Cancer Preventive and Therapeutic Effects of EGCG The Major Polyphenol in Green Tea
Cancer Preventive and Therapeutic Effects of EGCG The Major Polyphenol in Green Tea
Islam Rady, Hadir Mohamed, Mohamad Rady, Imtiaz A. Siddiqui & Hasan
Mukhtar
To cite this article: Islam Rady, Hadir Mohamed, Mohamad Rady, Imtiaz A. Siddiqui & Hasan
Mukhtar (2018) Cancer preventive and therapeutic effects of EGCG, the major polyphenol in green
tea, Egyptian Journal of Basic and Applied Sciences, 5:1, 1-23, DOI: 10.1016/j.ejbas.2017.12.001
© Mansoura University
Review Article
a r t i c l e i n f o a b s t r a c t
Article history: (-)-epigallocatechin-3-gallate (EGCG), the major bioactive catechin in green tea (GT) has been studied for
Received 14 November 2017 almost past thirty years as an agent initially for its cancer chemoprevention effects and then for its cancer
Accepted 6 December 2017 chemotherapeutic ability. This agent has shown considerable anti-cancer effects in a variety of preclinical
Available online 19 December 2017
cell culture and animal model systems. However, its clinical application to human patients is hampered
by a variety of reasons that includes its stability and bioavailability. As a result, an increased number of
Keywords: studies assessing the effects derived from the use of EGCG are been employed in combination with other
Green tea
agents or by utilizing innovative carrier settings. Here, we summarize the current understanding of the
EGCG
Anti-cancer effects
anticancer effects of EGCG and its effects with other combinations on different kinds of cancers.
Cancer nanochemoprevention Further, we also present the available information for the possible mechanism of action of EGCG.
Ó 2017 Mansoura University. Production and hosting by Elsevier B.V. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Contents
Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
EGCG anticancer effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Induction of apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Modulation of cellular proliferation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Inhibition of angiogenesis and related mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Anticancer effects of EGCG combinations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Induction of apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Modulation of cellular proliferation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Inhibition of angiogenesis and related mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Anticancer effects of EGCG combined with nanoparticls . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
https://doi.org/10.1016/j.ejbas.2017.12.001
2314-808X/Ó 2017 Mansoura University. Production and hosting by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
2 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
be caused by external factors such as infectious organisms, envi- Tea catechins were first isolated by Michiyo Tsujimura in 1929
ronmental pollutants, tobacco and an unhealthy diet or internal in Japan [16] and since then four main types of catechins have been
factors such as hormones, inherited genetic mutations and found in green tea leaves (Table 1): (-)-epigallocatechin-3-gallate
immune conditions may act together or singular to cause the incip- (EGCG) accounts for approximately 59% of the total catechins from
ience of cancer [7]. Since cancer is associated with such high mor- the leaves of the green tea, (-)-epigallocatechin (EGC) (19%), (-)-
bidity and mortality worldwide, there is an urgent need to epicatechin-3-gallate (ECG) (13.6%), and (-)-epicatechin (EC)
determine ways of management of this ailment where the current (6.4%) [17,18]. The functional and structural differences between
treatment modalities are mainly surgery, radiation based therapy, these catechins are attributed to the number of hydroxyl groups
chemotherapy, gene therapy and/or hormonal therapy [2]. Natural on the B-ring (Fig. 2) and the presence or absence of a galloyl moi-
products, especially those derived from plants, have been used to ety [18].
help mankind sustain its health since the dawn of medicine [8]. Among these catechins, EGCG is the most studied and is consid-
Nowadays, just like in ancient times, natural compounds are still ered to play a crucial role in cancer-preventive and therapeutic
determining factors in remedies [9]. activities [19–23]. Several studies have been performed to examine
Herbal medicine or Herbalism (also known as phytotherapy) is the effects of EGCG on various in vitro cancer-related molecular
the study of botany and use of plants intended for medicinal pur- targets and in vivo models for potential cancer chemoprevention
poses or for supplementing a diet, has been applied for thousands and therapy [24]. The overwhelming majority of these studies
of years, but researchers started to study their mode of action at observed that EGCG inhibits a vast array of anticancer molecular
the molecular, cellular and tissue levels only recently [10–12]. It targets (Fig. 3) and cancer-related cellular processes [25].
is also a firm belief that naturally occurring plant based natural Despite accomplished outcomes in preclinical settings, its appli-
compounds when properly formulated and administered have a cability to humans has met with limited success for many reasons
key role in cancer management. including inefficient systemic delivery and bioavailability. Several
Chemoprevention, especially through the use of naturally optimization approaches including utilization of nanoparticle
occurring phytochemicals capable of impeding the process of car- based delivery of EGCG have been utilized to circumvent the issues,
cinogenesis at one or more steps, is an ideal approach for cancer for instance, we in a seminal study [26] introduced the novel con-
management [13]. Among natural compounds, ever since our ini- cept of ‘‘nanochemoprevention” that utilizes nanotechnology for
tial work in describing its potential benefit against cancer, green enhancing the outcome of EGCG in cancer chemoprevention.
tea has been extensively studied worldwide in a variety of cancer Combination therapy or polytherapy (versus monotherapy) is
models and the resulting data has been very promising. Green therapy that consumes more than one medication. There has been
tea leaves comprises of a diverse number of components (Fig. 1) some emphasis on determining the effects of combining EGCG
that are demonstrated to be beneficial to human health. The green with other dietary agents. Several studies have indicated that
tea polyphenols are flavonols, commonly known as catechins [14] anti-cancer efficacy and scope of action of the individual agents
which are found in greater amounts in green tea than in black or can be further enhanced by combining them synergistically
Oolong tea [15]. with chemically similar or different compounds (Fig. 4). Such a
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 3
Table 1
Dried green tea composition.
combination might be effective in reducing the drug dosage and EGCG anticancer effects
resistance, and simultaneously exhibiting higher therapeutic out-
come [27–29]. Studies suggest that EGCG can synergistically inhi- Induction of apoptosis
bit cancer cells in vitro and in vivo when combined with other
dietary agents (Table 2), such as [6]-gingerol [30], curcumin [31], Apoptosis is a genetically encoded program leading to cell death
lovastatin [32], quercetin [33], sulforaphane [34], panaxadiol that is involved in normal development and homeostasis through-
[35], and pterostilbene [36]. Some evidence is also available for out the animal kingdom [50]. It is the main event that is known to
combination with chemotherapeutic agents such as 5- regulate the occurrence and/or spread of cancer [2]. The morpho-
fluorouracil [37], capecitabine [38], cisplatin [39], docetaxel [40], logical characteristicsof apoptosis include cell shrinkage, nuclear
doxorubicin [41] and temozolomide [42], or other agents like fragmentation, chromatin condensation and membrane blebbing
sodium butyrate [43], vitamin C and amino acids [44]. Combina- [50–52]. Apoptosis can undertake one or two pathways, intrinsic
tion of EGCG with these molecules can synergistically inhibit can- and/or extrinsic pathway (s) [53]. Intrinsic pathway, also known
cer cell proliferation [45,46], induce apoptosis [47,48] and suppress as mitochondrial pathway, can be induced through intracellular
tumor angiogenesis and growth [40] to name a few pathways that stresses via DNA damage or oxidative stress leading to release of
are effected by such an amalgamation. This synergistic effect, on mitochondrial Cytochrome C to form apoptosome complex [54].
one hand, may be associated with enhanced bioavailability of This complex is composed of Cytochrome C, apoptotic protease
EGCG [49]. Studies find that natural small molecules, such as quer- activating factor 1 (Apaf-1) and procaspase-9 [55], which activates
cetin can increase the bioavailability of EGCG in vitro and in vivo Caspase-9, Caspase-3 and Caspase-7 [56]. Otherwise, extrinsic
[33]. Current literature summarize the current understanding of pathway or death receptor pathway can be induced by death
the anti-cancer effects of EGCG alone and in combination with ligands Fas ligand (FasL), tumor necrosis factor a (TNFa) and
other dietary and pharmaceutical agents. TNF-related apoptosis inducing ligand (TRAIL) [57]. These ligands
4 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
Apaf-1 and BAD in NCI-H295 adrenal cancer cells [76] and BAK,
BAX, BCL-XS and PUMA in PANC-1, MIA-Pa-Ca-2, Hs 766T and
AsPC-1 pancreatic cancer cells [48]. Moreover, EGCG induced apop-
tosis through both intrinsic and extrinsic pathways, regulatory
proteins and endoplasmic reticulum stress via activation of
caspase-dependent, caspase-independent, death receptors, down-
regulation of anti-apoptotic proteins BCL-2, BCL-XL and XIAP and
upregulation of pro-apoptotic BAD and BAX in NCI-H295 human
adrenal cancer cells [76].
pEGCG is synthesized by modifying reactive hydroxyl groups
with peracetate groups and found to be converted as well as accu-
mulated into parental EGCG when cultured with MDA-MB-231
human breast cancer cell. pEGCG was found to inhibit significantly
tumor growth in vivo through apoptosis induction in tumor tissues
[79].
Fig. 3. Schematic drawing of the regulative actions of EGCG. This carton is based on the available literature about the anticancer effects of EGCG.
[96–98]. Furthermore, cancer cell motility, migration and invasion cells, EGCG inhibited invasion, epithelial-mesenchymal transition,
play fundamental roles in cancer metastasis [99]. Therefore, and tumor growth through downregulation of MMP-2, uPA, p-
inhibiting either cancer cell motility, migration or invasion impede FAK, p-Src, snail-1 and vimentin [111]. EGCG inhibits HGF-
metastasis, which is the cause of over 90% of patient deaths [100]. induced cell growth and invasion through suppression of HGF/c-
EGCG has demonstrated potential efficacy in inhibiting angio- Met signaling pathway [112]. Repressing of functional invadopodia
genesis, necrosis, motility, invasion, migration and metastasis formation was done by EGCG to inhibit in vitro and in vivo invasion
markers in a variety of human cancers tested under preclinical in HSC-3 and YD-10B oral cancer cells FAK/Src signaling [113]. In
model systems. In A549 lung cancer cells, EGCG was observed to Hypopharyngeal FaDu and laryngeal SNU-899 and SNU-1066 can-
inhibit angiogenesis and reduce xenograft tumor growth through cer cells, EGCG Inhibited HGF-induced cell growth and invasion
inhibiting IGF-1 via suppressing HIF-1a and VEGF protein expres- through suppression of HGF/c-Met signaling pathway [112]. In gas-
sion [101–104], upregulation of endostatin expression [102], inhi- tric cancer, EGCG inhibited xenograft angiogenesis and tumor
bition of HPV-16 oncoprotein induced angiogenesis conferred by growth in BGC-823 [40]. EGCG inhibited IL-6-induced angiogenesis
cancer cells via inhibition of HIF-1a protein expression and HIF- in vitro and in vivo through inhibition of VEGF expression via sup-
1a dependent expression of VEGF, IL-8, and CD31 and activation pressing Stat3 activity in AGS cells [114]. In SGC-7901 cancer cells,
of Akt [104]. In addition, EGCG inhibits nicotine-induced migration, EGCG inhibited tumor growth and angiogenesis through reducing
invasion through upregulation of HIF-1a, VEGF, COX-2, p-Akt and VEGF-induced endothelial cell proliferation, migration and tube
p-ERK [105]. Similarly, in lung NCI-H460 cancer cells, EGCG Inhi- formation [115]. In squamous cell carcinoma, EGCG inhibited
bits angiogenesis through inhibition of HIF-1a protein expression HGF-induced cell growth and invasion through suppression of
and HIF-1a dependent expression of VEGF, IL-8, and CD31 as well HGF/c-Met signaling pathway in SCC VII/SF cells while it inhibited
as activation of Akt [104]. EGCG also was able to inhibit cell motil- xenograft tumor growth in vivo via rising apoptosis [112]. In hep-
ity in vitro wound healing assay in H1299 and Lu99 lung cancer atocellular carcinoma, in Hepa 1c1c7 cells, EGCG inhibited cell
cells [106]. In ovarian cancer, EGCG inhibited cell motility through motility via suppression of Hsp90 chaperone system [107]. In cer-
suppression of Hsp90 chaperone system in SKOV3 cells [107]. On vical cancer, EGCG inhibited cell proliferation, invasion and migra-
breast cancer, only 10 lM of EGCG was able to inhibit cell migra- tion in HeLa cells through downregulation of MMP-9 gene and
tion trough downregulation of VEGF expression in Hs578T breast upregulation of TIMP-1 gene [85]. In colorectal cancer, EGCG inhib-
[130]. In NF639 breast cancer cells, EGCG inhibited branching col- ited tumor growth in vitro in SW837 cells and in vivo and through
ony growth and cell invasion through induction of estrogen recep- activation of VEGF/VEGFR axis via suppressing the expression of
tor a expression via activating FOXO3a signaling [108]. EGCG HIF-1a and several major growth factors [116]. EGCG also inhib-
inhibits cell migration and invasion through downregulation of ited migration and proliferation in SW620 cells in vitro through
VASP expression and Rac1 activity [109] in MCF-7 cancer cells. inactivation of PAR2-AP and factor VIIa and by the way inhibition
EGCG also Inhibits cell invasion, motility and migration in MDA- of the ERK1/2 and NF-jB pathways [117]. Moreover, EGCG inhib-
MB-231 through inhibition of EGF-induced MMP-9 via suppressing ited inhibits liver metastasis in vivo RKO colorectal cancer cells
FAK and ERK signaling pathways [110]. In oral cancer, in SCC-9 experiments and suppresses angiogenesis and induces apoptosis
6 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
Fig. 4. Schematic drawing of the regulative actions of EGCG combined with other dietary and pharmaceutical agents. This carton is based on the available literature.
