A.; Roszkowski, K.; Tomczyk, R.; Pawliszak, W.; Giamberini, M.; Mocek-Plociniak, A.; Tylkowski, B. Current Perspectives of the Applications of Polyphenols and Flavonoids in Cancer Therapy. Molecules 2020, 25, 3342. [Google Scholar] [CrossRef]
  25. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights Into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef]
  26. Gasmi, A.; Mujawdiya, P.K.; Noor, S.; Lysiuk, R.; Darmohray, R.; Piscopo, S.; Lenchyk, L.; Antonyak, H.; Dehtiarova, K.; Shanaida, M.; et al. Polyphenols in Metabolic Diseases. Molecules 2022, 27, 6280. [Google Scholar] [CrossRef]
  27. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [Green Version]
  28. Zhao, Y.; Zhang, X. Interactions of tea polyphenols with intestinal microbiota and their implication for anti-obesity. J. Sci. Food Agric. 2020, 100, 897–903. [Google Scholar] [CrossRef]
  29. Grosso, G.; Godos, J.; Lamuela-Raventos, R.; Ray, S.; Micek, A.; Pajak, A.; Sciacca, S.; D’Orazio, N.; Del Rio, D.; Galvano, F. A comprehensive meta-analysis on dietary flavonoid and lignan intake and cancer risk: Level of evidence and limitations. Mol. Nutr. Food Res. 2017, 61, 1600930. [Google Scholar] [CrossRef]
  30. Messina, M. Impact of Soy Foods on the Development of Breast Cancer and the Prognosis of Breast Cancer Patients. Forsch. Komplementmed. 2016, 23, 75–80. [Google Scholar] [CrossRef]
  31. Gardeazabal, I.; Romanos-Nanclares, A.; Martinez-Gonzalez, M.A.; Sanchez-Bayona, R.; Vitelli-Storelli, F.; Gaforio, J.J.; Aramendia-Beitia, J.M.; Toledo, E. Total polyphenol intake and breast cancer risk in the Seguimiento Universidad de Navarra (SUN) cohort. Br. J. Nutr. 2019, 122, 542–551. [Google Scholar] [CrossRef] [PubMed]
  32. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef] [PubMed]
  33. Jang, Y.G.; Go, R.E.; Hwang, K.A.; Choi, K.C. Resveratrol inhibits DHT-induced progression of prostate cancer cell line through interfering with the AR and CXCR4 pathway. J. Steroid Biochem. 2019, 192, 105406. [Google Scholar] [CrossRef] [PubMed]
  34. Deb, G.; Shankar, E.; Thakur, V.S.; Ponsky, L.E.; Bodner, D.R.; Fu, P.F.; Gupta, S. Green tea-induced epigenetic reactivation of tissue inhibitor of matrix metalloproteinase-3 suppresses prostate cancer progression through histone-modifying enzymes. Mol. Carcinog. 2019, 58, 1194–1207. [Google Scholar] [CrossRef] [PubMed]
  35. Yuan, L.; Zhou, M.M.; Huang, D.W.; Wasan, H.S.; Zhang, K.; Sun, L.T.; Huang, H.; Ma, S.L.; Shen, M.H.; Ruan, S.M. Resveratrol inhibits the invasion and metastasis of colon cancer through reversal of epithelial-mesenchymal transition via the AKT/GSK-3 beta/Snail signaling pathway. Mol. Med. Rep. 2022, 26, 354. [Google Scholar] [CrossRef]
  36. Li, P.H.; Pu, S.M.; Lin, C.; He, L.; Zhao, H.X.; Yang, C.; Guo, Z.Q.; Xu, S.S.; Zhou, Z.P. Curcumin selectively induces colon cancer cell apoptosis and S cell cycle arrest by regulates Rb/E2F/p53 pathway. J. Mol. Struct. 2022, 1263, 133180. [Google Scholar] [CrossRef]
  37. Zan, L.L.; Chen, Q.F.; Zhang, L.; Li, X.N. Epigallocatechin gallate (EGCG) suppresses growth and tumorigenicity in breast cancer cells by downregulation of miR-25. Bioengineered 2019, 10, 374–382. [Google Scholar] [CrossRef] [Green Version]
  38. Bayat, S.; Derakhshan, S.M.; Derakhshan, N.M.; Khaniani, M.S.; Alivand, M.R. Downregulation of HDAC2 and HDAC3 via oleuropein as a potent prevention and therapeutic agent in MCF-7 breast cancer cells. J. Cell. Biochem. 2019, 120, 9172–9180. [Google Scholar] [CrossRef]
  39. Alsamri, H.; Alneyadi, A.; Muhammad, K.; Ayoub, M.A.; Eid, A.; Iratni, R. Carnosol Induces p38-Mediated ER Stress Response and Autophagy in Human Breast Cancer Cells. Front. Oncol. 2022, 12, 911615. [Google Scholar] [CrossRef]
  40. Wu, M.F.; Huang, Y.H.; Chiu, L.Y.; Cherng, S.H.; Sheu, G.T.; Yang, T.Y. Curcumin Induces Apoptosis of Chemoresistant Lung Cancer Cells via ROS-Regulated p38 MAPK Phosphorylation. Int. J. Mol. Sci. 2022, 23, 8248. [Google Scholar] [CrossRef]
  41. Sun, X.L.; Xiang, Z.M.; Xie, Y.R.; Zhang, N.; Wang, L.X.; Wu, Y.L.; Zhang, D.Y.; Wang, X.J.; Sheng, J.; Zi, C.T. Dimeric-(−)-epigallocatechin-3-gallate inhibits the proliferation of lung cancer cells by inhibiting the EGFR signaling pathway. Chem.-Biol. Interact. 2022, 365, 110084. [Google Scholar] [CrossRef] [PubMed]
  42. Barros, T.M.B.; Lima, A.P.B.; Almeida, T.C.; da Silva, G.N. Inhibition of urinary bladder cancer cell proliferation by silibinin. Environ. Mol. Mutagen. 2020, 61, 445–455. [Google Scholar] [CrossRef] [PubMed]
  43. Adami, B.S.; Diz, F.M.; Goncalves, G.P.O.; Reghelin, C.K.; Scherer, M.; Dutra, A.P.; Papaleo, R.M.; de Oliveira, J.R.; Morrone, F.B.; Wieck, A.; et al. Morphological and mechanical changes induced by quercetin in human T24 bladder cancer cells. Micron 2021, 151, 103152. [Google Scholar] [CrossRef] [PubMed]
  44. Wei, R.; Penso, N.E.C.; Hackman, R.M.; Wang, Y.F.; Mackenzie, G.G. Epigallocatechin-3-Gallate (EGCG) Suppresses Pancreatic Cancer Cell Growth, Invasion, and Migration partly through the Inhibition of Akt Pathway and Epithelial-Mesenchymal Transition: Enhanced Efficacy When Combined with Gemcitabine. Nutrients 2019, 11, 1856. [Google Scholar] [CrossRef] [Green Version]
  45. Ratajczak, K.; Glatzel-Plucinska, N.; Ratajczak-Wielgomas, K.; Nowinska, K.; Borska, S. Effect of Resveratrol Treatment on Human Pancreatic Cancer Cells through Alterations of Bcl-2 Family Members. Molecules 2021, 26, 6560. [Google Scholar] [CrossRef]
  46. Jokar, M.H.; Sedighi, S.; Moradzadeh, M. A comparative study of anti-leukemic effects of kaempferol and epigallocatechin-3-gallate (EGCG) on human leukemia HL-60 cells. Avicenna J. Phytomed. 2021, 11, 314–323. [Google Scholar] [CrossRef]
