Next Article in Journal
Protaetia brevitarsis Hydrolysate Mitigates Muscle Dysfunction and Ectopic Fat Deposition Triggered by a High-Fat Diet in Mice
Previous Article in Journal
Postprandial Aminoacidemia Following the Ingestion of Alternative and Sustainable Proteins in Humans: A Narrative Review
Previous Article in Special Issue
The Impact of Minerals on Female Fertility: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Epigallocatechin Gallate in Selected Malignant Neoplasms in Women

1
Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
2
Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
3
Gynecological Oncology Center, Poznańska 58A, 60-850 Poznań, Poland
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(2), 212; https://doi.org/10.3390/nu17020212
Submission received: 19 December 2024 / Revised: 4 January 2025 / Accepted: 7 January 2025 / Published: 8 January 2025
(This article belongs to the Special Issue The Role of Nutrition in Gynecological Diseases)

Abstract

:
Tea is a significant source of flavonoids in the diet. Due to different production processes, the amount of bioactive compounds in unfermented (green) and (semi-)fermented tea differs. Importantly, green tea has a similar composition of phenolic compounds to fresh, unprocessed tea leaves. It consists primarily of monomeric flavan-3-ols, known as catechins, of which epigallocatechin gallate (EGCG) is the most abundant. Thanks to its antioxidant, antiproliferative, and antiangiogenic properties, EGCG has attracted the scientific community’s attention to its potential use in preventing and/or combating cancer. In this review article, we summarize the literature reports found in the Google Scholar and PubMed databases on the anticancer effect of EGCG on selected malignant neoplasms in women, i.e., breast, cervical, endometrial, and ovarian cancers, which have been published over the last two decades. It needs to be emphasized that EGCG concentrations reported as effective against cancer cells are typically higher than those found in plasma after polyphenol administration. Moreover, the low bioavailability and absorption of EGCG appear to be the main reasons for the differences in the effects between in vitro and in vivo studies. In this context, we also decided to look at possible solutions to these problems, consisting of combining the polyphenol with other bioactive components or using nanotechnology. Despite the promising results of the studies conducted so far, mainly in vitro and on animal models, there is no doubt that further, broad-based activities are necessary to unequivocally assess the potential use of EGCG in oncological treatment to combat cancer in women.

1. Introduction

Cancer is a significant health issue in modern societies. Global statistics indicate that from 2020 to 2022, approximately 20 million people were diagnosed with cancer each year, and nearly 10 million cancer patients died annually from the disease [1,2]. The outlook for the coming decades is not encouraging. It is estimated that in 2040, over 28 million people will be diagnosed with cancer, representing a 47% increase compared to current statistics [1]. In 2050, this number is expected to rise to more than 35 million cases, indicating a 77% increase based on currently available data [2]. The increase in cancer-related deaths is projected to be even higher; Australian scientists estimate that by 2050, 8.8 million (90%) more people will die of cancer compared to 2022 [3].
There are several reasons for this upward trend, but one of the most significant is the anticipated demographic changes. In 2022, the global population was approximately 8 billion, and it is expected to exceed 9 billion by 2050 [2]. Cancer is a health and significant social problem due to its economic consequences. The loss of human potential, a notable reduction in productivity (presenteeism), long-term or complete exclusion from the labor market, the costs associated with treatment and care, and diminished social well-being are just a few of the adverse effects on the economy. These factors can ultimately lead to a deterioration of public finances, potentially threatening their stability.
Despite the substantial advancements in cancer treatment, the effectiveness of therapies for patients with cancer, particularly those with malignant tumors, remains inadequate. As a result, there is increasing interest in exploring potential strategies to enhance traditional cancer therapies, such as adjuvant treatments, that utilize specific natural products.
Phenolic compounds are significant components of tea leaves, comprising 10–30% of the dry weight [4,5,6], but some literature sources indicate that this level can reach up to 36% [7,8]. The predominant compounds are catechins (flavan-3-ols), which account for up to approximately 30% of tea’s dry weight [9]. Variations in the tea chemical composition arise from such factors as plant cultivation conditions, geographical location, agricultural methods, and specific plant characteristics such as variety, age, and leaf position on the shoot [10]. Green tea, derived from the Camellia sinensis tea bush, is the second most popular beverage in the world after water, particularly in Asian countries. This unfermented tea contains various pharmacologically active compounds that have been successfully isolated and identified. These compounds offer a wide range of beneficial properties, including antioxidant, anti-inflammatory, hypoglycemic, antiviral, neuroprotective, cardioprotective, and anticancer effects [10,11,12,13,14,15,16]. In green tea production, heating the withered leaves deactivates endogenous oxidases, preventing the oxidation of the catechins. Fresh tea leaves and green tea share a similar composition of phenolic compounds [17]. There are four main types of monomeric catechins. Two of these catechins, (–)-epicatechin and (–)-epigallocatechin, have a free hydroxyl group at position C3. The other two are C3-esters of gallic acid, which are (–)-epicatechin gallate and (–)-epigallocatechin gallate (EGCG) (Figure 1). EGCG is regarded as the most intriguing compound in this group due to its abundant presence in green tea infusions, which can influence various physiological and pathological processes in humans [18].
The structure of EGCG consists of three aromatic rings (A, B, and D) and a heterocyclic ring, C, occupying the central position of the catechin molecule (Figure 1). Ring A, being the resorcinol component, is fused to ring C, while rings B (3,4,5-trihydroxyphenyl) and D (gallic acid residue) are connected to ring C directly or via an ester group at position C3 (Figure 1). The substituents at the C2 and C3 positions exhibit a syn (2R,3R) configuration, resulting in a (–)-epi(gallo)catechin composition (Figure 1). This differs from the (–)-(gallo)catechin arrangement with the corresponding substituents in an anti (2S,3R) configuration. The characteristic structure of EGCG, especially the presence of numerous hydroxyl groups, determines not only the favorable biological properties (including the highest antioxidant activity among catechins [19]) but also the relatively low bioavailability and metabolic stability of this natural polyphenol.

1.1. Bioavailability and Metabolic Biodegradation Pathways of EGCG

Although tea is a major source of dietary phenolic compounds, only a small portion can enter the bloodstream in their original form after consumption. The natural phenols in green tea travel through the mouth, esophagus, and stomach mostly unchanged. Only a limited percentage is absorbed by enterocytes and subsequently enters the circulatory system [17]. The pharmacokinetic properties of green tea polyphenols, especially EGCG, have been relatively well established in rodent studies (mice, rats), but there are some uncertainties about humans [20]. The total bioavailability of EGCG after oral administration of decaffeinated green tea to rats was only 0.1% [21]. In mice, polyphenol availability was higher (several percent) but still far from satisfactory [22]. In turn, studies conducted with the participation of healthy volunteers showed that after administering 800 mg of EGCG, the highest plasma concentration was 0.6–3.3 µM [23,24,25]. However, taking ≥800 mg of EGCG daily for 4 months or longer may be associated with increased alanine and aspartate aminotransferase levels, which may be a sign of liver dysfunction [26]. Nakagawa et al. [27] conducted a study involving four healthy individuals aged 21 to 28 years. They found that after the ingestion of 95 mg of polyphenol, the area under the plasma EGCG concentration-time curve was 857 ng·h/mL [27].
Limited bioavailability in the small intestine means that most phenols reach the large intestine in an unchanged form [28], raising questions about how these compounds can positively affect the body’s functioning. Native phenols found in green tea may possess health-promoting properties, at least in part, due to their direct impact on the microbiota, exhibiting prebiotic activity [17]. Additionally, some of these phenols can be converted into metabolites that are more accessible to the host by the microorganisms present in the large intestine (Figure 2) [29,30]. Specifically, the ester bond hydrolysis in EGCG, leading to gallic acid motif release, along with the C ring fission, is achieved exclusively by microbial enzymes, not mammalian ones [29]. Out of 169 strains of intestinal bacteria screened, Enterobacter aerogenes, Raoultella planticola, Klebsiella pneumoniae subsp. pneumoniae, and Bifidobacterium longum subsp. infantis have been identified as effective in hydrolyzing EGCG to EGC and gallic acid components, while Eubacterium oxidoreducens, E. ramulus, E. casseli avus, Clostridium orbiscidens, and the other ones belonging to the Butyrivibrio genus are involved in the fission of the C ring [29]. Different bacterial strains can also work together to degrade compounds from the catechin group [29]. Studies conducted on rats have demonstrated that when using EGCG labeled with tritium at position C4, the levels of radioactivity in the blood and urine were lower in the group that received initial antibiotic therapy compared to the control group that did not receive antibiotics [31]. Moreover, polyphenol absorption remained low for 4 h after oral administration, began to increase after 8 h, and reached maximum concentration 24 h post-ingestion [31].
The enzymatic activity of the intestinal microbiota enables efficient degradation of the organic compounds contained in tea, playing a key role in increasing the bioavailability of polyphenols, including EGCG. The principal metabolic pathway of EGCG degradation is based on the formation of (R)-5-(3,5-dihydroxyphenyl)-4-hydroxypentanoic acid 2 as the main metabolite through a series of transformations, including (i) hydrolysis of the ester group, (ii) opening of the C ring, (iii) removal of the hydroxyl group at position C4′, and (iv) cleavage of the A ring (Figure 2) [32,33]. The ɣ-hydroxyl acid metabolite 2 can, in turn, be converted into lactone 3 and then conjugated with glucuronic acid to form the 3′-O-β-glucuronide derivative 4 (Figure 2) [31,32,34]. Glucuronide conjugate 4, which is classified as a phase II hepatic metabolite and reaches the colon via enterohepatic circulation, may undergo deconjugation and thus undergo further metabolism by the intestinal microbiota [35]. EGCG metabolites are generally less complex than the parent polyphenol and thus more readily absorbed in the large intestine [34].

1.2. Basic Epidemiological Data on Selected Malignant Neoplasms in Women

Breast cancer (BC) is the most common malignant tumor in women and the 2nd most common cause of cancer worldwide, regardless of gender [2]. In 2022, ~2.3 million women worldwide fell ill with this type of cancer, which constituted 11.6% of all recorded cancer cases, and ~666,000 patients died (6.9% of all cancer deaths) [2]. BC is a heterogeneous disease, both in terms of epidemiology (sporadic tumors, tumors with BRCA mutations) and the expression of receptors: estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER2). About 10–15% of all diagnosed BCs do not have these receptors; these are triple-negative BCs (TNBCs) with an aggressive course and poor prognosis [36]. These cancers are usually more common in premenopausal women (under 40 years of age) who are black and have a BRCA1 mutation [36].
Cervical cancer (CC) is one of the most common malignancies in women; it ranks 4th in terms of incidence among all female cancers [2]. In 2022, over 660,000 women were diagnosed with CC, representing 6.8% of all female malignancies [2]. Nearly 350,000 patients died from this disease, accounting for 8.1% of all deaths caused by female malignancies [2]. A detailed analysis of data on the mortality of oncological patients indicates that CC was the leading cause of cancer deaths in 37 out of 185 countries (20%) listed in the 2022 study [2]. Persistent infection with high-risk HPV viruses, which include 13 types, is closely associated with the development of dysplastic changes and CC; viral oncoproteins E6 and E7 are responsible for oncogenic activity [37]. The basic preventive strategy to protect against CC is vaccination against HPV, while in the case of cancer development, in addition to surgical treatment, cytotoxic chemical treatment is recommended.
Endometrial cancer (EC) is the 6th most commonly diagnosed malignant tumor in women in developed countries [2]. In 2022, more than 420,000 women were diagnosed with this type of cancer, which represented 4.3% of all malignant tumors in females, and nearly 98,000 patients died [2]. Unfortunately, the EC survival rate has decreased over the last four decades [38].
Ovarian cancer (OC) is the leading cause of death in women with cancer. In 2022, this type of cancer was diagnosed in over 320,000 women (3.4% of all cases of female cancer), and over 206,000 women died, which accounted for 4.8% of all cancer-related deaths among oncology patients [2]. Over 70% of this type of cancer is diagnosed in advanced stages; despite cytoreduction and chemotherapy, approximately 70–80% of patients experience relapses with an unfavorable course, and the cause of this phenomenon may be the presence of a small subpopulation of cancer stem cells (CSCs) inside cancer tumors [39,40,41]. Of note, phytochemicals can attack and destroy CSCs [42,43].