in liver metastasis [118]. EGCG inhibited Met signaling which helps have been employed to induce apoptosis in different cancers. The
to attenuate tumor spread/metastasis, independent of H2O2- general idea is that combination of two or more agents could target
related mechanisms in HCT-116 and HT-29 colorectal cancer cells more pathways and thus will be more effective to increase the sta-
[119,120]. In bladder cancer, EGCG inhibited cell adhesion, migra- bility of the agent and reduce toxicity to simultaneously exhibit
tion and invasion in T-24 cells through downregulation of MMP-9 higher therapeutic outcome. Various studies found that EGCG syn-
expression via blocking of NF-jB signaling pathway [121]. In eso- ergistically induced cancer cells apoptosis in vitro and in vivo
phageal TE-8 and SKGT-4 cancer cells, EGCG reduced cell viability through different apoptotic signaling, upregulation of pro-
and invasion in vitro through reduction of p-ERK1/2, c-Jun and apoptotic proteins and inhibition of anti-apoptotic proteins when
COX-2, and activation of caspase-3 whereas it inhibits tumor combined with other natural molecules, such as vitexin-2-O-
growth in vivo through suppressing the expression of Ki67, p- xyloside and raphasatin [46], curcumin [125], N-acetylcysteine
ERK1/2 and COX-2 [32]. In prostate cancer, EGCG inhibited cell (NAC) [126], pterostilbene [36], tumor necrosis factor-related
motility and invasion through inhibition of c-Met signaling via apoptosis-inducing ligand (TRAIL) [74], quercetin [33], whole
altering the structure or function of lipid rafts in DU-145 cells green tea polyphenols (GTPs) [127], eicosapentaenoic acid-free
[122]. In addition, EGCG Inhibited tumor growth and angiogenesis fatty acid (EPA-FFA) and grape seed [GS] extract [128], 5-
in CWR22Rv1 cells but promoting apoptosis of the prostate cancer fluorouracil (5-FU) [37], sodium butyrate (NaB) [43] and [6]-
cells in vivo [123]. In BCaPT10 cancer cells, EGCG inhibited cell Gingerol and tocotrienol-rich fraction (TRF) [30].
motility in vitro via suppression of Hsp90 molecular chaperone sys- Combination of EGCG with curcumin induces apoptosis through
tem which supports malignant phenotype [124]. EGCG-P is more upregulation of caspase-dependent apoptotic signaling pathways
stable and effective than EGCG enhancing the inhibition of the in MCF-7 breast cancer cells [125]. However, a mixture of EGCG,
tumor growth, angiogenesis of CWR22Rv1 prostate cancer cells vitexin-2-O-xyloside and raphasatin was found to induce apoptosis
in vivo [123]. via mitochondrial pathway in breast MDA-MB-231 and MCF-7 and
colorectal Caco-2 and LoVo cancer cells [46]. In addition, NAC and
EGCG interact to form EGCG-20 -NAC adduct which induces cell cul-
Anticancer effects of EGCG combinations ture apoptosis in CL-13 lung cancer cells [126]. Also, pterostilbene
and EGCG combination induced apoptosis in both PANC-1 and
Induction of apoptosis MIA-Pa-Ca-2 pancreatic cancer cells [36] Adding TRAIL to EGCG
increases synergistically apoptosis induction via cleavage of
Several studies have stated that EGCG and its combinations procaspase-3 in MIA-Pa-Ca-2 cells [74]. Studies observed that nat-
have induced apoptosis in a variety of cancers. EGCG as a green ural small molecules, such as quercetin, can increase the bioavail-
tea polyphenol (GTP) or combined with different natural molecules ability of EGCG in rats and human [129] enhancing apoptosis
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 7
Table 2
Compounds have previously combined with EGCG.
Table 2 (continued)
EGC
EC
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 9
Table 2 (continued)
ECG
EGCG
induced against cancer cells due to EGCG treatment, such as in Gingerol or EGCG and TRF combinations both can induce apoptosis
LNCaP and PC-3 prostate cancer cells [33]. In vivo GTPs (including in 1321N1, LN18 and SW1783 glioma cells through activation of
that contains EGCG) oral infusion resulted in significant apoptosis caspase-3 [30].
of cancer cells causing inhibition of prostate cancer development, On the other hand, cancer stem cells (CSCs) have been identified
progression, and metastasis [127]. A combination of EGCG, eicos- in a number of solid tumors, including breast cancer, brain tumors,
apentaenoic acid-free fatty acid and grape seed extract [128] or a lung cancer, colon cancer, and melanoma [130]. CSCs have the
mixture of EGCG and Fluorouracil (5-FU) [37] induced apoptosis capacity to self-renew, to give rise to progeny that are different
in both HCT-116 and SW480 colorectal cancer cells [37,128]. Also from them, and to utilize common signaling pathways [130,131].
apoptosis through survivin downregulation has been noted when CSCs may be the source of all the tumor cells present in a malig-
HCT-116, HT-29 and RKO colorectal cancer cells are treated with nant tumor, the reason for the resistance to the chemotherapeutic
a combination of EGCG and NaB [43]. Finally, both EGCG and [6]- agent used to treat the malignant tumor, and the source of cells
Table 3
10
Anticancer effects of EGCG alone and combination.
Cancers Cell lines EGCG Combination Dose & IC50 Biological effects
Breast cancer MDA-MB-231 EGCG 50 and 100 lM [149]; 50 and 80 lg/ Inhibits cell proliferation and viability through suppression of Wnt signaling via inducing the HBP1
mL [75]; 20, 40 and 60 lM [150]; 10 transcriptional repressor [149], epigenetic repression of hTERT [150] and inhibition of glucose
and 20 lM [110]; 0.01–1000 lM uptake and metabolism [151]. Induces cell apoptosis through stimulation of pro-apoptotic
[151]. signaling and downregulation of MMP-9 expression [75]. Inhibits cell invasion, motility and
IC50: 50 lM (24 h) [149], 15.7 lM migration through inhibition of EGF-induced MMP-9 via suppressing FAK and ERK signaling
[151] pathways [110].
pEGCG 20 lM [150]; 50 lmol/L [79] Inhibit cell proliferation via Epigenetic repression of hTERT [150]. In addition, it inhibits
significantly tumor growth in vivo associated with increased proteasome inhibition and apoptosis
induction in tumor tissues [79].
EGCG + Curcumin E (20,25,35 and 40 lM) and/or C Inhibits cancer proliferation in vitro and in xenograft mouse models through inhibition of VEGFR-1,
(2,3,4 and 6 lM) EGFR and AKT protein level [31].
EGCG (E) + Vitexin-2-O- E (10,20,30,40,50 lg/mL), Mixture activates ROS mediated mitochondrial pathway causing G0/G1 cell cycle arrest and
xyloside (X) + Raphasatin (30,50,80, 100,120 lg/mL) and/or G induces apoptosis [46].
(G) (5,10,15,30,50 lg/mL).
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
IC50: E (135 ± 16), X (158 ± 13), G (36
± 5) lg/mL
EGCG-Ptx-PLGA-Casein-NPs – Induce apoptosis, inhibite NF-jB activation and downregulate the key genes associated with
angiogenesis, tumor metastasis and survival [141].
EGCG-LbL-PSS/PAH-NPs 1 or 5 lM [152] Inhibit HGF-induced c-Met signaling after prolonged pre-incubation [13,152].
MCF-7 EGCG 10, 20, 30, 40 and 50 [73]; 10, 25, 50 Induces growth inhibition and apoptosis through surviving expression downregulation via
and 100 [109]; 1 and 10 lM [153]; suppressing AKT pathway and activation of caspase-9 [73]. Inhibits cell migration and invasion
20, 40 and 60 [150]; 0.01–1000 lM through downregulation of VASP expression and Rac1 activity [109]. Inhibits nicotine and
[151]; 10, 20 and 40 lM [154]; 0.1, 1, estradiol-induced cell proliferation via downregulation of a9-nicotinic acetylcholine receptor
10, 50 and 100 lM [146]. IC50: 50 lg/ signaling pathway [153]. Inhibits cell viability and proliferation through epigenetic repression of
mL [73,109]; 44.1 lM [151] 40 lM hTERT [150], inhibition of glucose uptake and metabolism [151], epigenetic downregulation of ER-
[154]; 19–20 lM [146] a via p38MAPK/CK2 activation [154] and activation of Cav3.2 channels via elicit of Ca2 + spike
[146].
pEGCG 20 lM [150] Inhibit cell proliferation via epigenetic repression of hTERT [150]
EGCG + Curcumin E (2, 4, 10, 20, 40, 100) lM + C 20 lM Induce growth inhibition and apoptosis through upregulation of caspase-dependent apoptotic
signaling pathways and inhibition of P-glycoprotein pump function [125].
EGCG (E) + Vitexin-2-O- E (10,20,30,40,50 lg/mL), X Mixture activates ROS mediated mitochondrial pathway causing G0/G1 cell cycle arrest and
xyloside (X) + Raphasatin (30,50,80, 100,120 lg/mL) and G induces apoptosis [46].
(G) (5,10,15,30,50 lg/mL); IC50: E (135 ±
16), X (158 ± 13), G (36 ± 5) lg/mL
Hs578T EGCG 10 lM Inhibits cell proliferation and migration trough downregulation of VEGF expression [155].
T47D EGCG 10, 20 and 40 lM. Inhibits cell proliferation trough epigenetic downregulation of ER-a via p38MAPK/CK2 activation
IC50: 40 lM [154].
NF639 EGCG 20, 40, 60 and 80 mg/mL [156]; 40 lg/ Inhibits Her-2/Neu signaling, proliferation, and transformed phenotype of the Cancer Cells [156].
mL [108] Inhibit branching colony growth and cell invasion through induction of estrogen receptor a
expression via activating FOXO3a signaling [108].