  47. Kuttikrishnan, S.; Siveen, K.S.; Prabhu, K.S.; Khan, A.Q.; Ahmed, E.I.; Akhtar, S.; Ali, T.A.; Merhi, M.; Dermime, S.; Steinhoff, M.; et al. Curcumin Induces Apoptotic Cell Death via Inhibition of PI3-Kinase/AKT Pathway in B-Precursor Acute Lymphoblastic Leukemia. Front. Oncol. 2019, 9, 484. [Google Scholar] [CrossRef] [Green Version]
  48. Dong, C.Q.; Wang, Z.G.; Shen, P.; Chen, Y.G.; Wang, J.S.; Wang, H.B. Epigallocatechin-3-gallate suppresses the growth of human osteosarcoma by inhibiting the Wnt/beta-catenin signalling pathway. Bioengineered 2022, 13, 8490–8502. [Google Scholar] [CrossRef]
  49. De Luca, A.; Bellavia, D.; Raimondi, L.; Carina, V.; Costa, V.; Fini, M.; Giavaresi, G. Multiple Effects of Resveratrol on Osteosarcoma Cell Lines. Pharmaceuticals 2022, 15, 342. [Google Scholar] [CrossRef]
  50. Qin, J.L.; Fu, M.L.; Wang, J.; Huang, F.X.; Liu, H.P.; Huangfu, M.J.; Yu, D.; Liu, H.W.; Li, X.M.; Guan, X.; et al. PTEN/AKT/mTOR signaling mediates anticancer effects of epigallocatechin-3-gallate in ovarian cancer. Oncol. Rep. 2020, 43, 1885–1896. [Google Scholar] [CrossRef]
  51. Sun, S.F.; Fang, H.L. Curcumin inhibits ovarian cancer progression by regulating circ-PLEKHM3/miR-320a/SMG1 axis. J. Ovarian Res. 2021, 14, 158. [Google Scholar] [CrossRef] [PubMed]
  52. Dei Cas, M.; Ghidoni, R. Cancer Prevention and Therapy with Polyphenols: Sphingolipid-Mediated Mechanisms. Nutrients 2018, 10, 940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Bhosale, P.B.; Ha, S.E.; Vetrivel, P.; Kim, H.H.; Kim, S.M.; Kim, G.S. Functions of polyphenols and its anticancer properties in biomedical research: A narrative review. Transl. Cancer Res. 2020, 9, 7619–7631. [Google Scholar] [CrossRef] [PubMed]
  54. Selvakumar, P.; Badgeley, A.; Murphy, P.; Anwar, H.; Sharma, U.; Lawrence, K.; Lakshmikuttyamma, A. Flavonoids and Other Polyphenols Act as Epigenetic Modifiers in Breast Cancer. Nutrients 2020, 12, 761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Curti, V.; Di Lorenzo, A.; Dacrema, M.; Xiao, J.B.; Nabavi, S.M.; Daglia, M. In vitro polyphenol effects on apoptosis: An update of literature data. Semin. Cancer Biol. 2017, 46, 119–131. [Google Scholar] [CrossRef]
  56. Antonsson, B.; Martinou, J.C. The Bcl-2 protein family. Exp. Cell. Res. 2000, 256, 50–57. [Google Scholar] [CrossRef]
  57. Li, J.; Yuan, J. Caspases in apoptosis and beyond. Oncogene 2008, 27, 6194–6206. [Google Scholar] [CrossRef] [Green Version]
  58. Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef] [Green Version]
  59. Singla, R.K.; Dubey, A.K.; Garg, A.; Sharma, R.K.; Fiorino, M.; Ameen, S.M.; Haddad, M.A.; Al-Hiary, M. Natural Polyphenols: Chemical Classification, Definition of Classes, Subcategories, and Structures. J. Aoac. Int. 2019, 102, 1397–1400. [Google Scholar] [CrossRef]
  60. Lopez-Lazaro, M. Flavonoids as anticancer agents: Structure-activity relationship study. Curr. Med. Chem. Anticancer Agents 2002, 2, 691–714. [Google Scholar] [CrossRef]
  61. Khare, S.; Dewangan, R.P.; Kumar, A. Structure-Activity Relationship of Flavonoids: Recent Updates. In The Chemistry Inside Spices & Herbs: Research and Development; Bentham Science Publishers: Sharjah, United Arab Emirates, 2022; pp. 235–256. [Google Scholar] [CrossRef]
  62. Kim, K.; Vance, T.M.; Chun, O.K. Estimated intake and major food sources of flavonoids among US adults: Changes between 1999–2002 and 2007–2010 in NHANES. Eur. J. Nutr. 2016, 55, 833–843. [Google Scholar] [CrossRef] [PubMed]
  63. Janabi, A.H.W.; Kamboh, A.A.; Saeed, M.; Lu, X.Y.; BiBi, J.; Majeed, F.; Naveed, M.; Mughal, M.J.; Korejo, N.A.; Kamboh, R.; et al. Flavonoid-rich foods (FRF): A promising nutraceutical approach against lifespan-shortening diseases. Iran. J. Basic Med. Sci 2020, 23, 140–153. [Google Scholar] [CrossRef]
  64. Zaheer, K.; Humayoun Akhtar, M. An updated review of dietary isoflavones: Nutrition, processing, bioavailability and impacts on human health. Crit. Rev. Food Sci. Nutr. 2017, 57, 1280–1293. [Google Scholar] [CrossRef] [PubMed]
  65. Barreca, D.; Trombetta, D.; Smeriglio, A.; Mandalari, G.; Romeo, O.; Felice, M.R.; Gattuso, G.; Nabavi, S.M. Food flavonols: Nutraceuticals with complex health benefits and functionalities. Trends Food Sci. Tech. 2021, 117, 194–204. [Google Scholar] [CrossRef]
  66. Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. 2017, 8, 423–435. [Google Scholar] [CrossRef] [Green Version]
  67. Khan, M.K.; Dangles, O. A comprehensive review on flavanones, the major citrus polyphenols. J. Food Compos. Anal. 2014, 33, 85–104. [Google Scholar] [CrossRef]
  68. Luo, Y.; Jian, Y.Q.; Liu, Y.K.; Jiang, S.; Muhammad, D.; Wang, W. Flavanols from Nature: A Phytochemistry and Biological Activity Review. Molecules 2022, 27, 719. [Google Scholar] [CrossRef]
  69. Abudureheman, B.; Yu, X.; Fang, D.; Zhang, H. Enzymatic Oxidation of Tea Catechins and Its Mechanism. Molecules 2022, 27, 942. [Google Scholar] [CrossRef]
  70. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [Green Version]
  71. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef]
  72. Rodriguez-Garcia, C.; Sanchez-Quesada, C.; Toledo, E.; Delgado-Rodriguez, M.; Gaforio, J.J. Naturally Lignan-Rich Foods: A Dietary Tool for Health Promotion? Molecules 2019, 24, 917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Teka, T.; Zhang, L.; Ge, X.; Li, Y.; Han, L.; Yan, X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022, 197, 113128. [Google Scholar] [CrossRef]