2. Anticancer Activity of EGCG

A growing body of research data suggests that EGCG may influence several fundamental biological processes and characteristics that contribute to the development and progression of cancer [44]. EGCG is a compound capable of attacking cancer cells [11,12,45] and CSCs [42]. The scientific literature describes in detail the mechanisms underlying the anticancer effects of EGCG, which include (i) inhibition of cancer cell proliferation and growth, (ii) inhibition of blood vessel formation (angiogenesis), (iii) inhibition of metastasis, (iv) induction of programmed cancer cell death (apoptosis), as well as (v) immunomodulatory activity through its influence on various signaling pathways [14,32,44,46,47,48,49,50,51]. Equally intriguing seems to be the possible synergistic effects of using EGCG in combination with known cytostatics, immunotherapeutic drugs, or other bioactive components [14,32,50,52,53,54]. Polychemotherapy based on combining EGCG with other substances may increase the polyphenol’s bioavailability and enhance the inhibition of cancer cell proliferation, induction of apoptosis, and suppression of angiogenesis or tumor growth [49,53].
The levels of EGCG found in plasma after administration may be significantly lower than the concentrations shown to be effective against cancer cells in in vitro tests (usually 10–100 µM) (Table 1). The efficient distribution of EGCG in the human body faces several natural barriers and processes [44,46]. Given the relatively low bioavailability of EGCG when taken orally due to factors such as low solubility in gastrointestinal fluids, slow intestinal absorption, limited permeability across the blood-brain barrier, and high susceptibility to degradation by intestinal microbiota [55], there is considerable interest in exploring the use of nanotechnology. Research in this area focuses on developing various systems, such as nanoparticles and nanoliposomes, that can effectively and selectively deliver the polyphenol in its native form to targeted sites. This approach may enhance the potential clinical applications of EGCG. Of particular note are nanosystems that could be successfully used in tests on BC cells and/or common gynecological cancers.
To the best of our knowledge, there has not been a recent review article summarizing the potential benefits of EGCG—whether used alone, in combination with other bioactive compounds, or enhanced through nanotechnology—in combating certain female cancers. Consequently, we searched the Google Scholar and PubMed databases for original studies published in the past 20 years that explored the effects of EGCG on breast, cervical, endometrial, and ovarian cancers (Table 1). We employed a variety of keyword combinations related to the polyphenol, such as “EGCG”, “epigallocatechin gallate”, or “epigallocatechin-3-gallate”, along with specific cancer types, including “breast cancer”, “cervical cancer”, “endometrial cancer”, as well as “ovarian cancer”. Furthermore, we reviewed the reference lists of the selected articles to identify additional relevant publications for inclusion in this review.

2.1. Breast Cancer

EGCG may modify estrogenic activity via ERα and ERα-dependent transcription [90]. Additionally, epidemiological studies indicate a possible association between culture-related green tea consumption and a lower risk of BC in some populations [66,91,92], suggesting potential clinical implications of EGCG in preventing and treating BC. Romano and Martel [93] summarized the broad spectrum of anticancer effects of EGCG based on the induction of apoptosis of BC cells by modulating the expression of various anti- and pro-apoptotic factors and angiogenesis inhibition. Concerning TNBC, EGCG inhibited the growth and proliferation of cancer cells by affecting estrogen receptor gene expression [94] or various cell signaling pathways and also increased cytostatic-induced apoptosis in these cells [95]. Moreover, reports on the effectiveness of synthetic analogs constructed based on EGCG [96,97,98], as well as the benefits of combining green tea and tamoxifen in the fight against BC in vitro and in vivo studies, seem to be interesting [99].

2.1.1. Anti-Breast Cancer Activity of EGCG

A study published in 2017 found that EGCG inhibited the proliferation of MCF-7 cells in a concentration-dependent manner, with an IC50 value of 37.7 µM [56]. Additionally, EGCG promoted apoptosis, with an apoptosis index of 5.83% observed at a concentration of 30 µM (p = 0.0124) [56]. The authors associated these effects with polyphenol’s action on the p53/Bcl-2 pathway [56]. EGCG reduced telomerase activity (40–55% according to [57]), whose level is elevated in >90% of BC cases, thus becoming the subject of special attention in the context of diagnostic tests and the development of new anticancer therapies [57,58]. Polyphenol had no adverse effect on the growth of normal mammary gland cells [57]. Other studies have shown that EGCG can cause changes at the (epi)genetic level [59,100,101], suppress the activity of apoptosis inhibitors [102], affect steroid receptors [103,104], T-type calcium channels [105] and important signaling pathways [106,107,108], as well as inhibit cell growth, tumorigenesis, or invasiveness by modulating miRNA expression or activating MMPs [60,61,109,110]. Wei et al. [62] focused on the possible effect of polyphenol on glucose metabolism, which was inhibited by it, causing a reduction in the growth of BC cells in preclinical models. The activity of EGCG limiting glucose uptake by cancer cells was also confirmed by other authors [111]. In the studies conducted on mice that were given EGCG at doses of 50–100 mg/kg/day in their drinking water for four weeks, researchers observed its effects on angiogenesis and breast tumor growth [63]. The results showed a significant difference between the treatment group and the control group, with tumor growth measuring 0.37 ± 0.15 in the EGCG group compared to 1.16 ± 0.30 in the control group that did not receive EGCG (p < 0.01) [63]. The therapy was also associated with changes in the modulation of HIF-1α and NF-κB activation, as well as VEGF expression [63]; the effects on HIF-1α and VEGF were also observed in independent studies. [64]. Polyphenol did not negatively affect body weight, heart function, angiogenesis, or VEGF expression in the heart and skeletal muscle of mice [63].
Moreover, EGCG inhibited the proliferation, migration, and invasiveness of cells derived from stem-like inflammatory BC [65] or TNBC [67,68,69,112], among others, due to blocking of Wnt/β-catenin signaling in the latter group of tumors [68,112]. In vitro studies revealed the potential benefits of EGCG in inhibiting Met (hepatocyte growth factor receptor) signaling, which is a prognostic indicator of oncological treatment and survival of patients with BC [70]. Cancer development was delayed in the EGCG-treated mice inoculated with MDA-MB-231 human BC cells compared to the control group [66]. Immunohistochemical studies of tumor tissue sections also showed that EGCG inhibited proliferation and induced cell apoptosis [66].
On the other hand, other potentially beneficial effects of EGCG use during anticancer therapy include possible alleviation of post-radiation dermatitis symptoms. In a phase I clinical trial in a group of 24 patients with BC undergoing mastectomy who had dermatitis caused by adjuvant radiotherapy, the use of a polyphenol solution (40–660 µmol/L) sprayed onto the irradiation field resulted in a significant reduction of bothersome symptoms (redness, pain, burning, itching) [113]. Similarly, in a phase II double-blind, placebo-controlled clinical trial involving 180 patients with BC undergoing postoperative radiotherapy, prophylactic use of EGCG solution (660 µmol/L) reduced the severity and incidence of dermatitis [114]. The local application of EGCG effectively alleviated radiotherapy’s side effects in other clinical studies [115]. The molecular mechanisms underlying the beneficial effects of EGCG in acute radiation-induced skin toxicity are complex, involving antibacterial and anti-inflammatory processes [115].

2.1.2. Combined Action of EGCG Against Breast Cancer

Using the MCF-7 BC cell model, Hsieh and Wu [116] tested whether the combination of EGCG, resveratrol, and γ-tocotrienol at suboptimal doses (10 µM) could induce enhanced inhibition of cell proliferation, modulate gene expression, or enhance antioxidant properties compared to the effects of each of the three phytochemicals administered alone. The synergy between the compounds was observed in the induction of quinone reductase NQO1, suggesting their possible beneficial cooperation [116]. EGCG, in combination with trastuzumab, may represent a novel strategy for the eradication of HER2-expressing BCs [117], and in combination with curcumin, increased cytotoxicity and intracellular levels of doxorubicin in drug-resistant MCF-7 cells [118]. Also, in studies on the MCF-7 cell line, EGCG increased the level of ERα, which made cancer cells more susceptible to the antagonist of this protein, tamoxifen [119]; other authors also confirmed the beneficial effect of the combination of EGCG and tamoxifen [120].
Importantly, Chisholm et al. [121] demonstrated synergistic cytotoxicity of EGCG combined with 4-hydroxytamoxifen (afimoxifene)—a selective ER modulator and an active metabolite of tamoxifen—on the MDA-MB-231 cell line. Another potential method of limiting the growth of highly invasive BC cells may be the administration of EGCG in combination with suberoylanilide hydroxamic acid (vorinostat), a histone deacetylase inhibitor associated with the inhibition of the transcription process [122,123]. The combined use of both compounds in vitro modulated, among others, the expression of cellular inhibitor of apoptosis 2 (cIAP2), proapoptotic caspase 7, oncogenic miRNA-221/222, and tumor suppressors p27 and PTEN, and finally reduced the metastatic potential and migration of cancer cells [122,123]. Studies on MDA-MB-231 cells showed that EGCG (25 µM) in combination with curcumin (3 µM) has a synergistic cytotoxic effect and promotes cell cycle arrest in the G2/M phase [124]. Combined treatment of mice with 25 mg/kg/day EGCG plus 200 mg/kg/day curcumin resulted in a 49% reduction in tumor volume compared to the control group (p < 0.05), which correlated with a 78 ± 6% decrease in VEGFR-1 protein expression level [124]. Both the proliferation and apoptosis of MDA-MB-231 cells were negatively affected by the administration of EGCG and tapentadol, an opioid analgesic, due to disrupted cell cycle progression (p < 0.05) [125]. EGCG has been identified as a compound that sensitizes tumor cells to paclitaxel, an oncological drug often used in BC treatment [126].

2.1.3. EGCG-Based Nanosystems for Breast Cancer Treatment

Considering the limitations resulting from the relatively low solubility and bioavailability of EGCG (please see Section 1.1), worldwide research is underway on the possible use of nanotechnology to improve the anticancer properties of compounds belonging to the group of natural polyphenols [127]. Zeng et al. [128] constructed two types of EGCG-loaded nanoparticles to enhance the targeting of MCF-7 cells with polyphenol. The system developed by the authors not only improved this parameter but also increased the effectiveness of EGCG in inhibiting cancer cell proliferation (IC50 = 470.5 ± 33.0 μg/mL, IC50 = 65.9 ± 0.4 μg/mL and IC50 = 66.6 ± 0.6 μg/mL, respectively, for EGCG and two types of EGCG-containing nanoparticles modified with folic acid and polyethylene glycol) [128]. Moreover, EGCG-containing nanoparticles were found to be more active and more effective in inhibiting the development of aggressive forms of BC in mice overexpressing CCN5 than free polyphenols [129]. In a recent study, nanoparticles composed of EGCG, phosphatidylcholine, doxorubicin, and anthocyanin procyanidin significantly inhibited the growth of BT-474, EMT-6, MCF-7, and MDA-MB-231 cell lines [130]. EGCG, together with paclitaxel in the form of PLGA-casein core/shell nanoparticles or liposomal co-delivery system, effectively reduced the invasiveness of MDA-MB-231 cells [131,132].
In turn, EGCG-encapsulated chitosan-coated nanoliposomes increased cancer cells’ stability and intracellular polyphenol content, inhibiting their proliferation and inducing apoptosis [133]. This type of system maintained anticancer efficacy at a concentration of 10 µM or lower, at which EGCG in its native form was ineffective [133]. Similar results were obtained for EGCG encapsulated in lipid nanoparticles, whose cytotoxicity was over 8 times higher against MDA-MB-231 cells than that of free polyphenol [134]. Combining a similar system with bombesin, a peptide targeting gastrin-releasing peptide receptors overexpressed in BCs, resulted in increased specificity and improved cytotoxicity against cancer cell lines [135].

2.2. Cervical Cancer

More and more studies indicate the possible use of EGCG or its analogs in regulating HPV infection and thus preventing the occurrence of CC [71,136,137]. The use of a dietary supplement in a regimen of two tablets a day for eight weeks, containing EGCG (200 mg), vitamin B12 (1 mg), folic acid (400 μg), and hyaluronic acid (50 mg), effectively alleviated precancerous cytological changes of the cervix caused by HPV infection [138], which was confirmed by other authors in subsequent years [139]. Moreover, EGCG had a beneficial effect on treating anogenital warts caused by HPV [140,141]. EGCG can induce apoptosis of CC cells by stimulating the degradation of oncogenes/oncoproteins and modulating the expression of tumor suppressor genes, as well as cause the degradation of extracellular matrix components, including MMPs, which play a role in cancer invasion, migration, and metastasis [37].