4 T1 EGCG 10 mg/kg, IP injection on day 7, 9 and Suppresses tumor growth in vivo by inhibiting tumor-associated macrophage infiltration and M2
11 polarization [157].
ALDH1+ in SUM-149 EGCG 20, 40, 80 and 120 lg/mL Inhibits growth of cancer stem cells in vitro and in vivo. Inhibits spheroid formation and induces
and SUM-190 stem apoptosis [133].
cells
CD44+/CD24 in MDA- EGCG 20, 30 and 40 lM Inhibits tumorsphere formation and reduces cancer stem cell population through downregulation
MB-231 and MDA-MB- of estrogen receptor-a36, EGFR, p-ERK1/2 and p-AKT [158].
436 stem cells
Lung Cancer A549 EGCG 40–300 lM, IC50: 265 ± 7.1 lM (24 h) Induces cell apoptosis through inhibition of FASN activity and EGFR signaling pathway [101].
in vitro and 40 mg/kg/week by IP Reduces xenograft tumor growth and angiogenesis in vivo [101,102]through inhibiting of oncogene
injection into BALB/c nude female and IGF-1via suppressing HIF-1a and VEGF protein expression [103,104]. Inhibits cell proliferation
mice for 33 days [101]. 50 and 100 through upregulation of endostatin expression and suppression of VEGF expression [102] and
lM in vitro and 0.05% in drinking suppressing of the expression of the cell death-inhibiting gene, Bcl-xL [161]. Inhibits cell growth
water into BALB/c nude male mice for through induction of G0/G1 cell cycle arrest via inhibiting EGFR/cyclin D1 signaling [86] and
21 days [102]. 1, 5, 10, 20 and 40 lM upregulation of miR-210 expression via stabilizing HIF-1a [162]. Inhibits the anchorage-indepen-
[159]. 10, 25, 50 and 100 [104,105]. dent growth of cancer cells, induces p53 accumulation and upregulates its target genes, promotes
10–40 lM [86]. 80 lM [160]. 25 and the stability of p53 and MDM2, promotes nuclear localization and activity of p53, inhibits
100 [161]. 100 lM into flanks of nude proteasomal degradation-dependent p53 ubiquitination and inhibits the interaction of p53 and
mice [103–105]. 5,10,25 and 50 [162] MDM2 [159]. Inhibits HPV-16 oncoprotein induced angiogenesis conferred by cancer cells through
the inhibition of HIF-1a protein expression and HIF-1a dependent expression of VEGF, IL-8, and
CD31 as well as activation of Akt [104]. Inhibits cancer chemo-resistant variants through
downregulation of Axl and Tyro 3 expression [160]. Inhibits nicotine-induced migration, invasion
and upregulation of HIF-1a, VEGF, COX-2, p-Akt and p-ERK [105].
EGCG + Curcumin 10,20,40 lM EGCG and/or the same Inhibit cell growth in vitro and in vivo through induction of cell cycle arrest at G1 and S/G2 phases
concentrations for curcumin in vitro via downregulating cyclin D1 and cyclin B1 [45].
while fourteen 3 to 4-week old
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
female BALB/c nude mice were i.p.
implanted with 5 106 A549 cells.
At the third day after the A549 cells
injected, the mice were randomized
into two groups (7 mice/group) and
treated with control (NS, 100 mL/kg)
or EGCG and curcumin (20 mg/kg,
respectively) [45]
CL13 EGCG 5,10,25 and 50 Suppresses cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-
1a [162].
EGCG + N-acetylcysteine (0–100) lM of EGCG in the presence EGCG and NAC interact to form EGCG-20 -NAC adduct which induces cell culture apoptosis [126].
(NAC) or absence of 0–2 mM NAC.
H1299 EGCG 10,20,30,40 and 50 lM in vitro, 0.1, Inhibits cancer growth in vivo and in vitro and induces ROS and cell apoptosis [163]. Suppresses
0.3 and 0.5% in diets and 30 mg/kg/d cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1a [162].
by IP injection into male NCr nu/nu
mice for 45 days.
IC50: 20 lM in vitro and 0.15 lM
in vivo [163]; 5,10,25 and 50 [162]
H460 EGCG 1,5,10,20 and 40 lM [159]. 10–40 lM Inhibits the anchorage-independent growth of cancer cells, induces p53 accumulation and
[86]. 80 lM [160]. 5,10,25 and 50 upregulates its target genes, promotes the stability of p53 and MDM2, promotes nuclear
[162] localization and activity of p53, inhibits proteasomal degradation-dependent p53 ubiquitination
and inhibits the interaction of p53 and MDM2[159]. Inhibits cell growth through induction of G0/
G1 cell cycle arrest via inhibiting EGFR/cyclin D1 signaling [86]. Inhibit cancer cells proliferation
including their chemo-resistant variants through downregulation of Axl and Tyro 3 expression
[160]. Suppresses cancer cell growth through upregulating miR-210 expression caused by
stabilizing HIF-1a [162].
H1650 EGCG 1,5,10,20 and 40 lM [159]. 10–40 lM Inhibits the anchorage-independent growth of cancer cells, induces p53 accumulation and
[86] upregulates its target genes, promotes the stability of p53 and MDM2, promotes nuclear
localization and activity of p53, inhibits proteasomal degradation-dependent p53 ubiquitination
and inhibits the interaction of p53 and MDM2 [159]. Inhibits cell growth through induction of G0/
G1 cell cycle arrest via inhibiting EGFR/cyclin D1 signaling [86].
H1299 EGCG 5,10,50 and 100 lM [106] Reduces cell motility in vitro wound healing assay. Increases Young’s modulus of H1299 from 1.24
to 2.25 showing a 2-fold increase in cell stiffness, i.e. rigid elasticity of cell membrane. Furthermore,
inhibits high expression of vimentin and Slug in the cells at a leading edge of scratch. Induces
inhibition of EMT phenotypes by alteration of membrane organization [106].
Lu99 EGCG Reduces cell motility in vitro wound healing assay. Increases Young’s modulus of Lu99 from 1.29 to
2.28 showing a 2-fold increase in cell stiffness, i.e. rigid elasticity of cell membrane. Furthermore,
inhibits high expression of vimentin and Slug in the cells at a leading edge of scratch. Induces
inhibition of EMT phenotypes by alteration of membrane organization [106].
11
12
Table 3 (continued)
Cancers Cell lines EGCG Combination Dose & IC50 Biological effects
H69 EGCG 20,40,50,60,80,100,120,140,150 and Induces cell apoptosis, reduces telomerase activity and inducts a cell-cycle block in S phase [164].
H69VP 200 lM. IC50: 70 lM (24 h)
NCI-H460 EGCG 10,25,50 and 100 in vitro [104] Inhibits HPV-16 oncoprotein induced angiogenesis conferred by cancer cells through the inhibition
of HIF-1a protein expression and HIF-1a dependent expression of VEGF, IL-8, and CD31 as well as
activation of Akt [104].
EGCG + Curcumin 10,20,40 lM EGCG and/or the same Inhibit cell growth in vitro and in vivo through induction of cell cycle arrest at G1 and S/G2 phases
concentrations for curcumin via downregulating cyclin D1 and cyclin B1 [45].
– EGCG + Polyphenon E Benzo(a)pyrene [B(a)P]-induced lung Mixture inhibits significantly pulmonary adenoma formation and growth in vivo [148].
cancer in female A/J mice (100 mg/
kg).
Pancreatic cancer AsPC-1 EGCG 5–80 lM [48] Induces apoptosis through cell cycle via G1 cell cycle arrest, regulation of cyclin D1, cdk4, cdk6, p21
Hs 766 T and p27, activation of ROS-mediated, p53-indepdendent apoptosis signaling, inhibition of Ras/Raf-
PANC-1 EGCG 5–80 lM [48] 1/ERK1/2 signaling, induction of MEKK1, JNK1/2 and p38 MAPK activities [48].
EGCG + Pterostilbene 20, 30 and 40 lM EGCG with/without The combination have additive, antiproliferative effects in vitro altering the apoptotic mechanisms
30 lM Pterostilbene by modulation at different points in the mechanism as well as the cell cycle arrest effect [36].
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
MIA-Pa-Ca-2 EGCG 10, 100 and 1000 lM, IC50 below 50 Induces apoptosis through cell cycle via G1 cell cycle arrest, regulation of cyclin D1, cdk4, cdk6, p21
lM [165]; 5–80 lM [48] and p27, activation of ROS-mediated, p53-indepdendent apoptosis signaling, inhibition of Ras/Raf-
1/ERK1/2 signaling, induction of MEKK1, JNK1/2 and p38 MAPK activities [48]. Inhibits Hsp90
function by impairing Hsp90 association with co-chaperones resulting anti-proliferating effects
[165].
EGCG + TRAIL 50 lg/ml E+5 ng/ml T A synergistic increase in apoptosis and cleavage of procaspase-3. Furthermore, clonogenic cell
survival assay demonstrates the significant diminishment of cancer cell proliferation in the
presence of both EGCG and TRAIL [74].
EGCG + Pterostilbene 20, 30 and 40 lM EGCG with/without The combination have additive, antiproliferative effects in vitro altering the apoptotic mechanisms
30 lM Pterostilbene by modulation at different points in the mechanism as well as the cell cycle arrest effect [36].
Ovarian cancer SKOV3 EGCG 20,30,40 and 50 lg/mL [166]; Inhibits cell proliferation and induces apoptosis through inhibition of cell cycle and DNA synthesis
20,40,60,80,100 and 120 lg/mL inducting NDA damage [166]. Downregulates AQP5, nuclear p65 and Ij-Ba expressions [167].
[167]; 100 lM [107] Inhibit cell motility via suppression of Hsp90 chaperone system [107].
Oral cancer SAS EGCG 20 and 40 lM [168] Provides antitumor immunity through inhibition of indoleamide 2,3-dioxygenase (IDO) expression
Cal-27 via blocking the IFN-c-induced JAK-PKC-d-STAT1 signaling pathway [168].
Ca-922
SCC-9 EGCG 5,10,15 and 20 lM in vitro; 10 and 20 Inhibits invasion, epithelial-mesenchymal transition, and tumor growth through downregulation of
mg/day/kg by oral gavage into the MMP-2, uPA, p-FAK, p-Src, snail-1 and vimentin [111].
right front axilla of BALB/c nude mice
SCC-4 EGCG 20 and 40 lM [168]; 10, 20, 50, 100, Provides antitumor immunity through inhibition of indoleamide 2,3-dioxygenase (IDO) expression
and 200 lM [169] via blocking the IFN-c-induced JAK-PKC-d-STAT1 signaling pathway [168]. Suppresses cell
proliferation and promotes apoptosis and autophagy through upregulation of BAD, BAK, FAS, IGF1R,
WNT11, and ZEB1 genes expressions and downregulation of CASP8, MYC, and TP53 [169].
KB EGCG 5,10,20,40,80,100,150 and 200 lM Inhibits HGF-induced cell growth and invasion through suppression of HGF/c-Met signaling
pathway [112].
HSC-3 EGCG 20 and 40 lM [168]; 10,25,50 and Provides antitumor immunity through inhibition of indoleamide 2,3-dioxygenase (IDO) expression
100 lM [113] via blocking the IFN-c-induced JAK-PKC-d-STAT1 signaling pathway [168]. Inhibits cancer invasion
via repressing functional invadopodia formation [113].
YD-10B EGCG 10,25,50 and 100 lM in vitro; 20 mg/ Inhibits cancer invasion in vitro and in vivo via repressing functional invadopodia formation and
d/kg IP injection into the tongue of FAK/Src signaling [113].
male BALB/c athymic nude mice
every other day for 4 weeks
Hypopharyngeal FaDu EGCG 5,10,20,40,80,100,150 and 200 lM Inhibits HGF-induced cell growth and invasion through suppression of HGF/c-Met signaling
cancer pathway [112].