  74. Ramesh, T.N.; Paul, M.; Manikanta, K.; Girish, K.S. Structure and morphological studies of curcuminoids and curcuminoid mixture. J. Cryst. Growth 2020, 547, 125812. [Google Scholar] [CrossRef]
  75. D’Archivio, M.; Filesi, C.; Vari, R.; Scazzocchio, B.; Masella, R. Bioavailability of the Polyphenols: Status and Controversies. Int. J. Mol. Sci. 2010, 11, 1321–1342. [Google Scholar] [CrossRef] [PubMed]
  76. Day, A.J.; Canada, F.J.; Diaz, J.C.; Kroon, P.A.; Mclauchlan, R.; Faulds, C.B.; Plumb, G.W.; Morgan, M.R.A.; Williamson, G. Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. Febs Lett. 2000, 468, 166–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ray, S.K.; Mukherjee, S. Evolving Interplay Between Dietary Polyphenols and Gut Microbiota-An Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef] [PubMed]
  78. Nocito, M.C.; De Luca, A.; Prestia, F.; Avena, P.; La Padula, D.; Zavaglia, L.; Sirianni, R.; Casaburi, I.; Puoci, F.; Chimento, A.; et al. Antitumoral Activities of Curcumin and Recent Advances to ImProve Its Oral Bioavailability. Biomedicines 2021, 9, 1476. [Google Scholar] [CrossRef]
  79. Chimento, A.; De Amicis, F.; Sirianni, R.; Sinicropi, M.S.; Puoci, F.; Casaburi, I.; Saturnino, C.; Pezzi, V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. Int. J. Mol. Sci. 2019, 20, 1381. [Google Scholar] [CrossRef] [Green Version]
  80. Enaru, B.; Socaci, S.; Farcas, A.; Socaciu, C.; Danciu, C.; Stanila, A.; Diaconeasa, Z. Novel Delivery Systems of Polyphenols and Their Potential Health Benefits. Pharmaceuticals 2021, 14, 946. [Google Scholar] [CrossRef]
  81. Sharma, E.; Attri, D.C.; Sati, P.; Dhyani, P.; Szopa, A.; Sharifi-Rad, J.; Hano, C.; Calina, D.; Cho, W.C. Recent updates on anticancer mechanisms of polyphenols. Front. Cell Dev. Biol. 2022, 10, 1005910. [Google Scholar] [CrossRef]
  82. Suraweera, T.L.; Rupasinghe, H.P.V.; Dellaire, G.; Xu, Z.L. Regulation of Nrf2/ARE Pathway by Dietary Flavonoids: A Friend or Foe for Cancer Management? Antioxidants 2020, 9, 973. [Google Scholar] [CrossRef] [PubMed]
  83. Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Busselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2019, 11, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hazafa, A.; Iqbal, M.O.; Javaid, U.; Tareen, M.B.K.; Amna, D.; Ramzan, A.; Piracha, S.; Naeem, M. Inhibitory effect of polyphenols (phenolic acids, lignans, and stilbenes) on cancer by regulating signal transduction pathways: A review. Clin. Transl. Oncol. 2022, 24, 432–445. [Google Scholar] [CrossRef] [PubMed]
  85. Hazafa, A.; Rehman, K.U.; Jahan, N.; Jabeen, Z. The Role of Polyphenol (Flavonoids) Compounds in the Treatment of Cancer Cells. Nutr. Cancer 2020, 72, 386–397. [Google Scholar] [CrossRef] [PubMed]
  86. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  87. Hengartner, M.O. The biochemistry of apoptosis. Nature 2000, 407, 770–776. [Google Scholar] [CrossRef]
  88. Kale, J.; Osterlund, E.J.; Andrews, D.W. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death Differ. 2018, 25, 65–80. [Google Scholar] [CrossRef] [Green Version]
  89. Chimento, A.; De Luca, A.; Avena, P.; De Amicis, F.; Casaburi, I.; Sirianni, R.; Pezzi, V. Estrogen Receptors-Mediated Apoptosis in Hormone-Dependent Cancers. Int. J. Mol. Sci. 2022, 23, 1242. [Google Scholar] [CrossRef]
  90. Fridman, J.S.; Lowe, S.W. Control of apoptosis by p53. Oncogene 2003, 22, 9030–9040. [Google Scholar] [CrossRef] [Green Version]
  91. Singh, K.K. Mitochondria damage checkpoint in apoptosis and genome stability. Fems Yeast Res. 2004, 5, 127–132. [Google Scholar] [CrossRef]
  92. Rapoport, B.L.; Anderson, R. Realizing the Clinical Potential of Immunogenic Cell Death in Cancer Chemotherapy and Radiotherapy. Int. J. Mol. Sci. 2019, 20, 959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Fan, Y.; Wang, M.; Li, Z.; Jiang, H.; Shi, J.; Shi, X.; Liu, S.; Zhao, J.; Kong, L.; Zhang, W.; et al. Intake of Soy, Soy Isoflavones and Soy Protein and Risk of Cancer Incidence and Mortality. Front. Nutr. 2022, 9, 847421. [Google Scholar] [CrossRef] [PubMed]
  94. Tuli, H.S.; Tuorkey, M.J.; Thakral, F.; Sak, K.; Kumar, M.; Sharma, A.K.; Sharma, U.; Jain, A.; Aggarwal, V.; Bishayee, A. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Front. Pharmacol. 2019, 10, 1336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yu, L.; Rios, E.; Castro, L.; Liu, J.; Yan, Y.; Dixon, D. Genistein: Dual Role in Women’s Health. Nutrients 2021, 13, 3048. [Google Scholar] [CrossRef]
  96. Kuiper, G.G.; Lemmen, J.G.; Carlsson, B.; Corton, J.C.; Safe, S.H.; van der Saag, P.T.; van der Burg, B.; Gustafsson, J.A. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 1998, 139, 4252–4263. [Google Scholar] [CrossRef] [PubMed]
  97. Choi, E.J.; Jung, J.Y.; Kim, G.H. Genistein inhibits the proliferation and differentiation of MCF-7 and 3T3-L1 cells via the regulation of ER alpha expression and induction of apoptosis. Exp. Ther. Med. 2014, 8, 454–458. [Google Scholar] [CrossRef] [Green Version]
  98. Ullah, M.F.; Ahmad, A.; Zubair, H.; Khan, H.Y.; Wang, Z.W.; Sarkar, F.H.; Hadi, S.M. Soy isoflavone genistein induces cell death in breast cancer cells through mobilization of endogenous copper ions and generation of reactive oxygen species. Mol. Nutr. Food Res. 2011, 55, 553–559. [Google Scholar] [CrossRef]
  99. Chan, L.J.; Pang, Y.H.; Wang, Y.J.; Zhu, D.; Taledaohan, A.; Jia, Y.J.; Zhao, L.C.; Wang, W.J. Genistein-induced mitochondrial dysfunction and FOXO3a/PUMA expression in non-small lung cancer cells. Pharm. Biol. 2022, 60, 1876–1883. [Google Scholar] [CrossRef]