2.2.1. Anti-Cervical Cancer Activity of EGCG

The use of EGCG was associated with the degradation of E6 and E7 oncoproteins and increased expression of their associated tumor suppressor genes [142]. The limiting effect of EGCG on CC cell lines may be related to the modulation of miRNA expression [71] or fused toes homolog (FTS) mRNA via p53 [143]. Other potential mechanisms of the anticancer effect of EGCG may be the modulation of the angiogenesis signaling cascade [72] or the suppression of epithelial-mesenchymal transition (EMT) [73]. In studies on HPV-positive cancer cell lines (CaSki, HeLa, and SiHa), it was found that the addition of EGCG to cell culture inhibits their growth in a manner dependent on the concentration of polyphenol (p < 0.05) [71]. The limitation of CaSki (IC50 = 27.3 µM) and HeLa (IC50 = 47.9 µM) cell growth could result from cell cycle arrest and induction of apoptosis [74]. EGCG at a concentration of 5 µg/mL or 10 µg/mL increased the level of proapoptotic caspase 3 [144]. Other in vitro studies have shown that EGCG affects epigenetic modulators, signaling pathways, or suppressor genes [75,76]. The reduction in expression caused by EGCG concerned key signaling pathways (MAPK, PI3K, Wnt) regulating the cell cycle and the potential for metastasis of cancer cells [75]. Japanese researchers found that the anticancer mechanism of EGCG action may also be due to the inhibition of telomerase activity [77,78]. Moreover, a beneficial effect of EGCG on the inhibition of epidermal growth factor (EGF) activity has been described [79]. It has been established that elevated levels of EGF in the tumor microenvironment may be associated with the ability of tumor cells to metastasize; in cell culture of both HPV-positive and HPV-negative CC lines, EGCG effectively blocked the action of EGF [79].

2.2.2. Combined Action of EGCG Against Cervical Cancer

In the studies on the HeLa cell line, the anticancer effect of EGCG in combination with another polyphenolic compound present in black tea, theaflavin (Figure 3), was determined [145]. Both components acted synergistically, disrupting cancer cell growth due to microtubule depolymerization, which affected the reduction of PI3K/AKT pathway signaling and led to cell death by apoptosis [145]. An enhanced anticancer effect on HeLa cells was also demonstrated after using EGCG with eugenol and amarogentin [146]. In this case, the inhibitory effect on proliferation resulted from the decreased expression of cyclin D1 and the increased expression of cell cycle inhibitors [146]. Reports indicate that EGCG acts synergistically against cancer cell proliferation when combined with enoxacin, a fluoroquinolone agent showing antibacterial activity [147]. Similar beneficial effects were observed with EGCG (25 μM) and cisplatin (250 nM) [148]. EGCG and retinoic acid administered together induced apoptosis and inhibited telomerase activity in cancer cell lines, which was not observed for either of these natural compounds used alone [149]. On the other hand, according to Raish et al. [150], photodynamic therapy (PDT) combined with EGCG can induce a significant tumor suppressive response compared with PDT alone.

2.2.3. EGCG-Based Nanosystems for Cervical Cancer Treatment

There are few literature reports on the influence of nanotechnology on the improvement of anti-CC properties of EGCG. For example, the use of gold nanoparticles containing paclitaxel and EGCG functionalized with poly-D-lysine grafted polyethylene glycol and triphenylphosphonium cations (TPP+) targeting mitochondria caused apoptotic death of HeLa cells used in the tests [151].

2.3. Endometrial Cancer

Interestingly, the results of some studies suggest that EGCG may have an impact on reducing the risk of occurrence and slowing the course of EC [80,152,153], including the genetic Lynch syndrome [154]. Huang et al. [50] summarized the potential of EGCG against EC cell lines, indicating, among others, the modulating effect of the polyphenol on selected signaling pathways and the nuclear transcription factor Nrf2, which resulted in inhibition of proliferation and invasive potential, as well as induction of apoptosis. Apart from EGCG in its native form, interesting results were also obtained using in vitro and in vivo tests with derivatives (prodrugs) obtained by chemical modification of the polyphenol molecule [155,156,157].

Anti-Endometrial Cancer Activity of EGCG

The studies performed by Manohar et al. [80] indicated that EGCG may limit EC cell proliferation by inhibiting ERK activation and inducing apoptosis mediated by reactive oxygen species (ROS) and activation of p38. The action of EGCG (100 µM) caused a decrease in the amount of VEGF isolated from the supernatant of cultured EC cells (82.39 ± 19.54 pg/mL vs. 436.06 ± 86.74 pg/mL in the control group, p < 0.0001), suggesting the potential of the polyphenol to inhibit the angiogenesis process [81].

2.4. Ovarian Cancer

A growing number of literature reports indicate a beneficial effect of EGCG on the course of OC [50,86,88,158,159,160,161]. The mechanism of the anticancer effect of EGCG in this type of cancer is diverse. It may include inhibition of proliferation and induction of cell apoptosis by modulating the expression of NF-κB, apoptotic proteins (Bax, Bcl-2), or caspase 3 [50]. Other described molecular mechanisms of EGCG action included disruption of the activation of protein kinases, reduction of VEGF production, and MMP activities [158].

2.4.1. Anti-Ovarian Cancer Activity of EGCG

EGCG can inhibit the growth of OC cells and induce their apoptosis, including cisplatin-resistant cells [82], among others, by arresting the cell cycle, regulating proteins associated with it, or modulating p38 kinase and selected MMPs [83,84,85]. Polyphenols can also increase oxidative stress in cancer cells [162]. It has been proven that EGCG can exert a strong inhibitory effect on proliferation in tests on SKOV3 cells [86,87,163], which was accompanied by, among others, a decrease in the activity of the PTEN/AKT/mTOR pathway and an increase in the expression of the suppressor gene PTEN [86] or a reduction in the expression of AQP5 [163]. The effect on other important signaling pathways has also been described in the scientific literature [89]. The effect of EGCG on inhibiting OC growth has been observed in both in vitro and in vivo studies by various authors [88,89]. The molecular mechanism of action of EGCG consisted of the activation of the nuclear transcription factor FOXO3a responsible for cell hemostasis in response to changes in the extracellular and intracellular environment, including oxidative stress [88], and a decrease in basal and ET-1-induced angiogenesis and invasion tendency [89]. The use of EGCG also inhibited the expression of c-Myc, promoting the neoplastic process [88]. Moreover, EGCG administered to mice caused a stronger inhibitory effect on tumor growth compared to paclitaxel [86]. However, it should be noted that the dose of EGCG (50 mg/kg) was 10-fold higher than the dose of paclitaxel (5 mg/kg) [86].
In a single-arm phase II study of 16 patients with advanced stage III–IV OC who underwent cytoreductive surgery followed by chemotherapy, the effect of EGCG supplementation in maintenance therapy was investigated [160]. The study lasted 18 months, during which patients consumed 500 mL of EGCG-enriched green tea daily [160]. No disease relapses were observed in 5 of 16 women (~31%) enrolled in the study after 1.5 years of treatment [160]. At the same time, no toxicity was observed when drinking EGCG-enriched tea [160].

2.4.2. Combined Action of EGCG Against Ovarian Cancer

EGCG and sulforaphane (an organosulfur compound from the isothiocyanate group with potential chemopreventive properties) induced apoptosis in paclitaxel-resistant OC cells by reducing the expression of hTERT and Bcl-2 [164]; the combination of these two compounds also enhanced cisplatin-mediated apoptosis [165]. Rodriguez Torres et al. [159] found that EGCG inhibited the growth of ovarian CSCs in a dose-dependent manner. Src expression was reduced, which increased the cytotoxicity of cisplatin and paclitaxel; a decrease in the activity of JAK/STAT3 signaling as well as the transcription factor NANOG was noted, which improved the efficacy of the mentioned oncological drugs [159]. EGCG increased sensitivity to cisplatin by regulating the expression of the copper and cisplatin influx transporter CTR1 [166] or by increasing oxidative stress [162] and also acted synergistically in combination with this chemotherapeutic [167]. The synergism of sequenced combinations of EGCG and curcumin with cisplatin in killing human OC cells has also been demonstrated [168].
In the clinical trial by Kiselev et al. [161], a method of potential maintenance treatment of advanced OC was described based on indole-3-carbinol (an indole derivative with multi-target anticancer activity) and EGCG, which was used in three of the five arms of the study together with chemotherapy (in the fourth arm only chemotherapy was applied, and in the fifth only observational treatment was performed). The study included 284 women with clinically advanced stage III–IV serous OC [161]. After five years, both progression-free survival (PFS) and overall survival (OS) were prolonged in women receiving additional EGCG treatment [161]. The median OS was 60 months in the arms of the study using the combination of indole-3-carbinol and EGCG compared to 46 months without these compounds [161]. Additionally, the percentage of patients with tumor relapse with ascites after combined treatment was also significantly lower [161].

2.4.3. EGCG-Based Nanosystems for Ovarian Cancer Treatment

The nanoparticle-mediated systems can safely and target-specifically deliver various bioactive compounds, such as flavonoids, to tumors, improving their therapeutic potential [169,170,171]. The nanoparticles containing flavonoids can also enhance the effects of classical chemotherapy on cancer cells [171]. Several studies have already implemented the strategy of encapsulating EGCG for cancer treatment, including OC [172,173,174]. For example, to improve the targeting of cancer cells, Alizadeh et al. [175] prepared chitosan-coated silica nanoparticles and attached them to EGCG. The free amino groups of chitosan were then attached to the aptamer AS1411 [175]. The obtained nanomaterial improved the cytotoxic effect of EGCG (about 93% of cells underwent late apoptosis) [175]. Moreover, the functionalized nanoparticles caused a decrease in the number of cells in the S and G2/M phases, an increase in the G0/G1 phase, and cell arrest in the G1 phase [175]. A doxorubicin delivery system with folic acid-modified EGCG enhanced the uptake of an oncological drug by SKOV3 cells and effectively inhibited tumor growth in vivo [176]. On the other hand, Bae et al. [177] showed that micellar nanocomplexes with hyaluronic acid-EGCG conjugate and cisplatin enable efficient delivery of a platinum-based agent into CD44-overexpressing OC cells by receptor-mediated endocytosis (RME).

3. Conclusions

Epigallocatechin gallate (EGCG) is green tea’s primary polyphenol (catechin). EGCG has been widely studied for its potential health benefits, and its anticancer properties seem particularly interesting. Many authors have indicated polyphenol’s antiproliferative, antiangiogenic, and proapoptotic effects on various cancer cell lines and in vivo tests on animal models. EGCG can modulate signaling pathways associated with, among others, cell proliferation and division in multiple types of cancer, including breast cancer and gynecological cancers (cervical, endometrial, and ovarian). Despite the beneficial anticancer potential of EGCG, significant pharmacokinetic problems should be considered, which may affect the polyphenol’s ineffective systemic delivery and bioavailability. Various strategies have been adopted to improve the poor systemic bioavailability and cellular uptake of EGCG, including potential combination or polytherapy, which uses EGCG with one or more bioactive compounds. Furthermore, the approach using nanotechnology is fundamental, as it can help overcome the pharmacokinetic problems of EGCG by controlling its cytotoxicity and increasing bioavailability (the concept of nanochemoprevention).
This review article has been developed based on an analysis of data published over the last two decades. However, it should be noted that the topic of the potential beneficial effects of EGCG and other natural polyphenols has been so extensively explored that continuous updates will be necessary to keep up with emerging new research data. Given the encouraging anticancer activity of EGCG, it is important to validate and expand our understanding of the potential of this polyphenol against cancer cells. It should be made through both in vitro and in vivo tests, particularly using concentrations observed in plasma. The potential use of EGCG in chemopreventive treatment, adjuvant treatment, or supporting the action of classical oncological drugs also requires further verification, including clinical trials. Only on this basis will it be possible to draw more clear conclusions about the real anticancer potential of EGCG in the fight against selected malignant tumors in women.