Laryngeal cancer SNU-899 EGCG 5,10,20,40,80,100,150 and 200 lM Inhibits HGF-induced cell growth and invasion through suppression of HGF/c-Met signaling
SNU-1066 pathway [112].
Nasopharyngeal (Oct4high/Nanoghigh/b- EGCG+Cisplatin (20 and 40 lM) E and/or (0.01, 0.1, 1 Mixture inhibits spheroid formation and cell viability as well as EGCG enhances chemo-sensitivity
cancer cateninhigh/ABCG2high/ and 10 lg/mL) C of cisplatin in vitro through downregulation of Oct4, b-Catenin, Nanog, ABCG2, MRP-1, MDR-1, p-
MRP-1high/MDR-1high) STAT3, Bcl-2, Survivin and c-Myc [170].
in TW01 CSCs
(Oct4high/Nanoghigh/b-
cateninhigh/ABCG2high/
MRP-1high/MDR-1high)
in TW06 CSCs
CNE2 EGCG + Cisplatin EGCG (20 lM) + Cisplatin (10 lM) EGCG increases chemo-sensitivity of cisplatin in vivo [138].
were injected subcutaneously into
the flanks of nude mice and allowed
to grow for 8 weeks
CD44+ in CNE2 CSCs EGCG 25,50 and 75 lM Inhibits nasopharyngeal cancer stem cell self-renewal and migration and reverses the epithelial–
mesenchymal transition via NF-jB p65 inactivation [138].
C666-1 EGCG + Cisplatin EGCG (20 lM) + Cisplatin (10 lM) EGCG increases chemo-sensitivity of cisplatin in vivo [138].
were injected subcutaneously into
the flanks of nude mice and allowed
to grow for 8 weeks
CD44+ in C666-1 CSCs EGCG 25,50 and 75 lM Inhibits nasopharyngeal cancer stem cell self-renewal and migration and reverses the epithelial–
mesenchymal transition via NF-jB p65 inactivation [138].
Gastric cancer BGC-823 EGCG 1.5 mg/d IP injection for 56 days Inhibits xenograft angiogenesis and tumor growth [40].
EGCG + Docetaxel –
EGCG + Capecitabine 200 mg/kg capecitabine daily by oral Inhibits xenograft angiogenesis and tumor growth [38].
gavage + EGCG (IP injection of 1.5 mg
EGCG daily
AGS EGCG 5,10,25 and 50 lM in vitro and 50 lM Inhibits IL-6-induced angiogenesis in vitro and in vivo through inhibition of VEGF expression via
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
EGCG + AGS cells in vivo into BALB/c suppressing Stat3 activity [114]. Inhibits cell proliferation and induces cell apoptosis through
nude female mice for 7 days [114]; downregulation of Id1 expression [171].
20,40,60,80,120 and 240 lg/mL [171]
AZ521 EGCG 5,10,20 and 40 lM Inhibits cell proliferation through downregulation of DEAD-box RNA helicase p68 [172].
NUGC-3 EGCG 25,50,75 and 100 lM Induces cell apoptosis through inhibition of survivin expression downstream of p73 [78].
MKN-1
MKN-28
MKN-45
TMK-1
SGC-7901 EGCG 1.5 mg/d IP injection into female Inhibit tumor growth and angiogenesis through reducing VEGF-induced endothelial cell
BALB/c nude for 28 days proliferation, migration and tube formation [115].
Hepatocellular Hepa 1c1c7 EGCG 100 lM Inhibit cell motility via suppression of Hsp90 chaperone system [107].
Carcinoma SMMC-7721 EGCG + Ascorbic acid – Mixture strongly suppress proliferation and metastasis through scavenging of reactive oxygen
species [136].
HepG2 EGCG 15,30,60,120 and 240 Induces non-apoptotic cell death via ROS-mediated lysosomal membrane permeabilization [173].
EGCG + 5-FU 5-FU (0.05 lg) and EGCG (25 lmol) Combination significantly decreased the viability of cells, compared with EGCG or 5-FU-treated
cells [174].
CD133 and NANOG in EGCG 10 and 20 lM Inhibit sphere formation through inhibition of CD133, Nanog, ABCC1, ABCG2, Nek2 and p-Akt [174].
HepG2 cancer stem
cells
Squamous cell SCC VII/SF EGCG 5,10,20,40,80,100,150 and 200 lM Inhibits HGF-induced cell growth and invasion through suppression of HGF/c-Met signaling
carcinoma in vitro; 25,50 and 75 mg/kg/day IP pathway while it inhibits xenograft tumor growth in vivo via rising apoptosis [112].
injection into the flank of syngeneic
C3H/HeJ micefor 21 days
Prostate cancer PC-3 EGCG 0–50 lM. IC50 : 39.0 lM Antiproliferative effects through ERK1/2 activation via MEK-independent, PI3-K-dependent
signaling pathway [175].
1 and 25 lM. IC50: 25 lM Induces cell apoptosis through upregulation of caspase-9a expression [47].
EGCG + Curcumin 50 and 100 lM E and/or 50 lM C Induction of cell cycle arrest at both S and G2/M phases via upregulating p21 protein level [135].
EGCG + Quercetin 80 lM EGCG and/or 10 and 20 lM Mixture demonstrates enhanced inhibition of cell proliferation and induction of cell apoptosis
Quercetin in vitro by increasing the intracellular concentration of EGCG and decreasing EGCG methylation
[33].
EGCG + Sulforaphane 20 and 100 lM E and/or 25 lM S Reduction of cell viability via inhibition of AP-1 activation [34].
EGCG + Ascorbic acid + 50 microg/mL-500 lg/mL of the Combination inhibits cell proliferation and invasion through downregulation of MMP-2 and MMP-
Lysine + Proline + Arginine mixture 9 expressions [44].
EGCg-198AuNPs single-dose intra-tumoral 80% reduction of tumor volumes after 28 d demonstrating significant inhibition of tumor growth
administration in human prostate compared to controls [176].
cancer-bearing SCID mice
EGCG-LDH nanohybrids 25,50,75 and 100 lM/L; IC50: 16.66 Induce apoptosis within over 5-fold dose advantages compared to EGCG alone in in-vitro system
lM/L (24 h), 15.47 lM/L (48 h), [142].
16.33 lM/L (72 h)
13
14
Table 3 (continued)
Cancers Cell lines EGCG Combination Dose & IC50 Biological effects
EGCG-PLA-PEG-NPs 0.7,1.37,2.74 and 5.48 lmol/L in vitro, NPs enhance the bioavailability and limited unwanted toxicity of EGCG within 10-fold dose
IC50: 3.74 lmol/L (24 h); in vivo, mice advantage [13]. In addition, induce apoptosis within remarkably significant increase in pro-
each received 100 lg dissolved in PBS apoptotic Bax with a concomitant decrease in anti-apoptotic Bcl-2, increase in PARP cleavage and
thrice weekly [26] marked induction of p21 and p27 [26].
EGCG-PLGA-PEG-DCL-NPs 20 lM In vitro, NPs enhance the anti-proliferative activity compared to the free EGCG modulating
EGCG-PLGA-PEG-AG-NPs apoptosis and cell-cycle. In vivo, NPs enhance the bioavailability and limited unwanted toxicity
[49].
PC-3ML EGCG + Doxorubicin 30 and 60 lM E + D in vitro; 0.14–57 Combination enhances the inhibition of metastatic tumor growth [41].
mg/kg E and/or 2 mol/L–0.07 mg/kg
D into immunodeficiency mice
(CD44+CD133+in PC-3) EGCG 30 and 60 lM Inhibits growth and spheroid formation, induces apoptosis, inhibits EMT, inhibits migration and
CSC invasion and downregulates Casp3/7, Bcl-2, Survivin, XIAP, Vimentin, Slug, Snail and nuclear b-
Catenin [132].
LNCaP EGCG 12 lM [177]; 20,40 and 80 lM [178]; Modulates cell growth, by affecting mitogenesis as well as inducing apoptosis, in cell-type-specific
10, 20, 40, and 80 lg/mL [87] manner which may be mediated by WAF1/p21-caused G0/G1-phase cell-cycle arrest, irrespective
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
of the androgen association or p53 status of the cells [87].Inhibits cell proliferation through
inhibition of PKC-a inhibition [177], prostate specific antigen (PSA) expression, AR transcriptional
activity, growth of relapsing R1Ad tumors and tumor derived serum PSA in vivo [178].
Green tea polyphenols 10–80 lg/mL In vitro and in vivo inhibition of testosterone-mediated induction of ornithine decarboxylase1
(GTP) [147].
EGCG + Quercetin 80 lM EGCG and/or 10 and 20 lM Quercetin enhances the anti-proliferative effects of EGCG and induction of cell apoptosis in vitro
Quercetin through increasing the intracellular concentration of EGCG and decreasing EGCG methylation [33].
EGCG + Ascorbic acid 50 microg/mL–500 lg/mL of the Combination inhibits cell proliferation and invasion through downregulation of MMP-2 and MMP-
+Lysine+Proline+Arginine mixture 9 expressions [44].
EGCG-PLGA-PEG-DCL-NPs 20 lM In vitro, NPs enhance the anti-proliferative activity compared to the free EGCG modulating
EGCG-PLGA-PEG-AG-NPs apoptosis and cell-cycle. In vivo, NPs enhance the bioavailability and limited unwanted toxicity
[49].
(CD44+CD133+in EGCG 30 and 60 lM Inhibits growth and spheroid formation, induces apoptosis, inhibits EMT, inhibits migration and
LNCaP) CSCs invasion and downregulates Casp3/7, Bcl-2, Survivin, XIAP, Vimentin, Slug, Snail and nuclear b-
Catenin [132].
DU-145 EGCG 5 lM; 10, 20, 40, and 80 lg/mL [87] Modulates cell growth, by affecting mitogenesis as well as inducing apoptosis, in cell-type-specific
manner which may be mediated by WAF1/p21-caused G0/G1-phase cell-cycle arrest, irrespective
of the androgen association or p53 status of the cells [87].
Inhibit cell motility and invasion through inhibition of c-Met signaling via altering the structure or
function of lipid rafts [122].
EGCG + Ascorbic acid + 50 microg/mL–500 lg/mL of the Combination inhibits cell proliferation and invasion through downregulation of MMP-2 and MMP-
Lysine + Proline + Arginine mixture 9 expressions [44].
EGCG-PLGA-PEG-DCL-NPs 20 lM In vitro, NPs enhance the anti-proliferative activity compared to the free EGCG modulating
EGCG-PLGA-PEG-AG-NPs apoptosis and cell-cycle. In vivo, NPs enhance the bioavailability and limited unwanted toxicity
[49].
CWR22R EGCG 50 mg/kg/d IP injection into nude Inhibits tumor growth and angiogenesis while promoting apoptosis of the prostate cancer cells
mice for 20 days. in vivo [123].
EGCG-P 86.7 mg/kg/d IP injection into nude EGCG-P is more stable and effective than EGCG enhancing the inhibition of the tumor growth,
mice for 20 days. angiogenesis and induces apoptosis of the prostate cancer cells in vivo [123].
BCaPT10 EGCG 2–200 lM in vitro; 0.06% in water Inhibits cell motility in vitro via suppression of Hsp90 molecular chaperone system which supports
into male athymic mice for 1 week malignant phenotype [124].
before xenograft surgery
BCaPM-T10 EGCG 0.06% in water into male athymic Inhibits a molecular chaperone supportive of the malignant phenotype [124].
mice for 1 week before xenograft
surgery
22Rm1 EGCG-CS-NPs 3 and 6 mg/kg by oral administration Inhibit AR-positive 22Rm1 tumor xenograft growth and secreted prostate-specific antigen levels
into athymic nude mice for 25 days compared with EGCG and control groups. Significant induction of poly (ADP-ribose) polymerases
cleavage, increase in the protein expression of Bax with concomitant decrease in Bcl-2, activation of
caspases and reduction in Ki-67 and proliferating cell nuclear antigen [143].