  100. Hsiao, Y.C.; Peng, S.F.; Lai, K.C.; Liao, C.L.; Huang, Y.P.; Lin, C.C.; Lin, M.L.; Liu, K.C.; Tsai, C.C.; Ma, Y.S.; et al. Genistein induces apoptosis in vitro and has antitumor activity against human leukemia HL-60 cancer cell xenograft growth in vivo. Environ. Toxicol. 2019, 34, 443–456. [Google Scholar] [CrossRef]
  101. Park, C.; Cha, H.J.; Lee, H.; Hwang-Bo, H.; Ji, S.Y.; Kim, M.Y.; Hong, S.H.; Jeong, J.W.; Han, M.H.; Choi, S.H.; et al. Induction of G2/M Cell Cycle Arrest and Apoptosis by Genistein in Human Bladder Cancer T24 Cells through Inhibition of the ROS-Dependent PI3k/Akt Signal Transduction Pathway. Antioxidants 2019, 8, 327. [Google Scholar] [CrossRef]
  102. Zhang, Q.; Bao, J.; Yang, J. Genistein-triggered anticancer activity against liver cancer cell line HepG2 involves ROS generation, mitochondrial apoptosis, G2/M cell cycle arrest and inhibition of cell migration. Arch. Med. Sci. 2019, 15, 1001–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Shafiee, G.; Saidijam, M.; Tavilani, H.; Ghasemkhani, N.; Khodadadi, I. Genistein Induces Apoptosis and Inhibits Proliferation of HT29 Colon Cancer Cells. Int. J. Mol. Cell. Med. 2016, 5, 178–191. [Google Scholar] [PubMed]
  104. Qin, J.; Teng, J.A.; Zhu, Z.; Chen, J.X.; Huang, W.J. Genistein induces activation of the mitochondrial apoptosis pathway by inhibiting phosphorylation of Akt in colorectal cancer cells. Pharm. Biol. 2016, 54, 74–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Solomon, L.A.; Ali, S.; Banerjee, S.; Munkarah, A.R.; Morris, R.T.; Sarkar, F.H. Sensitization of ovarian cancer cells to cisplatin by genistein: The role of NF-kappaB. J. Ovarian Res. 2008, 1, 9. [Google Scholar] [CrossRef] [Green Version]
  106. Szliszka, E.; Krol, W. Soy isoflavones augment the effect of TRAIL-mediated apoptotic death in prostate cancer cells. Oncol. Rep. 2011, 26, 533–541. [Google Scholar] [CrossRef] [PubMed]
  107. Dong, X.; Xu, W.Q.; Sikes, R.A.; Wu, C.Q. Combination of low dose of genistein and daidzein has synergistic preventive effects on isogenic human prostate cancer cells when compared with individual soy isoflavone. Food Chem. 2013, 141, 1923–1933. [Google Scholar] [CrossRef] [PubMed]
  108. Asgharian, P.; Tazekand, A.P.; Hosseini, K.; Forouhandeh, H.; Ghasemnejad, T.; Ranjbar, M.; Hasan, M.; Kumar, M.; Beirami, S.M.; Tarhriz, V.; et al. Potential mechanisms of quercetin in cancer prevention: Focus on cellular and molecular targets. Cancer Cell Int. 2022, 22, 257. [Google Scholar] [CrossRef]
  109. Akbari Kordkheyli, V.; Khonakdar Tarsi, A.; Mishan, M.A.; Tafazoli, A.; Bardania, H.; Zarpou, S.; Bagheri, A. Effects of quercetin on microRNAs: A mechanistic review. J. Cell. Biochem. 2019, 120, 12141–12155. [Google Scholar] [CrossRef] [PubMed]
  110. Duo, J.; Ying, G.G.; Wang, G.W.; Zhang, L. Quercetin inhibits human breast cancer cell proliferation and induces apoptosis via Bcl-2 and Bax regulation. Mol. Med. Rep. 2012, 5, 1453–1456. [Google Scholar] [CrossRef]
  111. Chou, C.C.; Yang, J.S.; Lu, H.F.; Ip, S.W.; Lo, C.; Wu, C.C.; Lin, J.P.; Tang, N.Y.; Chung, J.G.; Chou, M.J.; et al. Quercetin-mediated Cell Cycle Arrest and Apoptosis Involving Activation of a Caspase Cascade through the Mitochondria! Pathway in Human Breast Cancer MCF-7 Cells. Arch. Pharm. Res. 2010, 33, 1181–1191. [Google Scholar] [CrossRef]
  112. Niu, G.M.; Yin, S.M.; Xie, S.F.; Li, Y.Q.; Nie, D.N.; Ma, L.P.; Wang, X.J.; Wu, Y.D. Quercetin induces apoptosis by activating caspase-3 and regulating Bcl-2 and cyclooxygenase-2 pathways in human HL-60 cells. Acta Biochim. Biophys. Sin. 2011, 43, 30–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Farrag, I.M.; Belal, A.; Badawi, M.H.A.; Abdelhady, A.A.; Galala, F.; El-Sharkawy, A.; El-Dahshan, A.A.; Mehany, A.B.M. Antiproliferative, Apoptotic Effects and Suppression of Oxidative Stress of Quercetin against Induced Toxicity in Lung Cancer Cells of Rats: In vitro and In vivo Study. J. Cancer 2021, 12, 5249–5259. [Google Scholar] [CrossRef]
  114. Zhou, J.; Gong, J.; Ding, C.; Chen, G. Quercetin induces the apoptosis of human ovarian carcinoma cells by upregulating the expression of microRNA-145. Mol. Med. Rep. 2015, 12, 3127–3131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gong, C.; Yang, Z.Y.; Zhang, L.Y.; Wang, Y.H.; Gong, W.; Liu, Y. Quercetin suppresses DNA double-strand break repair and enhances the radiosensitivity of human ovarian cancer cells via p53-dependent endoplasmic reticulum stress pathway. OncoTargets Ther. 2018, 11, 17–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Seo, H.S.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, M.; Kim, I.; Na, C.H.; Hur, H.; Jang, B.H.; et al. Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep. 2016, 36, 31–42. [Google Scholar] [CrossRef] [Green Version]
  117. Kedhari Sundaram, M.; Raina, R.; Afroze, N.; Bajbouj, K.; Hamad, M.; Haque, S.; Hussain, A. Quercetin modulates signaling pathways and induces apoptosis in cervical cancer cells. Biosci. Rep. 2019, 39, BSR20190720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Imran, M.; Aslam Gondal, T.; Atif, M.; Shahbaz, M.; Batool Qaisarani, T.; Hanif Mughal, M.; Salehi, B.; Martorell, M.; Sharifi-Rad, J. Apigenin as an anticancer agent. Phytother. Res. 2020, 34, 1812–1828. [Google Scholar] [CrossRef]
  119. Rahmani, A.H.; Alsahli, M.A.; Almatroudi, A.; Almogbel, M.A.; Khan, A.A.; Anwar, S.; Almatroodi, S.A. The Potential Role of Apigenin in Cancer Prevention and Treatment. Molecules 2022, 27, 6051. [Google Scholar] [CrossRef] [PubMed]
  120. Shukla, S.; Fu, P.F.; Gupta, S. Apigenin induces apoptosis by targeting inhibitor of apoptosis proteins and Ku70-Bax interaction in prostate cancer. Apoptosis 2014, 19, 883–894. [Google Scholar] [CrossRef]