Author Contributions

Writing—original draft preparation, A.M. and M.A.; writing—review and editing, M.A., A.H. and J.M.; supervision, A.H.; funding acquisition, A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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] [PubMed]
  2. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  3. Bizuayehu, H.M.; Ahmed, K.Y.; Kibret, G.D.; Dadi, A.F.; Belachew, S.A.; Bagade, T.; Tegegne, T.K.; Venchiarutti, R.L.; Kibret, K.T.; Hailegebireal, A.H.; et al. Global disparities of cancer and its projected burden in 2050. JAMA Netw. Open 2024, 7, e2443198. [Google Scholar] [CrossRef] [PubMed]
  4. Farhoosh, R.; Golmovahhed, G.A.; Khodaparast, M.H.H. Antioxidant activity of various extracts of old tea leaves and black tea wastes (Camellia sinensis L.). Food Chem. 2007, 100, 231–236. [Google Scholar] [CrossRef]
  5. Pan, X.; Niu, G.; Liu, H. Microwave-assisted extraction of tea polyphenols and tea caffeine from green tea leaves. Chem. Eng. Process. 2003, 42, 129–133. [Google Scholar] [CrossRef]
  6. Cabrera, C.; Artacho, R.; Giménez, R. Beneficial effects of green tea—A review. J. Am. Coll. Nutr. 2006, 25, 79–99. [Google Scholar] [CrossRef]
  7. Graham, H.N. Green tea composition, consumption, and polyphenol chemistry. Prev. Med. 1992, 21, 334–350. [Google Scholar] [CrossRef]
  8. Dai, J.; Sameen, D.E.; Zeng, Y.; Li, S.; Qin, W.; Liu, Y. An overview of tea polyphenols as bioactive agents for food packaging applications. LWT 2022, 167, 113845. [Google Scholar] [CrossRef]
  9. Wang, H.; Provan, G.J.; Helliwell, K. Tea flavonoids: Their functions, utilisation and analysis. Trends Food Sci. Technol. 2000, 11, 152–160. [Google Scholar] [CrossRef]
  10. Legeay, S.; Rodier, M.; Fillon, L.; Faure, S.; Clere, N. Epigallocatechin gallate: A review of its beneficial properties to prevent metabolic syndrome. Nutrients 2015, 7, 5443–5468. [Google Scholar] [CrossRef]
  11. Kochman, J.; Jakubczyk, K.; Antoniewicz, J.; Mruk, H.; Janda, K. Health benefits and chemical composition of Matcha green tea: A review. Molecules 2020, 26, 85. [Google Scholar] [CrossRef] [PubMed]
  12. Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial properties of green tea catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [PubMed]
  13. Zhao, T.; Li, C.; Wang, S.; Song, X. Green tea (Camellia sinensis): A review of its phytochemistry, pharmacology, and toxicology. Molecules 2022, 27, 3909. [Google Scholar] [CrossRef] [PubMed]
  14. Li, X.X.; Liu, C.; Dong, S.L.; Ou, C.S.; Lu, J.L.; Ye, J.H.; Liang, Y.R.; Zheng, X.Q. Anticarcinogenic potentials of tea catechins. Front. Nutr. 2022, 9, 1060783. [Google Scholar] [CrossRef]
  15. Zeb, F.; Naqeeb, H.; Osaili, T.; Faris, M.E.; Ismail, L.C.; Obaid, R.S.; Naja, F.; Radwan, H.; Hasan, H.; Hashim, M.; et al. Molecular crosstalk between polyphenols and gut microbiota in cancer prevention. Nutr. Res. 2024, 124, 21–42. [Google Scholar] [CrossRef]
  16. Upadhyay, P.K.; Singh, S.; Vishwakarma, V.K. Natural polyphenols in cancer management: Promising role, mechanisms, and chemistry. Curr. Pharm. Biotechnol. 2024, 25, 694–712. [Google Scholar] [CrossRef]
  17. Liu, Z.; Vincken, J.P.; de Bruijn, W.J.C. Tea phenolics as prebiotics. Trends Food Sci. Technol. 2022, 127, 156–168. [Google Scholar] [CrossRef]
  18. Chu, C.; Deng, J.; Man, Y.; Qu, Y. Green tea extracts epigallocatechin-3-gallate for different treatments. Biomed. Res. Int. 2017, 2017, 5615647. [Google Scholar] [CrossRef]
  19. Rice-Evans, C. Implications of the mechanisms of action of tea polyphenols as antioxidants in vitro for chemoprevention in humans. Exp. Biol. Med. 1999, 220, 262–266. [Google Scholar] [CrossRef]
  20. Lee, M.J.; Maliakal, P.; Chen, L.; Meng, X.; Bondoc, F.Y.; Prabhu, S.; Lambert, G.; Mohr, S.; Yang, C.S. Pharmacokinetics of tea catechins after ingestion of green tea and (−)-epigallocatechin-3-gallate by humans: Formation of different metabolites and individual variability. Cancer Epidemiol. Biomark. Prev. 2002, 11, 1025–1032. [Google Scholar]
  21. Chen, L.; Lee, M.J.; Li, H.; Yang, C.S. Absorption, distribution, elimination of tea polyphenols in rats. Drug Metab. Dispos. 1997, 25, 1045–1050. [Google Scholar] [PubMed]
  22. Yang, C.S.; Sang, S.; Lambert, J.D.; Lee, M.J. Bioavailability issues in studying the health effects of plant polyphenolic compounds. Mol. Nutr. Food Res. 2008, 52, S139–S151. [Google Scholar] [CrossRef] [PubMed]
  23. Chow, H.H.; Cai, Y.; Alberts, D.S.; Hakim, I.; Dorr, R.; Shahi, F.; Crowell, J.A.; Yang, C.S.; Hara, Y. Phase I pharmacokinetic study of tea polyphenols following single-dose administration of epigallocatechin gallate and polyphenon E. Cancer Epidemiol. Biomark. Prev. 2001, 10, 53–58. [Google Scholar]
  24. Chow, H.H.S.; Cai, Y.; Hakim, I.A.; Crowell, J.A.; Shahi, F.; Brooks, C.A.; Dorr, R.T.; Hara, Y.; Alberts, D.S. Pharmacokinetics and safety of green tea polyphenols after multiple-dose administration of epigallocatechin gallate and polyphenon E in healthy individuals. Clin. Cancer Res. 2003, 9, 3312–3319. [Google Scholar] [PubMed]
  25. Chow, H.H.S.; Hakim, I.A.; Vining, D.R.; Crowell, J.A.; Ranger-Moore, J.; Chew, W.M.; Celaya, C.A.; Rodney, S.R.; Hara, Y.; Alberts, D.S. Effects of dosing condition on the oral bioavailability of green tea catechins after single-dose administration of polyphenon E in healthy individuals. Clin. Cancer Res. 2005, 11, 4627–4633. [Google Scholar] [CrossRef] [PubMed]
  26. Younes, M.; Aggett, P.; Aguilar, F.; Crebelli, R.; Dusemund, B.; Filipič, M.; Frutos, M.J.; Galtier, P.; Gott, D.; Gundert-Remy, U.; et al. Scientific opinion on the safety of green tea catechins. EFSA J. 2018, 16, 5239. [Google Scholar] [CrossRef]
  27. Nakagawa, K.; Nakayama, K.; Nakamura, M.; Sookwong, P.; Tsuduki, T.; Niino, H.; Kimura, F.; Miyazawa, T. Effects of co-administration of tea epigallocatechin-3-gallate (EGCG) and caffeine on absorption and metabolism of EGCG in humans. Biosci. Biotechnol. Biochem. 2009, 73, 2014–2017. [Google Scholar] [CrossRef]
  28. Crozier, A.; Jaganath, I.B.; Clifford, M.N. Dietary phenolics: Chemistry, bioavailability and effects on health. Nat. Prod. Rep. 2009, 26, 1001. [Google Scholar] [CrossRef]
  29. Chen, T.; Yang, C.S. Biological fates of tea polyphenols and their interactions with microbiota in the gastrointestinal tract: Implications on health effects. Crit. Rev. Food Sci. Nutr. 2020, 60, 2691–2709. [Google Scholar] [CrossRef]
  30. Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 2012, 148, 1258–1270. [Google Scholar] [CrossRef]
  31. Kohri, T.; Matsumoto, N.; Yamakawa, M.; Suzuki, M.; Nanjo, F.; Hara, Y.; Oku, N. Metabolic fate of (−)-[4-(3)H]epigallocatechin gallate in rats after oral administration. J. Agric. Food Chem. 2001, 49, 4102–4112. [Google Scholar] [CrossRef] [PubMed]
  32. Marín, V.; Burgos, V.; Pérez, R.; Maria, D.A.; Pardi, P.; Paz, C. The potential role of epigallocatechin-3-gallate (EGCG) in breast cancer treatment. Int. J. Mol. Sci. 2023, 24, 10737. [Google Scholar] [CrossRef] [PubMed]
  33. Takagaki, A.; Nanjo, F. Metabolism of (−)-epigallocatechin gallate by rat intestinal flora. J. Agric. Food Chem. 2010, 58, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
  34. Mena, P.; Bresciani, L.; Brindani, N.; Ludwig, I.A.; Pereira-Caro, G.; Angelino, D.; Llorach, R.; Calani, L.; Brighenti, F.; Clifford, M.N.; et al. Phenyl-γ-valerolactones and phenylvaleric acids, the main colonic metabolites of flavan-3-ols: Synthesis, analysis, bioavailability, and bioactivity. Nat. Prod. Rep. 2019, 36, 714–752. [Google Scholar] [CrossRef]
  35. van der Hooft, J.J.J.; de Vos, R.C.H.; Mihaleva, V.; Bino, R.J.; Ridder, L.; de Roo, N.; Jacobs, D.M.; van Duynhoven, J.P.M.; Vervoort, J. Structural elucidation and quantification of phenolic conjugates present in human urine after tea intake. Anal. Chem. 2012, 84, 7263–7271. [Google Scholar] [CrossRef]
  36. Zagami, P.; Carey, L.A. Triple negative breast cancer: Pitfalls and progress. NPJ Breast Cancer 2022, 8, 95. [Google Scholar] [CrossRef]
  37. Laganà, A.S.; Chiantera, V.; Gerli, S.; Proietti, S.; Lepore, E.; Unfer, V.; Carugno, J.; Favilli, A. Preventing persistence of HPV infection with natural molecules. Pathogens 2023, 12, 416. [Google Scholar] [CrossRef]
  38. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  39. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef]
  40. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer stem cells—Origins and biomarkers: Perspectives for targeted personalized therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef]
  41. Medema, J.P. Cancer stem cells: The challenges ahead. Nat. Cell Biol. 2013, 15, 338–344. [Google Scholar] [CrossRef] [PubMed]
  42. Naujokat, C.; McKee, D.L. The “big five” phytochemicals targeting cancer stem cells: Curcumin, EGCG, sulforaphane, resveratrol and genistein. Curr. Med. Chem. 2021, 28, 4321–4342. [Google Scholar] [CrossRef] [PubMed]
  43. Pistollato, F.; Calderón Iglesias, R.; Ruiz, R.; Aparicio, S.; Crespo, J.; Dzul Lopez, L.; Giampieri, F.; Battino, M. The use of natural compounds for the targeting and chemoprevention of ovarian cancer. Cancer Lett. 2017, 411, 191–200. [Google Scholar] [CrossRef] [PubMed]
  44. Talib, W.H.; Awajan, D.; Alqudah, A.; Alsawwaf, R.; Althunibat, R.; Abu AlRoos, M.; Al Safadi, A.; Abu Asab, S.; Hadi, R.W.; Al Kury, L.T. Targeting cancer hallmarks with epigallocatechin gallate (EGCG): Mechanistic basis and therapeutic targets. Molecules 2024, 29, 1373. [Google Scholar] [CrossRef]
  45. Hung, S.W.; Li, Y.; Chen, X.; Chu, K.O.; Zhao, Y.; Liu, Y.; Guo, X.; Man, G.C.W.; Wang, C.C. Green tea epigallocatechin-3-gallate regulates autophagy in male and female reproductive cancer. Front. Pharmacol. 2022, 13, 906746. [Google Scholar] [CrossRef]
  46. Min, K.; Kwon, T.K. Anticancer effects and molecular mechanisms of epigallocatechin-3-gallate. Integr. Med. Res. 2014, 3, 16–24. [Google Scholar] [CrossRef]
  47. Ferrari, E.; Bettuzzi, S.; Naponelli, V. The potential of epigallocatechin gallate (EGCG) in targeting autophagy for cancer treatment: A narrative review. Int. J. Mol. Sci. 2022, 23, 6075. [Google Scholar] [CrossRef]