– EGCG Five-week-old male TRAMP offspring Inhibits cell proliferation and induces apoptosis through downregulation of AR, IGF-1, IGF-1R, p-
were fed AIN-76A diet and 0.06% ERK 1/2, COX-2, and iNOS [179].
EGCG in tap water for28 weeks
– GTP – In vivo GTP oral infusion resulted in almost complete inhibition of distant site metastases.
Furthermore, GTP consumption caused significant apoptosis of cancer cells causing inhibition of
prostate cancer development, progression, and metastasis [127].
Melanoma Mel 928 EGCG-CS-NPs 0.5,1.0,2.0,4.0 and 8.0 lM in vitro; Induct apoptosis and cell cycle inhibition along with the growth of Mel 928 tumor xenograft.
IC50: 7 lM (48 h). 100 lg/mice;120 Inhibited proliferation (Ki-67 and PCNA) and induced apoptosis (Bax, PARP) in tumors harvested
lL treatment volume into Athymic from the treated mice within 8-fold dose advantage of nanoformulation over native EGCG [144].
(nu/nu) male nude mice.
Leukemia CEM (MSN@EGCG)-NPs 0.4, 0.8, 1.2 and 1.6 lM in vitro and Inhibit cell viability and proliferation in vitro and in vivo within a highly biocompatible and
100; NPS are i.v. injected into female biodegradable EGCG-coated MSN nanoparticles [180].
Balb/c mice
Cervical cancer HeLa EGCG 10, 25, 50 and 100 lM [88]; Cell growth inhibition trough gene expression regulation and induction of cell cycle arrest [88].
10,20,30,40,50,60,70,80,90 and 100 Inhibit cell proliferation, invasion and migration, and induce cell apoptosis through G1 cell cycle
lM [85]; IC50: 47.9 lM [88]; 100 lM arrest and DNA damage, downregulation of MMP-9 gene and upregulation of TIMP-1 gene [85].
(24 h) and 50 lM (48 h) [85].
CaSki EGCG 10, 25, 50 and 100 lM. IC50: 27.3 lM Cell growth inhibition trough gene expression regulation and induction of cell cycle arrest [88].
(24 h)
Colorectal cancer HCT-116 EGCG 0.05, 0.1, 0.5, 1, 5 and 10 lM [119]; Suppresses cancer cells growth through cyclin D1 degradation and p21 transcriptional activation
0.1, 0.5, 1, 5 and 10 lM [120]; 1, 10 via ERK, IKK and PI3K signaling pathway [89]. Inhibits Met signaling [119] which helps to attenuate
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
and 50 lM [89]; 50 and 100 lM tumor spread/metastasis, independent of H2O2-related mechanisms [120]. Inhibits cell prolifera-
[181]; 12.5,25,50 and 100 lM [118]. tion through inhibition of HGF-induced Met/ERK/AKT signaling pathway [119]and through
IC50: 3 lM [119] inhibition of Akt, activation and induction of p38 MAPK activation [118]. Induces cell apoptosis
through cancer-specific induction of ROS and epigenetic modulation of expression of apoptosis-
related genes, such as hTERT [181].
EGCG + Panaxadiol E (0,10,20 and 30 lM) and/or P (0,10 Inhibit cell proliferation and induce cell cycle arrest [35].
and 20 lM)
EGCG + EPA-FFA + GS EGCG (0–175 lM) + EPA-FFA (0–150 Combination completely inhibited the mTOR signaling. Moreover, the treatment led to changes of
lM) + GS extract (0–15 lM) for 24 h protein translation of ribosomal proteins, c-Myc and cyclin D1. In addition, combination reduces
clonal capability of cells, with block of cell cycle in G0/G1 and induction of apoptosis [128].
EGCG + 5-FU 5-FU (0.05 lg) and EGCG (25 lmol) Combination significantly decreased the viability of cells, compared with EGCG or 5-FU-treated
[174]; 25–400 lM of EGCG and/or cells [174] like the EGCG enhances 5-FU sensitivity and induces apoptosis in 5-FU resistant cancer
2.5–40 lM 5-FU [37]. cells [37].
EGCG + NaB 10 lM E +(1,2,3,4,5 and 6 mM) N. The combination treatment induces apoptosis and G2/M cell cycle arrest through decrease in
IC50: 10 lM E + 5 mM N HDAC1, DNMT1, survivin and HDAC activity [43].
CD133 and NANOG in EGCG 10 and 20 lM Inhibit sphere formation through inhibition of CD133, Nanog, ABCC1, ABCG2, Nek2, and p-Akt
HCT-116 stem cells [174].
CD44, CD133 and EGCG 50, 100, 200 and 400 lM Induces apoptosis and cell cycle arrest, attenuate spheroid formation and enhance
ALDH1 in HCT-116 chemosensitivity of 5-FU in vivo [37].
stem cells
HCT-8 EGCG 10,20 and 35 lg/mL [182] Inhibits proliferation in vitro and in vivo, induces apoptosis and affects cell cycle of cancer cells via
inhibiting of HES1 and Notch2 expressions [182].
Caco-2 EGCG 1, 5 and 10 lM [183]; 1, 10 and 50 Induces cell growth inhibition as EGCG and 67LR at a physiological concentration can activate
lM [89] myosin phosphatase by reducing MYPT1 phosphorylation [183] or through cyclin D1 degradation
and p21 transcriptional activation via ERK, IKK and PI3K signaling pathway [89].
EGCG (E) + Vitexin-2-O- E (10,20,30,40,50 lg/mL), X Mixture activates ROS mediated mitochondrial pathway causing G0/G1 cell cycle arrest and
xyloside (X) + Raphasatin (30,50,80, 100,120 lg/mL) and G induces apoptosis [46].
(G) (5,10,15,30,50 lg/mL);
IC50: E (135 ± 16), X (158 ± 13), G (36
± 5) lg/mL
EGCG-CS-CPP-NPs 0.063, 0.125 and 0.250 mg/mL Nanoparticles enhance stability, penetration and transportation of EGCG through cancer cells [184].
HT-29 EGCG 0.1, 0.5, 1, 5 and 10 lM [120]; 1, 10 Inhibits Met signaling and helps to attenuate tumor spread/metastasis, independent of H2O2-
and 50 lM [89]; 10,20 and 35 lg/mL related mechanisms [120]. Suppresses cancer cells growth through cyclin D1 degradation and p21
[182]; 5, 10 and 20 mg/kg/d oral transcriptional activation via ERK, IKK and PI3K signaling pathway [89]. Inhibits proliferation
gavage to male BALB nude mice for in vitro and in vivo and induces apoptosis and affectes the cell cycle of cancer cells via inhibiting of
28 days [185] or/14 days [182]. HES1 and Notch2 expressions [182]. Inhibit tumor growth and metastasis in vivo by upregulating
the Nrf2-UGT1A signaling [185].
EGCG + NaB 10 lM E + (1,2,3,4,5 and 6 mM) N. The combination treatment induces apoptosis and G2/M cell cycle arrest through decrease in
IC50: 10 lM E + 5 mM N HDAC1, DNMT1, survivin and HDAC activity [43].
15
16
Table 3 (continued)
Cancers Cell lines EGCG Combination Dose & IC50 Biological effects
SW837 EGCG 25 lg/mL in vitro; 0.01% and 0.1% in Inhibits tumor growth in vivo and in vitro through activation of VEGF/VEGFR axis via suppressing
drinking water to BALB/c nude mice the expression of HIF-1a and several major growth factors [116]. Provides antitumor
for 35 days [116]; 10,50 and 100 lM immunotherapy through inhibiting the expression and function of indoleamine 2,3–dioxygenase
[186] via suppression of STAT1 activation [186].
SW480 EGCG 1, 10 and 50 lM [89]; 10,20 and 35 Suppresses cancer cells growth through cyclin D1 degradation and p21 transcriptional activation
lg/mL [182] via ERK, IKK and PI3K signaling pathway [89]. Inhibits proliferation in vitro and in vivo and induces
apoptosis and affectes the cell cycle of cancer cells via inhibiting of HES1 and Notch2 expressions
[182].
EGCG + Panaxadiol E (0,10,20 and 30 lM) and/or P (0,10 Inhibit cell proliferation and induce cell cycle arrest [35].
and 20 lM)
EGCG + EPA-FFA + GS EGCG (0–175 lM) + EPA-FFA (0–150 Combination completely inhibits the mTOR signaling and lead to changes in protein translation of
lM) + GS extract (0–15 lM) for 24 h ribosomal proteins, c-Myc and cyclin D1 and reduces clonal capability of cells, with block of cell
cycle in G0/G1 and induction of apoptosis [128].
EGCG + 5-FU 25–400 lM of EGCG and/or 2.5–40 EGCG enhances 5-FU sensitivity and induces apoptosis in 5-FU resistant cancer cells [37]
lM 5-FU [37].
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
SW620 EGCG 25,50,75 and 100 lg/mL Inhibits proliferation and migration in vitro through inactivation of PAR2-AP and factor VIIa and by
the way inhibition of the ERK1/2 and NF-jB pathways [117].
LoVo EGCG 10,20 and 35 lg/mL Inhibits proliferation in vitro and in vivo and induces apoptosis and affects the cell cycle of cancer
cells via inhibiting of HES1 and Notch2 expressions [182].
EGCG (E) + Vitexin-2-O- E (10,20,30,40,50 lg/mL), Mixture activates ROS mediated mitochondrial pathway causing G0/G1 cell cycle arrest and
xyloside (X) + Raphasatin (30,50,80, 100,120 lg/mL) and G induces apoptosis [46].
(G) (5,10,15,30,50 lg/mL); IC50: E (135 ±
16), X (158 ± 13), G (36 ± 5) lg/mL
RKO EGCG 12.5,25,50 and 100 lM in vitro; 30 Inhibits cell proliferation and induces cell apoptosis through inhibition of Akt, activation and
mg/kg IP injection to SCID male mice induction of p38 MAPK activation; inhibits liver metastasis in vivo and suppresses angiogenesis and
every other day for 2 weeks [118] induces apoptosis in liver metastasis [118].
EGCG + NaB 10 lM E + (1,2,3,4,5 and 6 mM) N. The combination treatment induces apoptosis and G2/M cell cycle arrest through decrease in
IC50: 10 lM E + 5 mM N HDAC1, DNMT1, survivin and HDAC activity. Furthermore, p53-dependent induction of p21 and an
increase in nuclear factor kappa B (NF-kB)-p65 [43].
– EGCG-(poly[lactic-co- – Modify DNA damage in human lymphocytes from colon cancer patients and healthy individuals
glycolic acid])-NPs treated in vitro with platinum-based chemotherapeutic drugs [187].
Glioma 1321N1 EGCG 50,100,150,200,250 and 300 lg/mL; Inhibits proliferation and induces apoptosis through activation of caspase-3 [30].
IC50: 82.0 ± 10.31 lg/mL (24 h)
EGCG + [6]-Gingerol 50 lg/mL EGCG + GING; IC50: 40 ± Mixture inhibits proliferation and induces apoptosis through activation of caspase-3 [30].
8.62 lg/mL
EGCG + Tocotrienol-rich 100 lg/mL EGCG + TRF; IC50: 100 ±
fraction (TRF) 9.5 lg/mL
LN18 EGCG 50,100,150,200,250 and 300 lg/mL; Inhibits proliferation and induces apoptosis through activation of caspase-3 [30].
IC50: 134.0 ± 11.36 lg/mL (24 h)
EGCG + [6]-Gingerol 50 lg/mL EGCG + GING; IC50: 24 ± Mixture inhibits proliferation and induces apoptosis through activation of caspase-3[30].