  121. Shi, M.D.; Shiao, C.K.; Lee, Y.C.; Shih, Y.W. Apigenin, a dietary flavonoid, inhibits proliferation of human bladder cancer T-24 cells via blocking cell cycle progression and inducing apoptosis. Cancer Cell Int. 2015, 15, 33. [Google Scholar] [CrossRef]
  122. Woo, J.S.; Choo, G.S.; Yoo, E.S.; Kim, S.H.; Lee, J.H.; Han, S.H.; Kim, H.J.; Jung, S.H.; Park, Y.S.; Kim, B.S.; et al. Apigenin induces apoptosis by regulating Akt and MAPK pathways in human melanoma cell A375SM. Mol. Med. Rep. 2020, 22, 4877–4889. [Google Scholar] [CrossRef]
  123. Chan, L.P.; Chou, T.H.; Ding, H.Y.; Chen, P.R.; Chiang, F.Y.; Kuo, P.L.; Liang, C.H. Apigenin induces apoptosis via tumor necrosis factor receptor- and Bcl-2-mediated pathway and enhances susceptibility of head and neck squamous cell carcinoma to 5-fluorouracil and cisplatin. Biochim. Biophys. Acta-Gen. Subj. 2012, 1820, 1081–1091. [Google Scholar] [CrossRef] [PubMed]
  124. Masuelli, L.; Benvenuto, M.; Mattera, R.; Di Stefano, E.; Zago, E.; Taffera, G.; Tresoldi, I.; Giganti, M.G.; Frajese, G.V.; Berardi, G.; et al. In Vitro and In Vivo Anti-tumoral Effects of the Flavonoid Apigenin in Malignant Mesothelioma. Front. Pharmacol. 2017, 8, 373. [Google Scholar] [CrossRef] [Green Version]
  125. Sohel, M.; Sultana, H.; Sultana, T.; Al Amin, M.; Aktar, S.; Ali, M.C.; Rahim, Z.B.; Hossain, M.A.; Al Mamun, A.; Amin, M.N.; et al. Chemotherapeutic potential of hesperetin for cancer treatment, with mechanistic insights: A comprehensive review. Heliyon 2022, 8, e08815. [Google Scholar] [CrossRef]
  126. Zhang, J.X.; Wu, D.D.; Song, J.; Wang, J.; Yi, J.S.; Dong, W.G. Hesperetin Induces the Apoptosis of Gastric Cancer Cells via Activating Mitochondrial Pathway by Increasing Reactive Oxygen Species. Dig. Dis. Sci. 2015, 60, 2985–2995. [Google Scholar] [CrossRef] [PubMed]
  127. Wu, D.D.; Zhang, J.X.; Wang, J.; Li, J.; Liao, F.; Dong, W.G. Hesperetin induces apoptosis of esophageal cancer cells via mitochondrial pathway mediated by the increased intracellular reactive oxygen species. Tumor Biol. 2016, 37, 3451–3459. [Google Scholar] [CrossRef] [PubMed]
  128. Sivagami, G.; Vinothkumar, R.; Preethy, C.P.; Riyasdeen, A.; Akbarsha, M.A.; Menon, V.P.; Nalini, N. Role of hesperetin (a natural flavonoid) and its analogue on apoptosis in HT-29 human colon adenocarcinoma cell line—A comparative study. Food Chem. Toxicol. 2012, 50, 660–671. [Google Scholar] [CrossRef] [PubMed]
  129. Palit, S.; Kar, S.; Sharma, G.; Das, P.K. Hesperetin Induces Apoptosis in Breast Carcinoma by Triggering Accumulation of ROS and Activation of ASK1/JNK Pathway. J. Cell. Physiol. 2015, 230, 1729–1739. [Google Scholar] [CrossRef]
  130. Elango, R.; Athinarayanan, J.; Subbarayan, V.P.; Lei, D.K.Y.; Alshatwi, A.A. Hesperetin induces an apoptosis-triggered extrinsic pathway and a p53-independent pathway in human lung cancer H522 cells. J. Asian Nat. Prod. Res. 2018, 20, 559–569. [Google Scholar] [CrossRef]
  131. Tanaka, M.; Endo, H.; Sakusa, K.; Yano, M. Hesperetin induces apoptosis in A549 cells via the Hsp70-mediated activation of Bax. Int. J. Oncol. 2022, 61, 143. [Google Scholar] [CrossRef]
  132. Alshatwi, A.A.; Ramesh, E.; Periasamy, V.S.; Subash-Babu, P. The apoptotic effect of hesperetin on human cervical cancer cells is mediated through cell cycle arrest, death receptor, and mitochondrial pathways. Fundam. Clin. Pharmacol. 2013, 27, 581–592. [Google Scholar] [CrossRef] [PubMed]
  133. Prakash, M.; Basavaraj, B.V.; Murthy, K.N.C. Biological functions of epicatechin: Plant cell to human cell health. J. Funct. Foods 2019, 52, 14–24. [Google Scholar] [CrossRef]
  134. Pereyra-Vergara, F.; Olivares-Corichi, I.M.; Perez-Ruiz, A.G.; Luna-Arias, J.P.; Garcia-Sanchez, J.R. Apoptosis Induced by (-)-Epicatechin in Human Breast Cancer Cells is Mediated by Reactive Oxygen Species. Molecules 2020, 25, 1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kim, D.; Mollah, M.L.; Kim, K. Induction of Apoptosis of SW480 Human Colon Cancer Cells by (-)-Epicatechin Isolated from Bulnesia sarmienti. Anticancer Res. 2012, 32, 5353–5361. [Google Scholar]
  136. Thomas, P.; Dong, J. (-)-Epicatechin acts as a potent agonist of the membrane androgen receptor, ZIP9 (SLC39A9), to promote apoptosis of breast and prostate cancer cells. J. Steroid Biochem. 2021, 211, 105906. [Google Scholar] [CrossRef]
  137. Chen, J.L.; Xu, B.J.; Sun, J.X.; Jiang, X.W.; Bai, W.B. Anthocyanin supplement as a dietary strategy in cancer prevention and management: A comprehensive review. Crit. Rev. Food Sci. Nutr. 2022, 62, 7242–7254. [Google Scholar] [CrossRef]
  138. Sorrenti, V.; Vanella, L.; Acquaviva, R.; Cardile, V.; Giofre, S.; Di Giacomo, C. Cyanidin induces apoptosis and differentiation in prostate cancer cells. Int. J. Oncol. 2015, 47, 1303–1310. [Google Scholar] [CrossRef] [Green Version]
  139. Hosseini, M.M.; Karimi, A.; Behroozaghdam, M.; Javidi, M.A.; Ghiasvand, S.; Bereimipour, A.; Aryan, H.; Nassiri, F.; Jangholi, E. Cytotoxic and Apoptogenic Effects of Cyanidin-3-Glucoside on the Glioblastoma Cell Line. World Neurosurg. 2017, 108, 94–100. [Google Scholar] [CrossRef]
  140. Atashi, H.A.; Arani, H.Z.; Shekarriz, A.; Nazari, H.; Zabolian, A.; Rakhshan, R.; Olya, M. Cyanidin 3-O-Glucoside Induces the Apoptosis in the Osteosarcoma Cells through Upregulation of the PPAR gamma and P21: An In Vitro Study. Anticancer Agents Med. Chem. 2020, 20, 1087–1093. [Google Scholar] [CrossRef]
  141. Abotaleb, M.; Liskova, A.; Kubatka, P.; Busselberg, D. Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules 2020, 10, 221. [Google Scholar] [CrossRef] [Green Version]