  48. Alam, M.; Ali, S.; Ashraf, G.M.; Bilgrami, A.L.; Yadav, D.K.; Hassan, M.I. Epigallocatechin 3-gallate: From green tea to cancer therapeutics. Food Chem. 2022, 379, 132135. [Google Scholar] [CrossRef]
  49. Rady, I.; Mohamed, H.; Rady, M.; Siddiqui, I.A.; Mukhtar, H. Cancer preventive and therapeutic effects of EGCG, the major polyphenol in green tea. Egypt. J. Basic Appl. Sci. 2018, 5, 1–23. [Google Scholar] [CrossRef]
  50. Huang, Y.J.; Wang, K.L.; Chen, H.Y.; Chiang, Y.F.; Hsia, S.M. Protective effects of epigallocatechin gallate (EGCG) on endometrial, breast, and ovarian cancers. Biomolecules 2020, 10, 1481. [Google Scholar] [CrossRef]
  51. Cai, J.; Qiao, Y.; Chen, L.; Lu, Y.; Zheng, D. Regulation of the Notch signaling pathway by natural products for cancer therapy. J. Nutr. Biochem. 2024, 123, 109483. [Google Scholar] [CrossRef] [PubMed]
  52. Cheng, Z.; Zhang, Z.; Han, Y.; Wang, J.; Wang, Y.; Chen, X.; Shao, Y.; Cheng, Y.; Zhou, W.; Lu, X.; et al. A review on anti-cancer effect of green tea catechins. J. Funct. Foods 2020, 74, 104172. [Google Scholar] [CrossRef]
  53. Suganuma, M.; Saha, A.; Fujiki, H. New cancer treatment strategy using combination of green tea catechins and anticancer drugs. Cancer Sci. 2011, 102, 317–323. [Google Scholar] [CrossRef]
  54. Fujiki, H.; Sueoka, E.; Watanabe, T.; Suganuma, M. Synergistic enhancement of anticancer effects on numerous human cancer cell lines treated with the combination of EGCG, other green tea catechins, and anticancer compounds. J. Cancer Res. Clin. Oncol. 2015, 141, 1511–1522. [Google Scholar] [CrossRef]
  55. Dai, W.; Ruan, C.; Zhang, Y.; Wang, J.; Han, J.; Shao, Z.; Sun, Y.; Liang, J. Bioavailability enhancement of EGCG by structural modification and nano-delivery: A review. J. Funct. Foods 2020, 65, 103732. [Google Scholar] [CrossRef]
  56. Huang, C.Y.; Han, Z.; Li, X.; Xie, H.H.; Zhu, S.S. Mechanism of EGCG promoting apoptosis of MCF-7 cell line in human breast cancer. Oncol. Lett. 2017, 14, 3623–3627. [Google Scholar] [CrossRef]
  57. Mittal, A.; Pate, M.; Wylie, R.; Tollefsbol, T.; Katiyar, S. EGCG down-regulates telomerase in human breast carcinoma MCF-7 cells, leading to suppression of cell viability and induction of apoptosis. Int. J. Oncol. 2004, 24, 703–710. [Google Scholar] [CrossRef]
  58. Moradzadeh, M.; Hosseini, A.; Erfanian, S.; Rezaei, H. Epigallocatechin-3-gallate promotes apoptosis in human breast cancer T47D cells through down-regulation of PI3K/AKT and telomerase. Pharmacol. Rep. 2017, 69, 924–928. [Google Scholar] [CrossRef]
  59. Sheng, J.; Shi, W.; Guo, H.; Long, W.; Wang, Y.; Qi, J.; Liu, J.; Xu, Y. The inhibitory effect of (−)-epigallocatechin-3-gallate on breast cancer progression via reducing SCUBE2 methylation and DNMT activity. Molecules 2019, 24, 2899. [Google Scholar] [CrossRef]
  60. Zan, L.; Chen, Q.; Zhang, L.; Li, X. Epigallocatechin gallate (EGCG) suppresses growth and tumorigenicity in breast cancer cells by downregulation of miR-25. Bioengineered 2019, 10, 374–382. [Google Scholar] [CrossRef]
  61. Sen, T.; Moulik, S.; Dutta, A.; Choudhury, P.R.; Banerji, A.; Das, S.; Roy, M.; Chatterjee, A. Multifunctional effect of epigallocatechin-3-gallate (EGCG) in downregulation of gelatinase-A (MMP-2) in human breast cancer cell line MCF-7. Life Sci. 2009, 84, 194–204. [Google Scholar] [CrossRef] [PubMed]
  62. Wei, R.; Mao, L.; Xu, P.; Zheng, X.; Hackman, R.M.; Mackenzie, G.G.; Wang, Y. Suppressing glucose metabolism with epigallocatechin-3-gallate (EGCG) reduces breast cancer cell growth in preclinical models. Food Funct. 2018, 9, 5682–5696. [Google Scholar] [CrossRef] [PubMed]
  63. Gu, J.W.; Makey, K.L.; Tucker, K.B.; Chinchar, E.; Mao, X.; Pei, I.; Thomas, E.Y.; Miele, L. EGCG, a major green tea catechin suppresses breast tumor angiogenesis and growth via inhibiting the activation of HIF-1α and NFκB, and VEGF expression. Vasc. Cell 2013, 5, 9. [Google Scholar] [CrossRef] [PubMed]
  64. Luo, H.; Xu, M.; Zhong, W.; Cui, Z.; Liu, F.; Zhou, K.; Li, X. EGCG decreases the expression of HIF-1α and VEGF and cell growth in MCF-7 breast cancer cells. J. BUON 2014, 19, 435–439. [Google Scholar]
  65. Mineva, N.D.; Paulson, K.E.; Naber, S.P.; Yee, A.S.; Sonenshein, G.E. Epigallocatechin-3-gallate inhibits stem-like inflammatory breast cancer cells. PLoS ONE 2013, 8, e73464. [Google Scholar] [CrossRef]
  66. Thangapazham, R.L.; Singh, A.K.; Sharma, A.; Warren, J.; Gaddipati, J.P.; Maheshwari, R.K. Green tea polyphenols and its constituent epigallocatechin gallate inhibits proliferation of human breast cancer cells in vitro and in vivo. Cancer Lett. 2007, 245, 232–241. [Google Scholar] [CrossRef]
  67. Braicu, C.; Gherman, C.D.; Irimie, A.; Berindan-Neagoe, I. Epigallocatechin-3-gallate (EGCG) inhibits cell proliferation and migratory behaviour of triple negative breast cancer cells. J. Nanosci. Nanotechnol. 2013, 13, 632–637. [Google Scholar] [CrossRef]
  68. Kim, J.; Zhang, X.; Rieger-Christ, K.M.; Summerhayes, I.C.; Wazer, D.E.; Paulson, K.E.; Yee, A.S. Suppression of Wnt signaling by the green tea compound (–)-epigallocatechin 3-gallate (EGCG) in invasive breast cancer cells. J. Biol. Chem. 2006, 281, 10865–10875. [Google Scholar] [CrossRef]
  69. Thangapazham, R.L.; Passi, N.; Maheshwari, R.K. Green tea polyphenol and epigallocatechin gallate induce apoptosis and inhibit invasion in human breast cancer cells. Cancer Biol. Ther. 2007, 6, 1938–1943. [Google Scholar] [CrossRef]
  70. Bigelow, R.L.H.; Cardelli, J.A. The green tea catechins, (−)-epigallocatechin-3-gallate (EGCG) and (−)-epicatechin-3-gallate (ECG), inhibit HGF/Met signaling in immortalized and tumorigenic breast epithelial cells. Oncogene 2006, 25, 1922–1930. [Google Scholar] [CrossRef]
  71. Zhu, Y.; Huang, Y.; Liu, M.; Yan, Q.; Zhao, W.; Yang, P.; Gao, Q.; Wei, J.; Zhao, W.; Ma, L. Epigallocatechin gallate inhibits cell growth and regulates miRNA expression in cervical carcinoma cell lines infected with different high-risk human papillomavirus subtypes. Exp. Ther. Med. 2018, 17, 1742–1748. [Google Scholar] [CrossRef] [PubMed]
  72. Tudoran, O.; Soritau, O.; Balacescu, O.; Balacescu, L.; Braicu, C.; Rus, M.; Gherman, C.; Virag, P.; Irimie, F.; Berindan-Neagoe, I. Early transcriptional pattern of angiogenesis induced by EGCG treatment in cervical tumour cells. J. Cell. Mol. Med. 2012, 16, 520–530. [Google Scholar] [CrossRef] [PubMed]
  73. Panji, M.; Behmard, V.; Zare, Z.; Malekpour, M.; Nejadbiglari, H.; Yavari, S.; Nayerpour dizaj, T.; Safaeian, A.; Maleki, N.; Abbasi, M.; et al. Suppressing effects of green tea extract and epigallocatechin-3-gallate (EGCG) on TGF-β-induced epithelial-to-mesenchymal transition via ROS/Smad signaling in human cervical cancer cells. Gene 2021, 794, 145774. [Google Scholar] [CrossRef] [PubMed]
  74. Qiao, Y.; Cao, J.; Xie, L.; Shi, X. Cell growth inhibition and gene expression regulation by (−)-epigallocatechin-3-gallate in human cervical cancer cells. Arch. Pharm. Res. 2009, 32, 1309–1315. [Google Scholar] [CrossRef]
  75. Kedhari Sundaram, M.; Haque, S.; Somvanshi, P.; Bhardwaj, T.; Hussain, A. Epigallocatechin gallate inhibits HeLa cells by modulation of epigenetics and signaling pathways. 3 Biotech. 2020, 10, 484. [Google Scholar] [CrossRef]
  76. Sharma, C.; Nusri, Q.E.A.; Begum, S.; Javed, E.; Rizvi, T.A.; Hussain, A. (−)-Epigallocatechin-3-gallate induces apoptosis and inhibits invasion and migration of human cervical cancer cells. Asian Pac. J. Cancer Prev. 2012, 13, 4815–4822. [Google Scholar] [CrossRef]
  77. Noguchi, M.; Yokoyama, M.; Watanabe, S.; Uchiyama, M.; Nakao, Y.; Hara, K.; Iwasaka, T. Inhibitory effect of the tea polyphenol, (−)-epigallocatechin gallate, on growth of cervical adenocarcinoma cell lines. Cancer Lett. 2006, 234, 135–142. [Google Scholar] [CrossRef]
  78. Yokoyama, M.; Noguchi, M.; Nakao, Y.; Pater, A.; Iwasaka, T. The tea polyphenol, (−)-epigallocatechin gallate effects on growth, apoptosis, and telomerase activity in cervical cell lines. Gynecol. Oncol. 2004, 92, 197–204. [Google Scholar] [CrossRef]
  79. Sabanayagam, R.; Krishnamoorthy, S.; Gnanagurusamy, J.; Muruganatham, B.; Muthusami, S. EGCG attenuate EGF triggered matrix abundance and migration in HPV positive and HPV negative cervical cancer cells. Med. Oncol. 2023, 40, 261. [Google Scholar] [CrossRef]
  80. Manohar, M.; Fatima, I.; Saxena, R.; Chandra, V.; Sankhwar, P.L.; Dwivedi, A. (−)-Epigallocatechin-3-gallate induces apoptosis in human endometrial adenocarcinoma cells via ROS generation and p38 MAP kinase activation. J. Nutr. Biochem. 2013, 24, 940–947. [Google Scholar] [CrossRef]
  81. Dann, J.M.; Sykes, P.H.; Mason, D.R.; Evans, J.J. Regulation of vascular endothelial growth factor in endometrial tumour cells by resveratrol and EGCG. Gynecol. Oncol. 2009, 113, 374–378. [Google Scholar] [CrossRef] [PubMed]
  82. Li, X.; Hou, Y.; Han, G.; Yang, Y.; Wang, S.; Lv, X.; Gao, M. S100A4/NF-κB axis mediates the anticancer effect of epigallocatechin-3-gallate in platinum-resistant ovarian cancer. iScience 2024, 27, 108885. [Google Scholar] [CrossRef] [PubMed]
  83. Huh, S.W.; Bae, S.M.; Kim, Y.-W.; Lee, J.M.; Namkoong, S.E.; Lee, I.P.; Kim, S.H.; Kim, C.K.; Ahn, W.S. Anticancer effects of (−)-epigallocatechin-3-gallate on ovarian carcinoma cell lines. Gynecol. Oncol. 2004, 94, 760–768. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, Y.W.; Bae, S.M.; Lee, J.M.; Namkoong, S.E.; Han, S.J.; Lee, B.R.; Lee, I.P.; Kim, S.H.; Lee, Y.J.; Kim, C.K.; et al. Activity of green tea polyphenol epigallocatechin-3-gallate against ovarian carcinoma cell lines. Cancer Res. Treat. 2004, 36, 315. [Google Scholar] [CrossRef]
  85. Wang, F.; Chang, Z.; Fan, Q.; Wang, L. Epigallocatechin-3-gallate inhibits the proliferation and migration of human ovarian carcinoma cells by modulating p38 kinase and matrix metalloproteinase-2. Mol. Med. Rep. 2014, 9, 1085–1089. [Google Scholar] [CrossRef]
  86. Qin, J.; Fu, M.; Wang, J.; Huang, F.; Liu, H.; Huangfu, M.; Yu, D.; Liu, H.; Li, X.; 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]