2.65 lg/mL
EGCG + Tocotrienol-rich 80 lg/mL EGCG + TRF; IC50: 88 ±
fraction (TRF) 11.14 lg/mL
SW1783 EGCG 50,100,150,200,250 and 300 lg/mL; Inhibits proliferation and induces apoptosis through activation of caspase-3 [30].
IC50: 300.0 ± 9.10 lg/mL (24 h)
EGCG + [6]-Gingerol 60 lg/mL EGCG + GING; IC50: 60 ± Mixture inhibits proliferation and induces apoptosis through activation of caspase-3 [30].
5.6 lg/mL
EGCG + Tocotrienol-rich 100 lg/mL EGCG + TRF; IC50: 270 ±
fraction (TRF) 4.16 lg/mL
U87 EGCG + Temozolomide 100 lM E + 100 lM TMZ EGCG inhibits properties of glioma stem-like cells (CD133high/ALDH1high) and synergizes with TMZ
U251 through downregulation of P-glycoprotein inhibition [42].
SHG-44
C6
Skin cancer A431 EGCG 5, 10, 20, 40, and 80 lg/mL [90];100– Inhibits cell growth, viability and induces cell apoptosis [90,91] through inhibition of EGFR
200 lM [91]; 10,20,40 and 60 lg/mL signaling [91], inhibition of pRb-E2F/DP pathway [90], inducing cell cycle arrest [90,91], inhibition
[188] of Cip1/p21 but no change in Kip1/27, CDK2, and cyclin D1 and a decrease in CDK4 only at low
doses [91] and inactivation of b-catenin signaling [188].
SCC-13 EGCG 10,20,40 and 60 lg/mL [188]; Reduces cell viability [188], induces cell apoptosis [188,189] and inhibits cell growth [189] through
10,20,40,60 and 100 lM [189] inactivation of b-catenin signaling [188] and influencing PcG-mediated epigenetic regulation of cell
cycle-related genes [189].
– Green tea polyphenols 200lL of a 5% solution Prevent the adverse effects of UV radiation in humans [190].
Esophageal cancer TE-8 EGCG 40 lM in vitro Reduces cell viability and invasion in vitro through reduction of p-ERK1/2, c-Jun and COX-2, and
EGCG (E) + Curcumin (C) E (40 lmol/L), C (40 lmol/L), and L (4 activation of caspase-3 whereas it inhibits tumor growth in vivo through suppressing the
EGCG (E) + Curcumin (C) + lmol/L) in vitro expression of Ki67, p-ERK1/2 and COX-2 [32].
Lovastatin (L)
SKGT-4 EGCG 40 lM in vitro and 50 lg/kg/d by oral
intake into nude mice for 30 days (5
day/week)
EGCG (E) + Curcumin (C) E (40 lmol/L), C (40 lmol/L), and L (4
EGCG (E) + Curcumin (C) + lmol/L) in vitro
Lovastatin (L)
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
Adrenal cancer NCI-H295 EGCG 10, 20, 30 and 40 mM in vitro; IC50: Induces cell apoptosis through activation of caspase-dependent, caspase-independent, the
20.34 mM (48 h) mitochondrial, death receptor and endoplasmic reticulum stress apoptotic signaling pathways.
EGCG also downregulate the expression of anti-apoptotic proteins, including BCL-2, BCL-XL and
XIAP. It upregulate the expression of pro-apoptotic proteins, including Apaf-1, BAD and BAX. It
regulate molecular chaperones, such as 70 kDa heat shock protein (HSP70), HSP90 and GRP78 [76].
Bladder cancer T-24 EGCG 10, 20, 40, 80 mg/mL Inhibit cell adhesion, migration and invasion through downregulion of MMP-9 expression via
blocking of NF-jB signaling pathway [121].
EGCG (in green tea extract) 10,50,100,500 and 1000 mg/mL of the Mixture inhibits critical steps of cancer development and spread, such as MMP-2 and -9 secretions
+ Ascorbic acid + Lysine + mixture and invasion [137].
Proline + Arginine
Pheochromocytoma PC-12 EGCG 15 mg/kg IP injection into male BALB/ Inhibits tumor growth and induces cancer cell apoptosis via acetylation of amyloid precursor
c nude mice every other day for 15 protein [77].
days
Neuroblastoma (Nanoghigh/Oct4high/ EGCG 1,10,50 and 100 lM Inhibits the development of TICs in BE(2)-C cells as well as inhibits sphere formation and induces
ATP7Alow/DKK2low)in apoptosis [134].
BE(2)-C CSCs
Ehrlich’s ascites EAC EGCG + HDHA-DOX-NPs E (20 mg/kg b.wt., orally through EGCG enhances the anticancer activities of HDHA-NPs significantly increasing the mean survival
carcinoma gavage) + HDHA-DOX-NPs (1.5 mg/kg time of the animals and inducing apoptosis [145].
b.wt.) intravenously into Swiss albino
mice.
Head and neck K3 EGCG + Cisplatin 5,10,20 and 50 lM E and/or 5,10 and Inhibit sphere formation and CD44+ cell population; enhances chemosensitivity of cisplatin in vitro
squamous K4 20 lM C and in vivo through downregulation of Oct4, Sox2, ABCC2, ABCG2 and Notch1 [39].
carcinoma (HNSC) K5
CSCs
17
18 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
that give rise to distant metastases [130]. Recently, studies found mixture induces G0/G1 cell cycle arrest in MDA-MB-231 and MCF-
that EGCG can induce apoptosis to inhibit CSCs in vitro and 7 breast, Caco-2 and LoVo colorectal cancer cells [46]. EGCG and
in vivo. Besides, its effect of spheroid formation inhibition in CSCs, pterostilbene combination has antiproliferative effects in vitro as
it induces apoptosis, and enhances chemo-sensitivity of chemo- a cell cycle arrest induction in pancreatic PANC-1 and MIA-Pa-
drugs in CSCs, for instance, EGCG induces apoptosis through down- Ca-2 cancer cells [36]. Similarly, EGCG and panaxadiol mixture
regulating Casp3/7, Bcl-2, survivin and XIAP in PC-3 and LNCaP inhibit cell proliferation and induce cell cycle arrest in both HCT-
prostate CSCs [132]. In addition, EGCG treatment induced apopto- 116 and SW480 colorectal cancer cells [35]. EGCG, EPA-FFA and
sis in the breast SUM-149 and SUM-190 CSCs [133], colon HCT-116 GS combination blocks cell cycle in G0/G1 in both HCT-116 and
CSCs [37] and neuroblastoma BE(2)-C CSCs[134], and enhance the SW480 colorectal cancer cells [128]. EGCG and NaB combination
chemosensitivity of 5-FU in vivo [37]. treatment induces G2/M cell cycle arrest through decreasing sur-
vivin in HCT-116, HT-29 and RKO colorectal cancer cells [43]. Fur-
Modulation of cellular proliferation thermore, EGCG inhibit cell proliferation via induction of cell cycle
arrest and attenuate spheroid formation in colorectal HCT-116
On the other hand, EGCG and curcumin combination inhibits CSCs [37].
cell proliferation and growth in vitro and in vivo in lung A549
and NCI-H460 cancer cells through induction of cell cycle arrest Inhibition of angiogenesis and related mechanisms
at G1 and S/G2 phases via downregulating cyclin D1 and cyclin
B1 [45] while same combination induces cell cycle arrest at both EGCG combinations has also been observed to inhibit angiogen-
S and G2/M phases via upregulating p21 protein level in PC-3 pros- esis, necrosis, motility, invasion, migration and metastasis in
tate cancer cells [135]. EGCG, vitexin-2-O-xyloside and raphasatin experimental cancer systems. In vivo GTP oral infusion resulted
Fig. 5. Schematic drawing of the regulative actions of EGCG combined with nanoparticles. This carton is based on the available literature.
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 19
in almost complete inhibition of distant site metastases in prostate cancer activities of HDHA-NPs significantly increasing the mean
cancer. Furthermore, GTP consumption caused significant apopto- survival time of the animals [145]. In vitro treatment of 20 lM of
sis of cancer cells causing inhibition of prostate cancer develop- EGCG-PLGA-PEG-DCL-NPs or EGCG-PLGA-PEG-AG-NPs into PC-3,
ment, progression, and metastasis [127]. A combination of EGCG LNCaP and DU-145 prostate cancer cells induces apoptosis upreg-
and curcumin or EGCG, curcumin and lovastatin was able to reduce ulating Bax, DR5, and P27 and decreasing Bcl2 and survivin [89].
cell viability and invasion in vitro in TE-8 and SKGT-4 esophageal Moreover, EGCG-PLA-PEG-NPs inhibit proliferation in PC-3
cancer cells through reduction of p-ERK1/2, c-Jun and COX-2, and prostate cancer cells through upregulation of p21 and p27 [26].
activation of caspase-3 whereas it inhibited tumor growth in vivo Finally, EGCG-NPs inhibit cell proliferation through cell cycle regu-
through suppressing the expression of Ki67, p-ERK1/2 and COX-2 latory proteins via downregulation of Cyclin A, Cyclin B1, Cyclin D3,
[32]. EGCG and doxorubicin mixture enhanced the inhibition of surviving, CDK2 and CDK6 and upregulation of P21 and P27 [49].
metastatic tumor growth in PC-3ML prostate cancer cells [41]. EGCG-NPs was also found to inhibit cancer angiogenesis and
EGCG, ascorbic acid, lysine, proline and arginine combination metastasis such as EGCG-Ptx-PLGA-Casein-NPs which downregu-
inhibited cell proliferation and invasion through downregulation lated the key genes associated with angiogenesis, tumor metastasis
of MMP-2 and MMP-9 expressions in LNCaP and DU-145 prostate and survival as well as induced apoptosis and inhibited NF-jB acti-
cancer cells [44]. In gastric cancer, xenograft angiogenesis and vation in MDA-MB-231 breast cancer cells [141].
tumor growth in BGC-823 cells were inhibited by a combination
of EGCG and docetaxel [40] or a mixture of EGCG and capecitabine Conclusion
[38]. EGCG and ascorbic acid combination strongly suppressed pro-
liferation and metastasis through scavenging of reactive oxygen Chemoprevention, also defined as ‘‘slowing the process of car-
species in SMMC-7721 hepatocellular carcinoma [136]. EGCG (in cinogenesis’’ concept appears to be a viable option for cancer con-
green tea extract), ascorbic acid, lysine, proline and arginine mix- trol. To be effective, chemopreventive intervention should be
ture inhibited critical steps of cancer development and spread, addressed during the early stages of the carcinogenesis process.