  142. ElKhazendar, M.; Chalak, J.; El-Huneidi, W.; Vinod, A.; Abdel-Rahman, W.M.; Abu-Gharbieh, E. Antiproliferative and proapoptotic activities of ferulic acid in breast and liver cancer cell lines. Trop. J. Pharm. Res. 2019, 18, 2571–2576. [Google Scholar] [CrossRef]
  143. Cheng, W.J.; Zhang, P.P.; Luo, Q.Q.; Deng, S.M.; Jia, A.Q. The chemosensitizer ferulic acid enhances epirubicin-induced apoptosis in MDA-MB-231 cells. J. Funct. Foods 2020, 73, 104130. [Google Scholar] [CrossRef]
  144. Luo, L.P.; Zhu, S.H.; Tong, Y.; Peng, S.W. Ferulic Acid Induces Apoptosis of HeLa and Caski Cervical Carcinoma Cells by Down-Regulating the Phosphatidylinositol 3-Kinase (PI3K)/Akt Signaling Pathway. Med. Sci. Monit. 2020, 26, e920095. [Google Scholar] [CrossRef] [PubMed]
  145. Tsai, C.L.; Chiu, Y.M.; Ho, T.Y.; Hsieh, C.T.; Shieh, D.C.; Lee, Y.J.; Tsay, G.J.; Wu, Y.Y. Gallic Acid Induces Apoptosis in Human Gastric Adenocarcinoma Cells. Anticancer Res. 2018, 38, 2057–2067. [Google Scholar] [CrossRef] [PubMed]
  146. Yahya, S.A.; Hassan, F.A. Cytotoxic Effects Of Gallic Acid In Human Breast Cancer Cell Mda-Mb-231. J. Pharm. Negat. Result 2022, 13, 1337–1344. [Google Scholar] [CrossRef]
  147. Aborehab, N.M.; Elnagar, M.R.; Waly, N.E. Gallic acid potentiates the apoptotic effect of paclitaxel and carboplatin via overexpression of Bax and P53 on the MCF-7 human breast cancer cell line. J. Biochem. Mol. Toxicol. 2021, 35, e22638. [Google Scholar] [CrossRef] [PubMed]
  148. Mottaghi, S.; Abbaszadeh, H. A comprehensive mechanistic insight into the dietary and estrogenic lignans, arctigenin and sesamin as potential anticarcinogenic and anticancer agents. Current status, challenges, and future perspectives. Crit. Rev. Food Sci. Nutr. 2022, 62, 7301–7318. [Google Scholar] [CrossRef]
  149. Ozgocmen, M.; Bayram, D.; Turel, G.Y.; Togay, V.A.; Calapoglu, N.S. Secoisolariciresinol diglucoside induces caspase-3-mediated apoptosis in monolayer and spheroid cultures of human colon carcinoma cells. J. Food Biochem. 2021, 45, e13719. [Google Scholar] [CrossRef]
  150. Chen, J.; Saggar, J.K.; Corey, P.; Thompson, L.U. Flaxseed and Pure Secoisolariciresinol Diglucoside, but Not Flaxseed Hull, Reduce Human Breast Tumor Growth (MCF-7) in Athymic Mice. J. Nutr. 2009, 139, 2061–2066. [Google Scholar] [CrossRef] [Green Version]
  151. Sirerol, J.A.; Rodriguez, M.L.; Mena, S.; Asensi, M.A.; Estrela, J.M.; Ortega, A.L. Role of Natural Stilbenes in the Prevention of Cancer. Oxid. Med. Cell. Longev. 2016, 2016, 3128951. [Google Scholar] [CrossRef] [Green Version]
  152. Liu, Y.W.; Wang, L.J.; Wu, Y.P.; Lv, C.W.; Li, X.K.; Cao, X.R.; Yang, M.; Feng, D.P.; Luo, Z.J. Pterostilbene exerts antitumor activity against human osteosarcoma cells by inhibiting the JAK2/STAT3 signaling pathway. Toxicology 2013, 304, 120–131. [Google Scholar] [CrossRef]
  153. Alosi, J.A.; McDonald, D.E.; Schneider, J.S.; Privette, A.R.; McFadden, D.W. Pterostilbene Inhibits Breast Cancer In Vitro Through Mitochondrial Depolarization and Induction of Caspase-Dependent Apoptosis. J. Surg. Res. 2010, 161, 195–201. [Google Scholar] [CrossRef] [PubMed]
  154. Elsherbini, A.M.; Sheweita, S.A.; Sultan, A.S. Pterostilbene as a Phytochemical Compound Induces Signaling Pathways Involved in the Apoptosis and Death of Mutant P53-Breast Cancer Cell Lines. Nutr. Cancer 2021, 73, 1976–1984. [Google Scholar] [CrossRef] [PubMed]
  155. Wakimoto, R.; Ono, M.; Takeshima, M.; Higuchi, T.; Nakano, S. Differential Anticancer Activity of Pterostilbene Against Three Subtypes of Human Breast Cancer Cells. Anticancer Res. 2017, 37, 6153–6159. [Google Scholar] [CrossRef] [PubMed]
  156. Wen, W.; Lowe, G.; Roberts, C.M.; Finlay, J.; Han, E.S.; Glackin, C.A.; Dellinger, T.H. Pterostilbene, a natural phenolic compound, synergizes the antineoplastic effects of megestrol acetate in endometrial cancer. Sci. Rep. 2017, 7, 12754. [Google Scholar] [CrossRef] [Green Version]
  157. Shin, H.J.; Han, J.M.; Choi, Y.S.; Jung, H.J. Pterostilbene Suppresses both Cancer Cells and Cancer Stem-Like Cells in Cervical Cancer with Superior Bioavailability to Resveratrol. Molecules 2020, 25, 228. [Google Scholar] [CrossRef] [Green Version]
  158. Guo, L.Y.; Tan, K.; Wang, H.; Zhang, X. Pterostilbene inhibits hepatocellular carcinoma through p53/SOD2/ROS-mediated mitochondrial apoptosis. Oncol. Rep. 2016, 36, 3233–3240. [Google Scholar] [CrossRef] [Green Version]
  159. Degot, P.; Huber, V.; Touraud, D.; Kunz, W. Curcumin extracts from Curcuma Longa—Improvement of concentration, purity, and stability in food-approved and water-soluble surfactant-free microemulsions. Food Chem. 2021, 339, 128140. [Google Scholar] [CrossRef]
  160. Yang, Z.J.; Huang, S.Y.; Zhou, D.D.; Xiong, R.G.; Zhao, C.N.; Fang, A.P.; Zhang, Y.J.; Li, H.B.; Zhu, H.L. Effects and Mechanisms of Curcumin for the Prevention and Management of Cancers: An Updated Review. Antioxidants 2022, 11, 1481. [Google Scholar] [CrossRef]
  161. Shakor, A.B.A.; Atia, M.; Ismail, I.A.; Alshehri, A.; El-Refaey, H.; Kwiatkowska, K.; Sobota, A. Curcumin induces apoptosis of multidrug-resistant human leukemia HL60 cells by complex pathways leading to ceramide accumulation. Bba-Mol. Cell Biol. L. 2014, 1841, 1672–1682. [Google Scholar] [CrossRef]
  162. Yu, T.; Ji, J.; Guo, Y.L. MST1 activation by curcumin mediates JNK activation, Foxo3a nuclear translocation and apoptosis in melanoma cells. Biochem. Bioph. Res. Commun. 2013, 441, 53–58. [Google Scholar] [CrossRef] [PubMed]
  163. Park, S.; Cho, D.H.; Andera, L.; Suh, N.; Kim, I. Curcumin enhances TRAIL-induced apoptosis of breast cancer cells by regulating apoptosis-related proteins. Mol. Cell. Biochem. 2013, 383, 39–48. [Google Scholar] [CrossRef] [PubMed]