  87. Rao, S.D.; Pagidas, K. Epigallocatechin-3-gallate, a natural polyphenol, inhibits cell proliferation and induces apoptosis in human ovarian cancer cells. Anticancer Res. 2010, 30, 2519–2523. [Google Scholar]
  88. Zhang, Z.; Zhang, Q.; Yu, Y.; Su, S. Epigallocatechin gallate inhibits ovarian cancer cell growth and induces cell apoptosis via activation of FOXO3A. Vitr. Cell. Dev. Biol. Anim. 2023, 59, 739–746. [Google Scholar] [CrossRef]
  89. Spinella, F.; Rosanò, L.; Di Castro, V.; Decandia, S.; Albini, A.; Nicotra, M.R.; Natali, P.G.; Bagnato, A. Green tea polyphenol epigallocatechin-3-gallate inhibits the endothelin axis and downstream signaling pathways in ovarian carcinoma. Mol. Cancer Ther. 2006, 5, 1483–1492. [Google Scholar] [CrossRef]
  90. Baker, K.M.; Bauer, A.C. Green tea catechin, EGCG, suppresses PCB 102-induced proliferation in estrogen-sensitive breast cancer cells. Int. J. Breast Cancer 2015, 2015, 163591. [Google Scholar] [CrossRef]
  91. Zhang, M.; Holman, C.D.J.; Huang, J.P.; Xie, X. Green tea and the prevention of breast cancer: A case-control study in Southeast China. Carcinogenesis 2006, 28, 1074–1078. [Google Scholar] [CrossRef] [PubMed]
  92. Ogunleye, A.A.; Xue, F.; Michels, K.B. Green tea consumption and breast cancer risk or recurrence: A meta-analysis. Breast Cancer Res. Treat. 2010, 119, 477–484. [Google Scholar] [CrossRef] [PubMed]
  93. Romano, A.; Martel, F. The role of EGCG in breast cancer prevention and therapy. Mini Rev. Med. Chem. 2021, 21, 883–898. [Google Scholar] [CrossRef] [PubMed]
  94. Gianfredi, V.; Vannini, S.; Moretti, M.; Villarini, M.; Bragazzi, N.L.; Izzotti, A.; Nucci, D. Sulforaphane and epigallocatechin gallate restore estrogen receptor expression by modulating epigenetic events in the breast cancer cell line MDA-MB-231: A systematic review and meta-analysis. J. Nutrigenet. Nutr. 2017, 10, 126–135. [Google Scholar] [CrossRef]
  95. Bimonte, S.; Cascella, M.; Barbieri, A.; Arra, C.; Cuomo, A. Current shreds of evidence on the anticancer role of EGCG in triple negative breast cancer: An update of the current state of knowledge. Infect. Agents Cancer 2020, 15, 2. [Google Scholar] [CrossRef]
  96. Meeran, S.M.; Patel, S.N.; Chan, T.H.; Tollefsbol, T.O. A novel prodrug of epigallocatechin-3-gallate: Differential epigenetic HTERT repression in human breast cancer cells. Cancer Prev. Res. 2011, 4, 1243–1254. [Google Scholar] [CrossRef]
  97. Giró-Perafita, A.; Rabionet, M.; Planas, M.; Feliu, L.; Ciurana, J.; Ruiz-Martínez, S.; Puig, T. EGCG-derivative G28 shows high efficacy inhibiting the mammosphere-forming capacity of sensitive and resistant TNBC models. Molecules 2019, 24, 1027. [Google Scholar] [CrossRef]
  98. Chen, D.; Pamu, S.; Cui, Q.; Chan, T.H.; Dou, Q.P. Novel epigallocatechin gallate (EGCG) analogs activate AMP-activated protein kinase pathway and target cancer stem cells. Bioorg. Med. Chem. 2012, 20, 3031–3037. [Google Scholar] [CrossRef]
  99. Sartippour, M.R.; Pietras, R.; Marquez-Garban, D.C.; Chen, H.-W.; Heber, D.; Henning, S.M.; Sartippour, G.; Zhang, L.; Lu, M.; Weinberg, O.; et al. The combination of green tea and tamoxifen is effective against breast cancer. Carcinogenesis 2006, 27, 2424–2433. [Google Scholar] [CrossRef]
  100. Guo, S.; Yang, S.; Taylor, C.; Sonenshein, G.E. Green tea polyphenol epigallocatechin-3 gallate (EGCG) affects gene expression of breast cancer cells transformed by the carcinogen 7,12-dimethylbenz[a]anthracene. J. Nutr. 2005, 135, 2978S–2986S. [Google Scholar] [CrossRef]
  101. Belguise, K.; Guo, S.; Sonenshein, G.E. Activation of FOXO3a by the green tea polyphenol epigallocatechin-3-gallate induces estrogen receptor α expression reversing invasive phenotype of breast cancer cells. Cancer Res. 2007, 67, 5763–5770. [Google Scholar] [CrossRef] [PubMed]
  102. Tang, Y.; Zhao, D.; Elliott, S.; Zhao, W.; Curiel, T.; Beckman, B.; Burow, M. Epigallocatechin-3 gallate induces growth inhibition and apoptosis in human breast cancer cells through survivin suppression. Int. J. Oncol. 2007, 31, 705–711. [Google Scholar] [CrossRef] [PubMed]
  103. De Amicis, F.; Russo, A.; Avena, P.; Santoro, M.; Vivacqua, A.; Bonofiglio, D.; Mauro, L.; Aquila, S.; Tramontano, D.; Fuqua, S.A.; et al. In vitro mechanism for downregulation of ER-α expression by epigallocatechin gallate in ER+/PR+ human breast cancer cells. Mol. Nutr. Food Res. 2013, 57, 840–853. [Google Scholar] [CrossRef] [PubMed]
  104. Hallman, K.; Aleck, K.; Quigley, M.; Dwyer, B.; Lloyd, V.; Szmyd, M.; Dinda, S. The regulation of steroid receptors by epigallocatechin-3-gallate in breast cancer cells. Breast Cancer 2017, 9, 365–373. [Google Scholar] [CrossRef]
  105. Ranzato, E.; Magnelli, V.; Martinotti, S.; Waheed, Z.; Cain, S.M.; Snutch, T.P.; Marchetti, C.; Burlando, B. Epigallocatechin-3-gallate elicits Ca2+ spike in MCF-7 breast cancer cells: Essential role of Cav3.2 channels. Cell Calcium 2014, 56, 285–295. [Google Scholar] [CrossRef]
  106. Tu, S.H.; Ku, C.Y.; Ho, C.T.; Chen, C.S.; Huang, C.S.; Lee, C.H.; Chen, L.C.; Pan, M.H.; Chang, H.W.; Chang, C.H.; et al. Tea polyphenol (−)-epigallocatechin-3-gallate inhibits nicotine- and estrogen-induced α9-nicotinic acetylcholine receptor upregulation in human breast cancer cells. Mol. Nutr. Food Res. 2011, 55, 455–466. [Google Scholar] [CrossRef]
  107. Zhang, Y.; Han, G.; Fan, B.; Zhou, Y.; Zhou, X.; Wei, L.; Zhang, J. Green tea (−)-epigallocatechin-3-gallate down-regulates VASP expression and inhibits breast cancer cell migration and invasion by attenuating Rac1 activity. Eur. J. Pharmacol. 2009, 606, 172–179. [Google Scholar] [CrossRef]
  108. Pan, M.H.; Lin, C.C.; Lin, J.K.; Chen, W.J. Tea polyphenol (−)-epigallocatechin 3-gallate suppresses heregulin-β1-induced fatty acid synthase expression in human breast cancer cells by inhibiting phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase cascade signaling. J. Agric. Food Chem. 2007, 55, 5030–5037. [Google Scholar] [CrossRef]
  109. Sen, T.; Chatterjee, A. Epigallocatechin-3-gallate (EGCG) downregulates EGF-induced MMP-9 in breast cancer cells: Involvement of integrin receptor α5β1 in the process. Eur. J. Nutr. 2011, 50, 465–478. [Google Scholar] [CrossRef]
  110. Sen, T.; Dutta, A.; Chatterjee, A. Epigallocatechin-3-gallate (EGCG) downregulates gelatinase-B (MMP-9) by involvement of FAK/ERK/NFκB and AP-1 in the human breast cancer cell line MDA-MB-231. Anticancer Drugs 2010, 21, 632–644. [Google Scholar] [CrossRef]
  111. Moreira, L.; Araújo, I.; Costa, T.; Correia-Branco, A.; Faria, A.; Martel, F.; Keating, E. Quercetin and epigallocatechin gallate inhibit glucose uptake and metabolism by breast cancer cells by an estrogen receptor-independent mechanism. Exp. Cell Res. 2013, 319, 1784–1795. [Google Scholar] [CrossRef] [PubMed]
  112. Hong, O.Y.; Noh, E.M.; Jang, H.Y.; Lee, Y.R.; Lee, B.K.; Jung, S.H.; Kim, J.S.; Youn, H.J. Epigallocatechin gallate inhibits the growth of MDA-MB-231 breast cancer cells via inactivation of the β-catenin signaling pathway. Oncol. Lett. 2017, 14, 441–446. [Google Scholar] [CrossRef] [PubMed]
  113. Zhao, H.; Zhu, W.; Jia, L.; Sun, X.; Chen, G.; Zhao, X.; Li, X.; Meng, X.; Kong, L.; Xing, L.; et al. Phase I study of topical epigallocatechin-3-gallate (EGCG) in patients with breast cancer receiving adjuvant radiotherapy. Br. J. Radiol. 2016, 89, 20150665. [Google Scholar] [CrossRef] [PubMed]
  114. Zhao, H.; Zhu, W.; Zhao, X.; Li, X.; Zhou, Z.; Zheng, M.; Meng, X.; Kong, L.; Zhang, S.; He, D.; et al. Efficacy of epigallocatechin-3-gallate in preventing dermatitis in patients with breast cancer receiving postoperative radiotherapy. JAMA Dermatol. 2022, 158, 779. [Google Scholar] [CrossRef]
  115. Zhu, W.; Jia, L.; Chen, G.; Zhao, H.; Sun, X.; Meng, X.; Zhao, X.; Xing, L.; Yu, J.; Zheng, M. Epigallocatechin-3-gallate ameliorates radiation-induced acute skin damage in breast cancer patients undergoing adjuvant radiotherapy. Oncotarget 2016, 7, 48607–48613. [Google Scholar] [CrossRef]
  116. Hsieh, T.C.; Wu, J.M. Suppression of cell proliferation and gene expression by combinatorial synergy of EGCG, resveratrol and γ-tocotrienol in estrogen receptor-positive MCF-7 breast cancer cells. Int. J. Oncol. 2008, 33, 851–859. [Google Scholar] [CrossRef]
  117. Eddy, S.F.; Kane, S.E.; Sonenshein, G.E. Trastuzumab-resistant HER2-driven breast cancer cells are sensitive to epigallocatechin-3 gallate. Cancer Res. 2007, 67, 9018–9023. [Google Scholar] [CrossRef]
  118. Wang, S.; Chen, R.; Zhong, Z.; Shi, Z.; Chen, M.; Wang, Y. Epigallocatechin-3-gallate potentiates the effect of curcumin in inducing growth inhibition and apoptosis of resistant breast cancer cells. Am. J. Chin. Med. 2014, 42, 1279–1300. [Google Scholar] [CrossRef]
  119. Zeng, L.; Holly, J.M.P.; Perks, C.M. Effects of physiological levels of the green tea extract epigallocatechin-3-gallate on breast cancer cells. Front. Endocrinol. 2014, 5, 61. [Google Scholar] [CrossRef]
  120. Scandlyn, M.J.; Stuart, E.C.; Somers-Edgar, T.J.; Menzies, A.R.; Rosengren, R.J. A new role for tamoxifen in oestrogen receptor-negative breast cancer when it is combined with epigallocatechin gallate. Br. J. Cancer 2008, 99, 1056–1063. [Google Scholar] [CrossRef]
  121. Chisholm, K.; Bray, B.J.; Rosengren, R.J. Tamoxifen and epigallocatechin gallate are synergistically cytotoxic to MDA-MB-231 human breast cancer cells. Anticancer Drugs 2004, 15, 889–897. [Google Scholar] [CrossRef] [PubMed]
  122. Steed, K.L.; Jordan, H.R.; Tollefsbol, T.O. SAHA and EGCG promote apoptosis in triple-negative breast cancer cells, possibly through the modulation of CIAP2. Anticancer Res. 2020, 40, 9–26. [Google Scholar] [CrossRef] [PubMed]
  123. Lewis, K.A.; Jordan, H.R.; Tollefsbol, T.O. Effects of SAHA and EGCG on growth potentiation of triple-negative breast cancer cells. Cancers 2018, 11, 23. [Google Scholar] [CrossRef]
  124. Somers-Edgar, T.J.; Scandlyn, M.J.; Stuart, E.C.; Le Nedelec, M.J.; Valentine, S.P.; Rosengren, R.J. The combination of epigallocatechin gallate and curcumin suppresses ERα-breast cancer cell growth in vitro and in vivo. Int. J. Cancer 2008, 122, 1966–1971. [Google Scholar] [CrossRef]