such as MMP-2 and -9 secretions and invasion in T-24 bladder can- A plethora of experimental evidences suggest that both dietary
cer cells [137]. and lifestyle factors act by balancing promotion/prevention of
In CSCs, EGCG was also found to inhibit migration and invasion, chronic inflammation and/or oxidative stress, sometime leading
for example, it inhibited growth, spheroid formation, migration alterations associated with cancer initiation. Within the chemopre-
and invasion and downregulates Casp3/7, Bcl-2, Survivin, XIAP, ventive armamentarium, the use of natural agents from dietary
Vimentin, Slug, Snail and nuclear b-Catenin in PC-3 and LNCaP sources is generally preferred with respect to bioactive molecules
CSCs [132]. In addition, EGCG inhibited self-renewal and migration deriving from other sources. Many of these natural occurring
and reversed the epithelial–mesenchymal transition via NF-jB p65 agents demonstrated antioxidant activity, and compounds belong-
inactivation in nasopharyngeal CNE2 and C666-1 CSCs [138]. ing to polyphenols chemical class may play a promising role for
cancer prevention. Epidemiological studies conducted in humans
support the existence of an association between natural polyphe-
Anticancer effects of EGCG combined with nanoparticls nols consumption and a reduced cancer risk. In the last decade, a
representative member of polyphenols, i.e. EGCG, has been the
On the other hand, nanotechnology mediated approaches to focus of a number of studies scrutinizing its beneficial effects on
develop drugs have attracted intense attention in cancer preven- health. Therefore, consumption of green tea has become more
tion and therapy research. Nanoparticles appears to hold great pro- and more popular in the world due to its versatile health benefits
mise in the field of cancer management because of its unique [29]. Moreover, interesting preclinical evidence and encouraging
physicochemical properties including nanometer size, large sur- initial clinical trials have been obtained testing EGCG as chemopre-
face area-to-mass ratio, and efficient interaction with cells [2]. Sid- ventive agent. However, despite its beneficial therapeutic poten-
diqui et al envisioned that nanoparticle-mediated delivery could be tial, EGCG presents important pharmacokinetics problems, due to
useful to control the toxicity and enhance the bioavailability of the inefficient systemic delivery and bioavailability. In order to
chemopreventive agents such as EGCG, and introduced the concept improve the poor systemic bioavailability and cellular uptake of
of ‘‘nanochemoprevention” [26,49,139,140]. These studies demon- EGCG, various strategies have been adopted, which include combi-
strated that EGCG encapsulated in polymeric nanoparticles (NPs) nation therapy or polytherapy that consumes EGCG with one or
exhibited over ten-fold dose advantage for exerting its apoptotic more medications. In particular, nanotechnology approaches could
and effects against cancer, both in vitro and in vivo [26,49]. In help overcome pharmacokinetics issues of EGCG by controlling its
breast cancer, EGCG-Ptx-PLGA-Casein-NPs induce apoptosis in toxicity and enhancing its bioavailability to introduce the concept
MDA-MB-231 cells through inhibiting NF-jB activation [141]. of ‘‘nanochemoprevention” [26,49,139,140]. In addition, recent
EGCG-LDH nanohybrids induce apoptosis within over 5-fold dose studies conducted implying both EGCG and CSCs to found that
advantages in vitro compared to EGCG alone in prostate PC-3 can- EGCG induces multiple of anticancer effects in CSCs and enhances
cer cells [142]. Similarly in PC-3, EGCG-PLA-PEG-NPs enhance the chemo-sensitivity of chemo-drugs in CSCs.
bioavailability and limited unwanted toxicity of EGCG within 10- In this review the current available studies of the anti-cancer
fold dose advantage [13] and induce apoptosis within remarkably effects of EGCG alone and combined with other dietary and phar-
significant increase in pro-apoptotic Bax with a concomitant maceutical agents as well as the recent novel nanotechnology
decrease in anti-apoptotic Bcl-2 (Fig. 5), increase in poly(ADP- approaches used to deliver sustained levels of EGCG have been
ribose) polymerase (PARP) cleavage and marked induction of p21 covered and discussed in order to introduce some furnish driving
and p27 [26]. In vivo oral administration of EGCG-CS-NPs induces force for further evolution of research on innovative database able
apoptosis in 22Rm1 prostate cancer cells increasing in Bax expres- to consolidate the chemopreventive potential of EGCG.
sion with a concomitant decrease in Bcl-2 and activation of cas-
pases [143]. Another study demonstrated apoptosis induction Acknowledgements
(Bax, PARP) in tumors harvested from the treated mice within 8-
fold dose advantage of nanoformulation over native EGCG [144]. The authors acknowledge support from Egyptian Ministry for
EGCG and HDHA-DOX-NPs combination induces apoptosis in Ehr- Higher Education for a fellowship to IR. IAS was supported by
lich’s ascites carcinoma (EAC) whereas EGCG enhances the anti- Mentored Research Scholar Grant (MRSG-11-019-01-CNE) from
20 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
the American Cancer Society. While preparing this review article [31] Somers-Edgar TJ, Scandlyn MJ, Stuart EC, Le Nedelec MJ, Valentine SP,
Rosengren RJ. The combination of epigallocatechin gallate and curcumin
the core resources of P30AR066524 were used.
suppresses ER alpha-breast cancer cell growth in vitro and in vivo. Int J
Cancer 2008;122:1966–71.
References [32] Ye F, Zhang GH, Guan BX, Xu XC. Suppression of esophageal cancer cell
growth using curcumin, (-)-epigallocatechin-3-gallate and lovastatin. World J
Gastroenterol 2012;18:126–35.
[1] Kohler BA, Sherman RL, Howlader N, Jemal A, Ryerson AB, Henry KA, et al.
[33] Wang P, Heber D, Henning SM. Quercetin increased the antiproliferative
Annual report to the nation on the status of cancer, 1975–2011, featuring
activity of green tea polyphenol (-)-epigallocatechin gallate in prostate cancer
incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J
cells. Nutr Cancer 2012;64:580–7.
Natl Cancer Inst 2015;107:djv048.
[34] Nair S, Barve A, Khor TO, Shen GX, Lin W, Chan JY, et al. Regulation of Nrf2-
[2] Rady I, Siddiqui IA, Rady M, Mukhtar H. Melittin, a major peptide component
and AP-1-mediated gene expression by epigallocatechin-3-gallate and
of bee venom, and its conjugates in cancer therapy. Cancer Lett
sulforaphane in prostate of Nrf2-knockout or C57BL/6J mice and PC-3 AP-1
2017;402:16–31.
human prostate cancer cells. Acta Pharmacol Sin 2010;31:1223–40.
[3] Frankish H. 15 million new cancer cases per year by 2020, says WHO. Lancet
[35] Du GJ, Wang CZ, Qi LW, Zhang ZY, Calway T, He TC, et al. The synergistic
2003;361:1278.
apoptotic interaction of panaxadiol and epigallocatechin gallate in human
[4] Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L.
colorectal cancer cells. Phytother Res 2013;27:272–7.
Current challenges in cancer treatment. Clin Ther 2016;38:1551–66.
[36] Kostin SF, McDonald DE, McFadden DW. Inhibitory effects of (-)-
[5] Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer
epigallocatechin-3-gallate and pterostilbene on pancreatic cancer growth
statistics. CA Cancer J Clin 2012;65(2015):87–108.
in vitro. J Surg Res 2012;177:255–62.
[6] Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer
[37] Toden S, Tran HM, Tovar-Camargo OA, Okugawa Y, Goel A. Epigallocatechin-
incidence and mortality worldwide: sources, methods and major patterns in
3-gallate targets cancer stem-like cells and enhances 5-fluorouracil
GLOBOCAN 2012. Int J Cancer 2015;136:E359–386.
chemosensitivity in colorectal cancer. Oncotarget 2016;7:16158–71.
[7] Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin 2016;66
[38] Wu H, Xin Y, Xu C, Xiao Y. Capecitabine combined with (-)-epigallocatechin-
(2016):7–30.
3-gallate inhibits angiogenesis and tumor growth in nude mice with gastric
[8] Moghadamtousi SZ, Fadaeinasab M, Nikzad S, Mohan G, Ali HM, Kadir HA.
cancer xenografts. Exp Ther Med 2012;3:650–4.
Annona muricata (Annonaceae): a review of its traditional uses, isolated
[39] Lee SH, Nam HJ, Kang HJ, Kwon HW, Lim YC. Epigallocatechin-3-gallate
acetogenins and biological activities. Int J Mol Sci 2015;16:15625–58.
attenuates head and neck cancer stem cell traits through suppression of
[9] Peter B, Bosze S, Horvath R. Biophysical characteristics of proteins and living
Notch pathway. Eur J Cancer 2013;49:3210–8.
cells exposed to the green tea polyphenol epigallocatechin-3-gallate (EGCg):
[40] Wu H, Xin Y, Xiao Y, Zhao J. Low-dose docetaxel combined with (-)-
review of recent advances from molecular mechanisms to nanomedicine and
epigallocatechin-3-gallate inhibits angiogenesis and tumor growth in nude
clinical trials. Eur Biophys J 2017;46:1–24.
mice with gastric cancer xenografts. Cancer Biother Radiopharm
[10] de Albuquerque UP, Soldati GT, Sieber SS, Ramos MA, de Sa JC, de Souza LC.
2012;27:204–9.
The use of plants in the medical system of the Fulni-o people (NE Brazil): a
[41] Stearns ME, Amatangelo MD, Varma D, Sell C, Goodyear SM. Combination
perspective on age and gender. J Ethnopharmacol 2011;133:866–73.
therapy with epigallocatechin-3-gallate and doxorubicin in human prostate
[11] Camargo EE, Bandeira MA, de Oliveira AG. Diagnosis of public programs
tumor modeling studies: inhibition of metastatic tumor growth in severe
focused on herbal medicines in Brazil. Nat Prod Commun 2011;6:1001–2.
combined immunodeficiency mice. Am J Pathol 2010;177:3169–79.
[12] Komlaga G, Agyare C, Dickson RA, Mensah ML, Annan K, Loiseau PM, et al.
[42] Zhang Y, Wang SX, Ma JW, Li HY, Ye JC, Xie SM, et al. EGCG inhibits properties
Medicinal plants and finished marketed herbal products used in the
of glioma stem-like cells and synergizes with temozolomide through
treatment of malaria in the Ashanti region, Ghana. J Ethnopharmacol
downregulation of P-glycoprotein inhibition. J Neurooncol 2015;121:41–52.
2015;172:333–46.
[43] Saldanha SN, Kala R, Tollefsbol TO. Molecular mechanisms for inhibition of
[13] Siddiqui IA, Adhami VM, Ahmad N, Mukhtar H. Nanochemoprevention:
colon cancer cells by combined epigenetic-modulating epigallocatechin
sustained release of bioactive food components for cancer prevention. Nutr
gallate and sodium butyrate. Exp Cell Res 2014;324:40–53.
Cancer 2010;62:883–90.
[44] Roomi MW, Ivanov V, Kalinovsky T, Niedzwiecki A, Rath M. Anti-tumor effect
[14] Chacko SM, Thambi PT, Kuttan R, Nishigaki I. Beneficial effects of green tea: a
of ascorbic acid, lysine, proline, arginine, and epigallocatechin gallate on
literature review. Chin Med 2010;5:13.
prostate cancer cell lines PC-3, LNCaP, and DU145. Res Commun Mol Pathol
[15] Vinson JA. Black and green tea and heart disease: a review. Biofactors
Pharmacol 2004;115–116:251–64.
2000;13:127–32.
[45] Zhou DH, Wang X, Yang M, Shi X, Huang W, Feng Q. Combination of low
[16] Snoussi C, Ducroc R, Hamdaoui MH, Dhaouadi K, Abaidi H, Cluzeaud F, et al.
concentration of (-)-epigallocatechin gallate (EGCG) and curcumin strongly
Green tea decoction improves glucose tolerance and reduces weight gain of
suppresses the growth of non-small cell lung cancer in vitro and in vivo
rats fed normal and high-fat diet. J Nutr Biochem 2014;25:557–64.
through causing cell cycle arrest. Int J Mol Sci 2013;14:12023–36.
[17] Sano M, Tabata M, Suzuki M, Degawa M, Miyase T, Maeda-Yamamoto M.
[46] Papi A, Farabegoli F, Iori R, Orlandi M, De Nicola GR, Bagatta M, et al. Vitexin-
Simultaneous determination of twelve tea catechins by high-performance
2-O-xyloside, raphasatin and (-)-epigallocatechin-3-gallate synergistically
liquid chromatography with electrochemical detection. Analyst
affect cell growth and apoptosis of colon cancer cells. Food Chem
2001;126:816–20.
2013;138:1521–30.
[18] Steinmann J, Buer J, Pietschmann T, Steinmann E. Anti-infective properties of
[47] Hagen RM, Chedea VS, Mintoff CP, Bowler E, Morse HR, Ladomery MR.
epigallocatechin-3-gallate (EGCG), a component of green tea. Br J Pharmacol
Epigallocatechin-3-gallate promotes apoptosis and expression of the caspase
2013;168:1059–73.