  164. Jung, E.M.; Park, J.W.; Choi, K.S.; Park, J.W.; Lee, H.I.L.; Lee, K.S.; Kwon, T.K. Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis through CHOP-independent DR5 upregulation. Carcinogenesis 2006, 27, 2008–2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kwon, D.; Oh, S.; Park, J.H.; Lee, S.; Lee, S. The TRAIL Sensitization Effect of Substituted Triazolyl Curcumin Mimics Against Brain Cancer Cells. Bull. Korean Chem. Soc. 2014, 35, 3339–3342. [Google Scholar] [CrossRef] [Green Version]
  166. Shankar, S.; Ganapathy, S.; Chen, Q.H.; Srivastava, R.K. Curcumin sensitizes TRAIL-resistant xenografts: Molecular mechanisms of apoptosis, metastasis and angiogenesis. Mol. Cancer 2008, 7, 16. [Google Scholar] [CrossRef] [Green Version]
  167. Wang, W.Z.; Li, L.; Liu, M.Y.; Jin, X.B.; Mao, J.W.; Pu, Q.H.; Meng, M.J.; Chen, X.G.; Zhu, J.Y. Curcumin induces FasL-related apoptosis through p38 activation in human hepatocellular carcinoma Huh7 cells. Life Sci. 2013, 92, 352–358. [Google Scholar] [CrossRef]
  168. Obaidi, I.; Blanco Fernandez, A.; McMorrow, T. Curcumin Sensitises Cancerous Kidney Cells to TRAIL Induced Apoptosis via Let-7C Mediated Deregulation of Cell Cycle Proteins and Cellular Metabolism. Int. J. Mol. Sci. 2022, 23, 9569. [Google Scholar] [CrossRef] [PubMed]
  169. Choudhuri, T.; Pal, S.; Agwarwal, M.L.; Das, T.; Sa, G. Curcumin induces apoptosis in human breast cancer cells through p53-dependent Bax induction. Febs Lett. 2002, 512, 334–340. [Google Scholar] [CrossRef] [Green Version]
  170. Mou, S.; Zhou, Z.; He, Y.; Liu, F.; Gong, L. Curcumin inhibits cell proliferation and promotes apoptosis of laryngeal cancer cells through Bcl-2 and PI3K/Akt, and by upregulating miR-15a. Oncol. Lett. 2017, 14, 4937–4942. [Google Scholar] [CrossRef] [Green Version]
  171. Zhou, Q.M.; Sun, Y.; Lu, Y.Y.; Zhang, H.; Chen, Q.L.; Su, S.B. Curcumin reduces mitomycin C resistance in breast cancer stem cells by regulating Bcl-2 family-mediated apoptosis. Cancer Cell Int. 2017, 17, 84. [Google Scholar] [CrossRef] [Green Version]
  172. Sha, J.; Li, J.; Wang, W.; Pan, L.; Cheng, J.; Li, L.; Zhao, H.; Lin, W. Curcumin induces G0/G1 arrest and apoptosis in hormone independent prostate cancer DU-145 cells by down regulating Notch signaling. Biomed. Pharmacother. 2016, 84, 177–184. [Google Scholar] [CrossRef] [PubMed]
  173. Bush, J.A.; Cheung, K.J., Jr.; Li, G. Curcumin induces apoptosis in human melanoma cells through a Fas receptor/caspase-8 pathway independent of p53. Exp. Cell. Res. 2001, 271, 305–314. [Google Scholar] [CrossRef] [PubMed]
  174. Aravindan, N.; Madhusoodhanan, R.; Ahmad, S.; Johnson, D.; Herman, T.S. Curcumin inhibits NFkappaB mediated radioprotection and modulate apoptosis related genes in human neuroblastoma cells. Cancer Biol. Ther. 2008, 7, 569–576. [Google Scholar] [CrossRef] [Green Version]
  175. Qiao, Q.; Jiang, Y.; Li, G. Inhibition of the PI3K/AKT-NF-kappaB pathway with curcumin enhanced radiation-induced apoptosis in human Burkitt’s lymphoma. J. Pharmacol. Sci. 2013, 121, 247–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Wu, S.H.; Hang, L.W.; Yang, J.S.; Chen, H.Y.; Lin, H.Y.; Chiang, J.H.; Lu, C.C.; Yang, J.L.; Lai, T.Y.; Ko, Y.C.; et al. Curcumin induces apoptosis in human non-small cell lung cancer NCI-H460 cells through ER stress and caspase cascade- and mitochondria-dependent pathways. Anticancer Res. 2010, 30, 2125–2133. [Google Scholar]
  177. Sohn, E.J.; Bak, K.M.; Nam, Y.K.; Park, H.T. Upregulation of microRNA 344a-3p is involved in curcumin induced apoptosis in RT4 schwannoma cells. Cancer Cell Int. 2018, 18, 199. [Google Scholar] [CrossRef] [Green Version]
  178. Khan, A.Q.; Ahmed, E.I.; Elareer, N.; Fathima, H.; Prabhu, K.S.; Siveen, K.S.; Kulinski, M.; Azizi, F.; Dermime, S.; Ahmad, A.; et al. Curcumin-Mediated Apoptotic Cell Death in Papillary Thyroid Cancer and Cancer Stem-Like Cells through Targeting of the JAK/STAT3 Signaling Pathway. Int. J. Mol. Sci. 2020, 21, 438. [Google Scholar] [CrossRef] [Green Version]
  179. Vladu, A.F.; Ficai, D.; Ene, A.G.; Ficai, A. Combination Therapy Using Polyphenols: An Efficient Way to Improve Antitumoral Activity and Reduce Resistance. Int. J. Mol. Sci. 2022, 23, 10244. [Google Scholar] [CrossRef]
  180. Patra, S.; Pradhan, B.; Nayak, R.; Behera, C.; Das, S.; Patra, S.K.; Efferth, T.; Jena, M.; Bhutia, S.K. Dietary polyphenols in chemoprevention and synergistic effect in cancer: Clinical evidences and molecular mechanisms of action. Phytomedicine 2021, 90, 153554. [Google Scholar] [CrossRef] [PubMed]
  181. Liu, H.; Lee, G.; Lee, J.I.; Ahn, T.G.; Kim, S.A. Effects of genistein on anti-tumor activity of cisplatin in human cervical cancer cell lines. Obstet. Gynecol. Sci. 2019, 62, 322–328. [Google Scholar] [CrossRef]
  182. Kaushik, S.; Shyam, H.; Agarwal, S.; Sharma, R.; Nag, T.C.; Dwivedi, A.K.; Balapure, A.K. Genistein potentiates Centchroman induced antineoplasticity in breast cancer via PI3K/Akt deactivation and ROS dependent induction of apoptosis. Life Sci. 2019, 239, 117073. [Google Scholar] [CrossRef] [PubMed]
  183. Xu, W.; Xie, S.; Chen, X.; Pan, S.; Qian, H.; Zhu, X. Effects of Quercetin on the Efficacy of Various Chemotherapeutic Drugs in Cervical Cancer Cells. Drug Des. Devel. Ther. 2021, 15, 577–588. [Google Scholar] [CrossRef] [PubMed]
  184. Mohammadi, E.; Alemi, F.; Maleki, M.; Malakoti, F.; Farsad-Akhtar, N.; Yousefi, B. Quercetin and Methotrexate in Combination have Anticancer Activity in Osteosarcoma Cells and Repress Oncogenic MicroRNA-223. Drug Res. 2022, 72, 226–233. [Google Scholar] [CrossRef] [PubMed]