  125. Bimonte, S.; Cascella, M.; Barbieri, A.; Arra, C.; Cuomo, A. Shining a light on the effects of the combination of (–)-epigallocatechin-3-gallate and tapentadol on the growth of human triple-negative breast cancer cells. Vivo 2019, 33, 1463–1468. [Google Scholar] [CrossRef]
  126. Luo, T.; Wang, J.; Yin, Y.; Hua, H.; Jing, J.; Sun, X.; Li, M.; Zhang, Y.; Jiang, Y. (−)-Epigallocatechin gallate sensitizes breast cancer cells to paclitaxel in a murine model of breast carcinoma. Breast Cancer Res. 2010, 12, R8. [Google Scholar] [CrossRef]
  127. Vieira, I.R.S.; Tessaro, L.; Lima, A.K.O.; Velloso, I.P.S.; Conte-Junior, C.A. Recent progress in nanotechnology improving the therapeutic potential of polyphenols for cancer. Nutrients 2023, 15, 3136. [Google Scholar] [CrossRef]
  128. Zeng, L.; Yan, J.; Luo, L.; Ma, M.; Zhu, H. Preparation and characterization of (−)-epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and their inhibitory effects on human breast cancer MCF-7 cells. Sci. Rep. 2017, 7, 45521. [Google Scholar] [CrossRef]
  129. Das, A.; Haque, I.; Ray, P.; Ghosh, A.; Dutta, D.; Quadir, M.; De, A.; Gunewardena, S.; Chatterjee, I.; Banerjee, S.; et al. CCN5 activation by free or encapsulated EGCG is required to render triple-negative breast cancer cell viability and tumor progression. Pharmacol. Res. Perspect. 2021, 9, e00753. [Google Scholar] [CrossRef]
  130. Wang, X.; He, J.; Jiang, S.; Gao, Y.; Zhang, L.K.; Yin, L.; You, R.; Guan, Y.Q. Multi-ligand modified PC@DOX-PA/EGCG micelles effectively inhibit the growth of ER+, PR+ or HER2+ breast cancer. J. Mater. Chem. B 2022, 10, 418–429. [Google Scholar] [CrossRef]
  131. Ramadass, S.K.; Anantharaman, N.V.; Subramanian, S.; Sivasubramanian, S.; Madhan, B. Paclitaxel/epigallocatechin gallate coloaded liposome: A synergistic delivery to control the invasiveness of MDA-MB-231 breast cancer cells. Colloids Surf. B Biointerfaces 2015, 125, 65–72. [Google Scholar] [CrossRef] [PubMed]
  132. Narayanan, S.; Mony, U.; Vijaykumar, D.K.; Koyakutty, M.; Paul-Prasanth, B.; Menon, D. Sequential release of epigallocatechin gallate and paclitaxel from PLGA-casein core/shell nanoparticles sensitizes drug-resistant breast cancer cells. Nanomedicine 2015, 11, 1399–1406. [Google Scholar] [CrossRef] [PubMed]
  133. de Pace, R.C.C.; Liu, X.; Sun, M.; Nie, S.; Zhang, J.; Cai, Q.; Gao, W.; Pan, X.; Fan, Z.; Wang, S. Anticancer activities of (−)-epigallocatechin-3-gallate encapsulated nanoliposomes in MCF7 breast cancer cells. J. Liposome Res. 2013, 23, 187–196. [Google Scholar] [CrossRef] [PubMed]
  134. Radhakrishnan, R.; Kulhari, H.; Pooja, D.; Gudem, S.; Bhargava, S.; Shukla, R.; Sistla, R. Encapsulation of biophenolic phytochemical EGCG within lipid nanoparticles enhances its stability and cytotoxicity against cancer. Chem. Phys. Lipids 2016, 198, 51–60. [Google Scholar] [CrossRef] [PubMed]
  135. Radhakrishnan, R.; Pooja, D.; Kulhari, H.; Gudem, S.; Ravuri, H.G.; Bhargava, S.; Ramakrishna, S. Bombesin conjugated solid lipid nanoparticles for improved delivery of epigallocatechin gallate for breast cancer treatment. Chem. Phys. Lipids 2019, 224, 104770. [Google Scholar] [CrossRef]
  136. Wang, Y.Q.; Lu, J.L.; Liang, Y.R.; Li, Q.S. Suppressive effects of EGCG on cervical cancer. Molecules 2018, 23, 2334. [Google Scholar] [CrossRef]
  137. Aarthy, M.; Panwar, U.; Singh, S.K. Structural dynamic studies on identification of EGCG analogues for the inhibition of human papillomavirus E7. Sci. Rep. 2020, 10, 8661. [Google Scholar] [CrossRef]
  138. Grandi, G.; Botticelli, L.; Fraia, P.D.; Babalini, C.; Masini, M.; Unfer, V. The association of four natural molecules—EGCG, folic acid, vitamin B12, and HA—To counteract HPV cervical lesions: A case report. J. Pers. Med. 2023, 13, 567. [Google Scholar] [CrossRef]
  139. Calcagno, M.; Incocciati, B.; Di Fraia, L.; Unfer, V. Counteracting HPV cervical and anal infection through dietary supplementation of EGCG, folic acid, vitamin B12 and hyaluronic acid: Clinical case reports. J. Clin. Med. 2024, 13, 3597. [Google Scholar] [CrossRef]
  140. Miyoshi, N.; Tanabe, H.; Suzuki, T.; Saeki, K.; Hara, Y. Applications of a standardized green tea catechin preparation for viral warts and human papilloma virus-related and unrelated cancers. Molecules 2020, 25, 2588. [Google Scholar] [CrossRef]
  141. Chu, Y.W.; Liu, S.T.; Yang, Y.L.; Huang, S.M.; Wang, W.M. The cytotoxic mechanism of epigallocatechin gallate on proliferative HaCaT keratinocytes. J. Biomed. Sci. 2017, 24, 55. [Google Scholar] [CrossRef] [PubMed]
  142. Yap, J.K.W.; Kehoe, S.T.; Woodman, C.B.J.; Dawson, C.W. The major constituent of green tea, epigallocatechin-3-gallate (EGCG), inhibits the growth of HPV18-infected keratinocytes by stimulating proteasomal turnover of the E6 and E7 oncoproteins. Pathogens 2021, 10, 459. [Google Scholar] [CrossRef] [PubMed]
  143. Muthusami, S.; Prabakaran, D.S.; An, Z.; Yu, J.R.; Park, W.Y. EGCG suppresses Fused Toes Homolog protein through p53 in cervical cancer cells. Mol. Biol. Rep. 2013, 40, 5587–5596. [Google Scholar] [CrossRef]
  144. Asif Siddiqui, F.; Naim, M.; Islam, N. Apoptotic effect of green tea polyphenol (EGCG) on cervical carcinoma cells. Diagn. Cytopathol. 2011, 39, 482–488. [Google Scholar] [CrossRef]
  145. Chakrabarty, S.; Nag, D.; Ganguli, A.; Das, A.; Ghosh Dastidar, D.; Chakrabarti, G. Theaflavin and epigallocatechin-3-gallate synergistically induce apoptosis through inhibition of PI3K/Akt signaling upon depolymerizing microtubules in HeLa cells. J. Cell. Biochem. 2019, 120, 5987–6003. [Google Scholar] [CrossRef]
  146. Pal, D.; Sur, S.; Roy, R.; Mandal, S.; Kumar Panda, C. Epigallocatechin gallate in combination with eugenol or amarogentin shows synergistic chemotherapeutic potential in cervical cancer cell line. J. Cell. Physiol. 2019, 234, 825–836. [Google Scholar] [CrossRef]
  147. McDonnell, A.M.; Pyles, H.M.; Diaz-Cruz, E.S.; Barton, C.E. Enoxacin and epigallocatechin gallate (EGCG) act synergistically to inhibit the growth of cervical cancer cells in culture. Molecules 2019, 24, 1580. [Google Scholar] [CrossRef]
  148. Kilic, U.; Sahin, K.; Tuzcu, M.; Basak, N.; Orhan, C.; Elibol-Can, B.; Kilic, E.; Sahin, F.; Kucuk, O. Enhancement of cisplatin sensitivity in human cervical cancer: Epigallocatechin-3-gallate. Front. Nutr. 2015, 1, 28. [Google Scholar] [CrossRef]
  149. Yokoyama, M.; Noguchi, M.; Nakao, Y.; Ysunaga, M.; Yamasaki, F.; Iwasaka, T. Antiproliferative effects of the major tea polyphenol, (−)-epigallocatechin gallate and retinoic acid in cervical adenocarcinoma. Gynecol. Oncol. 2008, 108, 326–331. [Google Scholar] [CrossRef]
  150. Raish, M.; Husain, S.Z.A.; Bae, S.M.; Han, S.J.; Park, C.H.; Shin, J.C.; Ahn, W.S. Photodynamic therapy in combination with green tea polyphenol EGCG enhances antitumor efficacy in human papillomavirus 16 (E6/E7) immortalized tumor cells. J. Appl. Res. 2010, 10, 58–67. [Google Scholar]
  151. Oladimeji, O.; Akinyelu, J.; Singh, M. Co-polymer functionalised gold nanoparticles show efficient mitochondrial targeted drug delivery in cervical carcinoma cells. J. Biomed. Nanotechnol. 2020, 16, 853–866. [Google Scholar] [CrossRef] [PubMed]
  152. Włodarczyk, M.; Ciebiera, M.; Nowicka, G.; Łoziński, T.; Ali, M.; Al-Hendy, A. Epigallocatechin gallate for the treatment of benign and malignant gynecological diseases—Focus on epigenetic mechanisms. Nutrients 2024, 16, 559. [Google Scholar] [CrossRef] [PubMed]
  153. Butler, L.M.; Wu, A.H. Green and black tea in relation to gynecologic cancers. Mol. Nutr. Food Res. 2011, 55, 931–940. [Google Scholar] [CrossRef] [PubMed]
  154. Aussem, A.; Ludwig, K. The potential for reducing Lynch syndrome cancer risk with nutritional Nrf2 activators. Nutr. Cancer 2021, 73, 404–419. [Google Scholar] [CrossRef]
  155. Man, G.C.W.; Wang, J.; Song, Y.; Wong, J.H.; Zhao, Y.; Lau, T.S.; Leung, K.T.; Chan, T.H.; Wang, H.; Kwong, J.; et al. Therapeutic potential of a novel prodrug of green tea extract in induction of apoptosis via ERK/JNK and Akt signaling pathway in human endometrial cancer. BMC Cancer 2020, 20, 964. [Google Scholar] [CrossRef]
  156. Wang, J.; Man, G.C.W.; Chan, T.H.; Kwong, J.; Wang, C.C. A prodrug of green tea polyphenol (–)-epigallocatechin-3-gallate (Pro-EGCG) serves as a novel angiogenesis inhibitor in endometrial cancer. Cancer Lett. 2018, 412, 10–20. [Google Scholar] [CrossRef]
  157. Wang, C.C.; Xu, H.; Man, G.C.W.; Zhang, T.; Chu, K.O.; Chu, C.Y.; Cheng, J.T.Y.; Li, G.; He, Y.X.; Qin, L.; et al. Prodrug of green tea epigallocatechin-3-gallate (Pro-EGCG) as a potent anti-angiogenesis agent for endometriosis in mice. Angiogenesis 2013, 16, 59–69. [Google Scholar] [CrossRef]
  158. Bimonte, S.; Cascella, M. The potential roles of epigallocatechin-3-gallate in the treatment of ovarian cancer: Current state of knowledge. Drug Des. Dev. Ther. 2020, 14, 4245–4250. [Google Scholar] [CrossRef]
  159. Rodriguez Torres, S.; Gresseau, L.; Benhamida, M.; Fernandez-Marrero, Y.; Annabi, B. Epigallocatechin-3-gallate prevents the acquisition of a cancer stem cell phenotype in ovarian cancer tumorspheres through the inhibition of Src/JAK/STAT3 signaling. Biomedicines 2023, 11, 1000. [Google Scholar] [CrossRef]
  160. Trudel, D.; Labbé, D.P.; Araya-Farias, M.; Doyen, A.; Bazinet, L.; Duchesne, T.; Plante, M.; Grégoire, J.; Renaud, M.C.; Bachvarov, D.; et al. A two-stage, single-arm, phase II study of EGCG-enriched green tea drink as a maintenance therapy in women with advanced stage ovarian cancer. Gynecol. Oncol. 2013, 131, 357–361. [Google Scholar] [CrossRef]
  161. Kiselev, V.I.; Ashrafyan, L.A.; Muyzhnek, E.L.; Gerfanova, E.V.; Antonova, I.B.; Aleshikova, O.I.; Sarkar, F.H. A new promising way of maintenance therapy in advanced ovarian cancer: A comparative clinical study. BMC Cancer 2018, 18, 904. [Google Scholar] [CrossRef] [PubMed]
  162. Chan, M.M.; Soprano, K.J.; Weinstein, K.; Fong, D. Epigallocatechin-3-gallate delivers hydrogen peroxide to induce death of ovarian cancer cells and enhances their cisplatin susceptibility. J. Cell. Physiol. 2006, 207, 389–396. [Google Scholar] [CrossRef] [PubMed]
  163. Yan, C.; Yang, J.; Shen, L.; Chen, X. Inhibitory effect of epigallocatechin gallate on ovarian cancer cell proliferation associated with aquaporin 5 expression. Arch. Gynecol. Obstet. 2012, 285, 459–467. [Google Scholar] [CrossRef]
  164. Chen, H.; Landen, C.N.; Li, Y.; Alvarez, R.D.; Tollefsbol, T.O. Epigallocatechin gallate and sulforaphane combination treatment induce apoptosis in paclitaxel-resistant ovarian cancer cells through hTERT and Bcl-2 down-regulation. Exp. Cell Res. 2013, 319, 697–706. [Google Scholar] [CrossRef]
  165. Chen, H.; Landen, C.N.; Li, Y.; Alvarez, R.D.; Tollefsbol, T.O. Enhancement of cisplatin-mediated apoptosis in ovarian cancer cells through potentiating G2/M arrest and P21 upregulation by combinatorial epigallocatechin gallate and sulforaphane. J. Oncol. 2013, 2013, 872957. [Google Scholar] [CrossRef]
  166. Wang, X.; Jiang, P.; Wang, P.; Yang, C.S.; Wang, X.; Feng, Q. EGCG enhances cisplatin sensitivity by regulating expression of the copper and cisplatin influx transporter CTR1 in ovary cancer. PLoS ONE 2015, 10, e0125402. [Google Scholar] [CrossRef]
  167. Mazumder, M.E.H.; Beale, P.; Chan, C.; Yu, J.Q.; Huq, F. Epigallocatechin gallate acts synergistically in combination with cisplatin and designed trans-palladiums in ovarian cancer cells. Anticancer Res. 2012, 32, 4851–4860. [Google Scholar]
  168. Yunos, N.M.; Beale, P.; Yu, J.Q.; Huq, F. Synergism from sequenced combinations of curcumin and epigallocatechin-3-gallate with cisplatin in the killing of human ovarian cancer cells. Anticancer Res. 2011, 31, 1131–1140. [Google Scholar]
  169. Sharma, T.; Singh, D.; Mahapatra, A.; Mahapatra, P.; Sahoo, S.; Sahoo, S.K. Advancements in clinical translation of flavonoid nanoparticles for cancer treatment. OpenNano 2022, 8, 100074. [Google Scholar] [CrossRef]
  170. Khan, H.; Ullah, H.; Martorell, M.; Valdes, S.E.; Belwal, T.; Tejada, S.; Sureda, A.; Kamal, M.A. Flavonoids nanoparticles in cancer: Treatment, prevention and clinical prospects. Semin. Cancer Biol. 2021, 69, 200–211. [Google Scholar] [CrossRef]
  171. Dobrzynska, M.; Napierala, M.; Florek, E. Flavonoid nanoparticles: A promising approach for cancer therapy. Biomolecules 2020, 10, 1268. [Google Scholar] [CrossRef] [PubMed]
  172. Sun, W.; Yang, Y.; Wang, C.; Liu, M.; Wang, J.; Qiao, S.; Jiang, P.; Sun, C.; Jiang, S. Epigallocatechin-3-gallate at the nanoscale: A new strategy for cancer treatment. Pharm. Biol. 2024, 62, 676–690. [Google Scholar] [CrossRef] [PubMed]
  173. Jiang, Y.; Jiang, Z.; Ma, L.; Huang, Q. Advances in nanodelivery of green tea catechins to enhance the anticancer activity. Molecules 2021, 26, 3301. [Google Scholar] [CrossRef] [PubMed]
  174. Granja, A.; Pinheiro, M.; Reis, S. Epigallocatechin gallate nanodelivery systems for cancer therapy. Nutrients 2016, 8, 307. [Google Scholar] [CrossRef]
  175. Alizadeh, L.; Alizadeh, E.; Zarebkohan, A.; Ahmadi, E.; Rahmati-Yamchi, M.; Salehi, R. AS1411 aptamer-functionalized chitosan-silica nanoparticles for targeted delivery of epigallocatechin gallate to the SKOV-3 ovarian cancer cell lines. J. Nanopart. Res. 2020, 22, 5. [Google Scholar] [CrossRef]
  176. Chuan, D.; Mu, M.; Hou, H.; Zhao, N.; Li, J.; Tong, A.; Zou, B.; Chen, H.; Han, B.; Guo, G. Folic acid-functionalized tea polyphenol as a tumor-targeting nanodrug delivery system. Mater. Des. 2021, 206, 109805. [Google Scholar] [CrossRef]
  177. Bae, K.H.; Tan, S.; Yamashita, A.; Ang, W.X.; Gao, S.J.; Wang, S.; Chung, J.E.; Kurisawa, M. Hyaluronic acid-green tea catechin micellar nanocomplexes: Fail-safe cisplatin nanomedicine for the treatment of ovarian cancer without off-target toxicity. Biomaterials 2017, 148, 41–53. [Google Scholar] [CrossRef]
Figure 1. The structure of the main components of green tea from the group of natural polyphenols and their content in tea leaves (% of the dry mass of tea leaves, based on [7]).
Figure 1. The structure of the main components of green tea from the group of natural polyphenols and their content in tea leaves (% of the dry mass of tea leaves, based on [7]).
Nutrients 17 00212 g001
Figure 2. Proposed main metabolic pathways of EGCG degradation by gut microbiota (based on [32,33]), where EGCG = (–)-epigallocatechin gallate, EGC = (–)-epigallocatechin.
Figure 2. Proposed main metabolic pathways of EGCG degradation by gut microbiota (based on [32,33]), where EGCG = (–)-epigallocatechin gallate, EGC = (–)-epigallocatechin.
Nutrients 17 00212 g002
Figure 3. The structure of theaflavin, an important polyphenol found in black tea.
Figure 3. The structure of theaflavin, an important polyphenol found in black tea.
Nutrients 17 00212 g003
Table 1. Selected in vitro and in vivo studies on the anticancer potential of EGCG.
Table 1. Selected in vitro and in vivo studies on the anticancer potential of EGCG.
Cancer TypeCancer Cell LinesIn Vitro EGCG
Concentration
In Vivo TestsAnticancer EffectsRef.
Breast cancerMCF-72, 4, 8, 16, 32, 64, 128 μM Inhibition of proliferation, promotion of apoptosis[56]
MCF-710–100 μg/mL Inhibition of reproductive potential and colony formation, reduction in cell viability, promotion of apoptosis[57]
T47D10–80 μM Reduction in cell viability, modulation of gene expression[58]
MCF-7,
MDA-MB 231
20 μM Inhibition of the progression of cancer[59]
MCF-70.5–20 μg/mLCB-17 SCID mice (6–8 weeks old); EGCG dose: 100 mg/kg, oral gavageInhibition of cell viability, induction of apoptosis, disruption of the cell cycle in the G2/M phase, inhibition of tumor growth[60]
MCF-720 μM Reduction in MMP-2 activity[61]
4T110–320 μMBALB/c mice (4 weeks old); EGCG dose: 5, 10, 20 mg/kg, intraperitoneal injectionsReduction in cell growth, induction of apoptosis, promotion of mitochondrial depolarization, inhibition of glucose metabolism, reduction in tumor mass[62]
E0771, MCF-7, MDA-MB-23110, 50 μg/mLC57BL/6 mice (7 weeks old); EGCG dose: 50–100 mg/kg, drinking waterImpact on cancer cells and tumor blood vessels, reduction in tumor mass[63]
MCF-725, 50, 100 mg/L Reduction in cell proliferation and growth[64]
SUM-149, SUM-1905, 10, 20, 40, 60, 80, 160 µg/mLNOD/SCID mice (6 weeks old); EGCG dose: 16.5 mg/kg, intraperitoneal injectionsReduction in cell growth, invasiveness, and survival, reduction in the growth of existing tumors[65]
MDA-MB-2311–200 μg/mLNCr-nu/nu mice (5 weeks old); EGCG dose: 1 mg in 100 μL of autoclaved distilled waterInhibition of cell invasiveness, induction of apoptosis, delay in the occurrence of cancer, reduction in tumor mass[66]
Hs578T10 μM Inhibition of proliferation, migration, and invasiveness[67]
MDA-MB-2310.5 μg/mL Inhibition of invasion[68]
MDA-MB-23150, 80 μg/mL Inhibition of invasiveness, induction of apoptosis[69]
MDA-MB-2310.07–20 μM Inhibition of Met signaling, blocking invasiveness[70]
Cervical
cancer
C33A, CaSki, HeLa, SiHa10, 20, 40, 60, 80, 100 µg/mL Inhibition of cell growth, probably due to regulation of miRNA expression[71]
HeLa10 μM Inhibition of proliferation, adhesion, spread and invasiveness[72]
HeLa, SiHa20, 40, 60, 80, 100 μM Inhibition of cell viability[73]
CaSki, HeLa10, 25, 50, 100 µM Cell cycle arrest, apoptosis induction[74]
HeLa50 µM Modulation of epigenetic factors and signaling pathways (MAPK, PI3K, Wnt)[75]
HeLa1–100 μM Inhibition of cell growth, invasion, and migration[76]
OMC-4, TMCC-150–100 μM Induction of apoptosis, inhibition of telomerase activity, dysregulation of the cell cycle[77]
HeLa, ME180, SiHa, TMCC-150, 100 μM Induction of apoptosis, inhibition of telomerase activity[78]
C33A, ME18050 μM Reduction in matrix abundance and cell migration[79]
Endometrial
cancer
Ishikawa50, 75, 100, 125, 150 μM Inhibition of cell proliferation, induction of apoptosis, modulation of pro- and anti-apoptotic proteins, generation of ROS[80]
Ishikawa, RL952100 μM Reduction in VEGF[81]
Ovarian
cancer
A2780/DDP, SKOV3/DDP0–40 μMBALB/c mice (5 weeks old); EGCG dose: 50 mg/kg, intraperitoneal injectionsInhibition of cell proliferation and motility, induction of apoptosis, inhibition of tumor growth[82]
OVCAR-3,
PA-1, SKOV-3
10–100 μM Inhibition of cell growth, cell cycle arrest, induction of apoptosis[83]
OVCAR-3,
PA-1, SKOV-3
10–100 μM Inhibition of cell growth, cell cycle arrest, induction of apoptosis[84]
OVCAR-30–200 μM Inhibition of cell proliferation and migration[85]
CAOV-3, OVCAR-3, SKOV35, 10, 20, 40, 80 µg/mLBALB/c mice (4–5 weeks old); EGCG dose: 10, 30, 50 mg/kgInhibition of cell proliferation and induction of apoptosis; increased inhibition of tumor growth compared to paclitaxel (50 mg/kg EGCG vs. 5 mg/kg paclitaxel)[86]
SKOV-320, 30, 40, 50 µg/mL Inhibition of cell proliferation and viability, DNA damage, induction of apoptosis[87]
A2780, SKOV30–20 nMBALB/c mice; EGCG dose: 200 mg/kg, intratumoral injectionInhibition of cell migration and survival, acceleration of apoptosis, inhibition of tumor growth[88]
HEY, OVCA 43310, 20, 40 μMAthymic (nu+/nu+) mice (4–6 weeks old); EGCG source: green tea leaves (12.4 g/L), drinking fluidInhibition of cell growth, induction of apoptosis, inhibition of angiogenesis and invasiveness; reduction in tumor growth[89]
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

Markowska, A.; Antoszczak, M.; Markowska, J.; Huczyński, A. Role of Epigallocatechin Gallate in Selected Malignant Neoplasms in Women. Nutrients 2025, 17, 212. https://doi.org/10.3390/nu17020212

AMA Style

Markowska A, Antoszczak M, Markowska J, Huczyński A. Role of Epigallocatechin Gallate in Selected Malignant Neoplasms in Women. Nutrients. 2025; 17(2):212. https://doi.org/10.3390/nu17020212

Chicago/Turabian Style

Markowska, Anna, Michał Antoszczak, Janina Markowska, and Adam Huczyński. 2025. "Role of Epigallocatechin Gallate in Selected Malignant Neoplasms in Women" Nutrients 17, no. 2: 212. https://doi.org/10.3390/nu17020212

APA Style

Markowska, A., Antoszczak, M., Markowska, J., & Huczyński, A. (2025). Role of Epigallocatechin Gallate in Selected Malignant Neoplasms in Women. Nutrients, 17(2), 212. https://doi.org/10.3390/nu17020212

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