  185. Xu, Z.X.; Zhao, D.M.; Zheng, X.Y.; Huang, B.; Xia, X.S.; Pan, X.J. Quercetin exerts bidirectional regulation effects on the efficacy of tamoxifen in estrogen receptor-positive breast cancer therapy: An in vitro study. Environ. Toxicol. 2020, 35, 1179–1193. [Google Scholar] [CrossRef] [PubMed]
  186. Hu, W.J.; Liu, J.; Zhong, L.K.; Wang, J. Apigenin enhances the antitumor effects of cetuximab in nasopharyngeal carcinoma by inhibiting EGFR signaling. Biomed. Pharmacother. 2018, 102, 681–688. [Google Scholar] [CrossRef]
  187. Liu, R.; Ji, P.; Liu, B.; Qiao, H.S.; Wang, X.; Zhou, L.K.; Deng, T.; Ba, Y. Apigenin enhances the cisplatin cytotoxic effect through p53-modulated apoptosis. Oncol. Lett. 2017, 13, 1024–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Xu, Y.M.; Xin, Y.Q.; Diao, Y.; Lu, C.Y.; Fu, J.; Luo, L.; Yin, Z.M. Synergistic Effects of Apigenin and Paclitaxel on Apoptosis of Cancer Cells. PLoS ONE 2011, 6, e29169. [Google Scholar] [CrossRef] [Green Version]
  189. Mahbub, A.A.; Le Maitre, C.L.; Cross, N.A.; Jordan-Mahy, N. The effect of apigenin and chemotherapy combination treatments on apoptosis-related genes and proteins in acute leukaemia cell lines. Sci. Rep. 2022, 12, 8858. [Google Scholar] [CrossRef]
  190. Mahbub, A.A.; Maitre, C.L.L.; Haywood-Small, S.; Cross, N.A.; Jordan-Mahy, N. Polyphenols enhance the activity of alkylating agents in leukaemia cell lines. Oncotarget 2019, 10, 4570–4586. [Google Scholar] [CrossRef] [Green Version]
  191. He, P.; Ma, J.; Liu, Y.; Deng, H.; Dong, W. Hesperetin Promotes Cisplatin-Induced Apoptosis of Gastric Cancer In Vitro and In Vivo by Upregulating PTEN Expression. Front. Pharmacol. 2020, 11, 1326. [Google Scholar] [CrossRef]
  192. Li, X.; Mu, J.; Lin, Y.; Zhao, J.; Meng, X. Combination of cyanidin-3-O-glucoside and cisplatin induces oxidative stress and apoptosis in HeLa cells by reducing activity of endogenous antioxidants, increasing bax/bcl-2 mRNA expression ratio, and downregulating Nrf2 expression. J. Food Biochem. 2021, 45, e13806. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, T.; Ma, L.; Wu, P.; Li, W.; Li, T.; Gu, R.; Dan, X.; Li, Z.; Fan, X.; Xiao, Z. Gallic acid has anticancer activity and enhances the anticancer effects of cisplatin in non--small cell lung cancer A549 cells via the JAK/STAT3 signaling pathway. Oncol. Rep. 2019, 41, 1779–1788. [Google Scholar] [CrossRef] [PubMed]
  194. Hsu, Y.H.; Chen, S.Y.; Wang, S.Y.; Lin, J.A.; Yen, G.C. Pterostilbene Enhances Cytotoxicity and Chemosensitivity in Human Pancreatic Cancer Cells. Biomolecules 2020, 10, 709. [Google Scholar] [CrossRef]
  195. Kostin, S.F.; McDonald, D.E.; McFadden, D.W. Inhibitory effects of (-)-epigallocatechin-3-gallate and pterostilbene on pancreatic cancer growth in vitro. J. Surg. Res. 2012, 177, 255–262. [Google Scholar] [CrossRef]
  196. Sivanantham, B.; Sethuraman, S.; Krishnan, U.M. Combinatorial Effects of Curcumin with an Anti-Neoplastic Agent on Head and Neck Squamous Cell Carcinoma Through the Regulation of EGFR-ERK1/2 and Apoptotic Signaling Pathways. Acs Comb. Sci. 2016, 18, 22–35. [Google Scholar] [CrossRef] [PubMed]
  197. Zhan, Y.Z.; Chen, Y.N.; Liu, R.; Zhang, H.; Zhang, Y.M. Potentiation of paclitaxel activity by curcumin in human breast cancer cell by modulating apoptosis and inhibiting EGFR signaling. Arch. Pharm. Res. 2014, 37, 1086–1095. [Google Scholar] [CrossRef]
  198. Calaf, G.M.; Ponce-Cusi, R.; Carrion, F. Curcumin and paclitaxel induce cell death in breast cancer cell lines. Oncol. Rep. 2018, 40, 2381–2388. [Google Scholar] [CrossRef]
  199. Dang, Y.P.; Yuan, X.Y.; Tian, R.; Li, D.G.; Liu, W. Curcumin improves the paclitaxel-induced apoptosis of HPV-positive human cervical cancer cells via the NF-kappa B-p53-caspase-3 pathway. Exp. Ther. Med. 2015, 9, 1470–1476. [Google Scholar] [CrossRef] [Green Version]
  200. Altundag, E.M.; Yilmaz, A.M.; Serdar, B.S.; Jannuzzi, A.T.; Kocturk, S.; Yalcin, A.S. Synergistic Induction of Apoptosis by Quercetin and Curcumin in Chronic Myeloid Leukemia (K562) Cells: II. Signal Transduction Pathways Involved. Nutr. Cancer 2021, 73, 703–712. [Google Scholar] [CrossRef]
Figure 1. Schematic classification of dietary polyphenols and main relevant phytochemical compounds belonging to the various groups.
Figure 1. Schematic classification of dietary polyphenols and main relevant phytochemical compounds belonging to the various groups.
Ijms 24 01680 g001
Figure 2. Effects of some polyphenols on extrinsic and intrinsic apoptotic pathways in cancer. ↑: increase, ↓: decrease. Adapted from Chimento, A. et al. [89].
Figure 2. Effects of some polyphenols on extrinsic and intrinsic apoptotic pathways in cancer. ↑: increase, ↓: decrease. Adapted from Chimento, A. et al. [89].
Ijms 24 01680 g002
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

Chimento, A.; De Luca, A.; D’Amico, M.; De Amicis, F.; Pezzi, V. The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 1680. https://doi.org/10.3390/ijms24021680

AMA Style

Chimento A, De Luca A, D’Amico M, De Amicis F, Pezzi V. The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy. International Journal of Molecular Sciences. 2023; 24(2):1680. https://doi.org/10.3390/ijms24021680

Chicago/Turabian Style

Chimento, Adele, Arianna De Luca, Maria D’Amico, Francesca De Amicis, and Vincenzo Pezzi. 2023. "The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy" International Journal of Molecular Sciences 24, no. 2: 1680. https://doi.org/10.3390/ijms24021680

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