9a splice variant in PC3 prostate cancer cells. Int J Oncol 2013;43:194–200.
[19] Yang CS, Wang H. Cancer therapy combination: green tea and a
[48] Shankar S, Suthakar G, Srivastava RK. Epigallocatechin-3-gallate inhibits cell
phosphodiesterase 5 inhibitor? J Clin Invest 2013;123:556–8.
cycle and induces apoptosis in pancreatic cancer. Front Biosci
[20] Bailey HH, Mukhtar H. Green tea polyphenols and cancer chemoprevention of
2007;12:5039–51.
genitourinary cancer. Am Soc Clin Oncol Educ Book 2013:92–6.
[49] Sanna V, Singh CK, Jashari R, Adhami VM, Chamcheu JC, Rady I, et al. Targeted
[21] Khan N, Mukhtar H. Cancer and metastasis: prevention and treatment by
nanoparticles encapsulating (-)-epigallocatechin-3-gallate for prostate cancer
green tea. Cancer Metastasis Rev 2010;29:435–45.
prevention and therapy. Sci Rep 2017;7:41573.
[22] Siddiqui IA, Sanna V. Impact of nanotechnology on the delivery of natural
[50] Tait SW, Green DR. Mitochondria and cell death: outer membrane
products for cancer prevention and therapy. Mol Nutr Food Res
permeabilization and beyond. Nat Rev Mol Cell Biol 2010;11:621–32.
2016;60:1330–41.
[51] Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with
[23] Saleem M, Adhami VM, Siddiqui IA, Mukhtar H. Tea beverage in chemo-
wide-ranging implications in tissue kinetics. Br J Cancer 1972;26:239–57.
prevention of prostate cancer: a mini-review. Nutr Cancer 2003;47:13–23.
[52] Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the
[24] Nagle DG, Ferreira D, Zhou YD. Epigallocatechin-3-gallate (EGCG): chemical
cellular level. Nat Rev Mol Cell Biol 2008;9:231–41.
and biomedical perspectives. Phytochemistry 2006;67:1849–55.
[53] Xu ML, Hu J, Guo BP, Niu YR, Xiao C, Xu YX. Exploration of intrinsic and
[25] Adhami VM, Ahmad N, Mukhtar H. Molecular targets for green tea in prostate
extrinsic apoptotic pathways in zearalenone-treated rat sertoli cells. Environ
cancer prevention. J Nutr 2003;133:2417S–24S.
Toxicol 2015.
[26] Siddiqui IA, Adhami VM, Bharali DJ, Hafeez BB, Asim M, Khwaja SI, et al.
[54] Chalah A, Khosravi-Far R. The mitochondrial death pathway. Adv Exp Med
Introducing nanochemoprevention as a novel approach for cancer control:
Biol 2008;615:25–45.
proof of principle with green tea polyphenol epigallocatechin-3-gallate.
[55] Langlais C, Hughes MA, Cain K, MacFarlane M. In vitro assembly and analysis
Cancer Res 2009;69:1712–6.
of the apoptosome complex. Cold Spring Harb Protoc 2015;2015. pdb.
[27] Wagner H, Ulrich-Merzenich G. Synergy research: approaching a new
prot087080.
generation of phytopharmaceuticals. Phytomedicine 2009;16:97–110.
[56] Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E, Boise LH.
[28] Saldanha SN, Tollefsbol TO. The role of nutraceuticals in chemoprevention
Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic
and chemotherapy and their clinical outcomes. J Oncol 2012;2012:192464.
apoptosis. BMC Cell Biol 2013;14:32.
[29] Gan RY, Li HB, Sui ZQ, Corke H. Absorption, metabolism, anti-cancer effect
[57] Siegmund D, Mauri D, Peters N, Juo P, Thome M, Reichwein M, et al. Fas-
and molecular targets of epigallocatechin gallate (EGCG): An updated review.
associated death domain protein (FADD) and caspase-8 mediate up-
Crit Rev Food Sci Nutr 2016:1–18.
regulation of c-Fos by Fas ligand and tumor necrosis factor-related
[30] Rahman AA, Makpol S, Jamal R, Harun R, Mokhtar N, Ngah WZ. Tocotrienol-
apoptosis-inducing ligand (TRAIL) via a FLICE inhibitory protein (FLIP)-
rich fraction, [6]-gingerol and epigallocatechin gallate inhibit proliferation
regulated pathway. J Biol Chem 2001;276:32585–90.
and induce apoptosis of glioma cancer cells. Molecules 2014;19:14528–41.
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 21
[58] Kavanagh E. Quantification of active caspase-3 and active caspase-8 in [87] Gupta S, Ahmad N, Nieminen AL, Mukhtar H. Growth inhibition, cell-cycle
microglia cells. Methods Mol Biol 2013;1041:113–20. dysregulation, and induction of apoptosis by green tea constituent (-)-
[59] Pan LL, Wang AY, Huang YQ, Luo Y, Ling M. Mangiferin induces apoptosis by epigallocatechin-3-gallate in androgen-sensitive and androgen-insensitive
regulating Bcl-2 and Bax expression in the CNE2 nasopharyngeal carcinoma human prostate carcinoma cells. Toxicol Appl Pharmacol 2000;164:82–90.
cell line. Asian Pac J Cancer Prev 2014;15:7065–8. [88] Qiao Y, Cao J, Xie L, Shi X. Cell growth inhibition and gene expression
[60] Xu W, Jing L, Wang Q, Lin CC, Chen X, Diao J, et al. Bax-PGAM5L-Drp1 complex regulation by (-)-epigallocatechin-3-gallate in human cervical cancer cells.
is required for intrinsic apoptosis execution. Oncotarget 2015;6:30017–34. Arch Pharm Res 2009;32:1309–15.
[61] Chang KT, Anishkin A, Patwardhan GA, Beverly LJ, Siskind LJ, Colombini M. [89] Zhang X, Min KW, Wimalasena J, Baek SJ. Cyclin D1 degradation and p21
Ceramide channels: destabilization by Bcl-xL and role in apoptosis. Biochim induction contribute to growth inhibition of colorectal cancer cells induced
Biophys Acta 1848;2015:2374–84. by epigallocatechin-3-gallate. J Cancer Res Clin Oncol 2012;138:2051–60.
[62] DeOcesano-Pereira C, Amaral MS, Parreira KS, Ayupe AC, Jacysyn JF, [90] Ahmad N, Adhami VM, Gupta S, Cheng P, Mukhtar H. Role of the
Amarante-Mendes GP, et al. Long non-coding RNA INXS is a critical retinoblastoma (pRb)-E2F/DP pathway in cancer chemopreventive effects of
mediator of BCL-XS induced apoptosis. Nucleic Acids Res 2014;42:8343–55. green tea polyphenol epigallocatechin-3-gallate. Arch Biochem Biophys
[63] Yu SY, Qi R. Role of bad in podocyte apoptosis induced by puromycin 2002;398:125–31.
aminonucleoside. Transplant Proc 2013;45:569–73. [91] Bhatia N, Agarwal C, Agarwal R. Differential responses of skin cancer-
[64] Ma S, Hockings C, Anwari K, Kratina T, Fennell S, Lazarou M, et al. Assembly of chemopreventive agents silibinin, quercetin, and epigallocatechin 3-gallate
the Bak apoptotic pore: a critical role for the Bak protein alpha6 helix in the on mitogenic signaling and cell cycle regulators in human epidermoid
multimerization of homodimers during apoptosis. J Biol Chem carcinoma A431 cells. Nutr Cancer 2001;39:292–9.
2013;288:26027–38. [92] Folkman J. Angiogenesis. Annu Rev Med 2006;57:1–18.
[65] Gao C, Guo WJ, Wang AY. Significance of decreased apoptosis, role of Bid [93] Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature
expression and Bcl-Xl/Bid ratio in human jejunal stromal tumors. APMIS 2000;407:249–57.
2012;120:171–81. [94] Birbrair A, Zhang T, Wang ZM, Messi ML, Olson JD, Mintz A, et al. Type-2
[66] Muthalagu N, Junttila MR, Wiese KE, Wolf E, Morton J, Bauer B, et al. BIM is pericytes participate in normal and tumoral angiogenesis. Am J Physiol Cell
the primary mediator of MYC-induced apoptosis in multiple solid tissues. Cell Physiol 2014;307:C25–38.
Rep 2014;8:1347–53. [95] Liekens S, De Clercq E, Neyts J. Angiogenesis: regulators and clinical
[67] Chen C, Qincao L, Xu J, Du S, Huang E, Liu C, et al. Role of PUMA in applications. Biochem Pharmacol 2001;61:253–70.
methamphetamine-induced neuronal apoptosis. Toxicol Lett [96] Proskuryakov SY, Gabai VL. Mechanisms of tumor cell necrosis. Curr Pharm
2016;240:149–60. Des 2010;16:56–68.
[68] Mukhopadhyay S, Panda PK, Sinha N, Das DN, Bhutia SK. Autophagy and [97] Bredholt G, Mannelqvist M, Stefansson IM, Birkeland E, Bo TH, Oyan AM, et al.
apoptosis: where do they meet? Apoptosis 2014;19:555–66. Tumor necrosis is an important hallmark of aggressive endometrial cancer
[69] He H, Zang LH, Feng YS, Chen LX, Kang N, Tashiro S, et al. Physalin A induces and associates with hypoxia, angiogenesis and inflammation responses.
apoptosis via p53-Noxa-mediated ROS generation, and autophagy plays a Oncotarget 2015;6:39676–91.
protective role against apoptosis through p38-NF-kappaB survival pathway [98] Stieler K, Schumacher U, Horst AK, Fischer N. XMRV induces cell migration,
in A375–S2 cells. J Ethnopharmacol 2013;148:544–55. cytokine expression and tumor angiogenesis: are 22Rv1 cells a suitable
[70] Zhang Y, Hao Y, Sun Q, Peng C. Role of Smac in apoptosis of lung cancer cells prostate cancer model? PLoS One 2012;7:e42321.
A549 induced by Taxol. Clin Lab 2015;61:17–21. [99] Matsuoka T, Yashiro M. Rho/ROCK signaling in motility and metastasis of
[71] Hahm ER, Lee J, Singh SV. Role of mitogen-activated protein kinases and Mcl- gastric cancer. World J Gastroenterol 2014;20:13756–66.
1 in apoptosis induction by withaferin A in human breast cancer cells. Mol [100] Zhou H, Zhang B, Zheng J, Yu M, Zhou T, Zhao K, et al. The inhibition of
Carcinog 2014;53:907–16. migration and invasion of cancer cells by graphene via the impairment of
[72] Safa AR. C-FLIP, a master anti-apoptotic regulator. Exp Oncol mitochondrial respiration. Biomaterials 2014;35:1597–607.
2012;34:176–84. [101] Relat J, Blancafort A, Oliveras G, Cufi S, Haro D, Marrero PF, et al. Different
[73] Tang Y, Zhao DY, Elliott S, Zhao W, Curiel TJ, Beckman BS, et al. fatty acid metabolism effects of (-)-epigallocatechin-3-gallate and C75 in
Epigallocatechin-3 gallate induces growth inhibition and apoptosis in adenocarcinoma lung cancer. BMC Cancer 2012;12:280.
human breast cancer cells through survivin suppression. Int J Oncol [102] Sakamoto Y, Terashita N, Muraguchi T, Fukusato T, Kubota S. Effects of
2007;31:705–11. epigallocatechin-3-gallate (EGCG) on A549 lung cancer tumor growth and
[74] Basu A, Haldar S. Combinatorial effect of epigallocatechin-3-gallate and TRAIL angiogenesis. Biosci Biotechnol Biochem 2013;77:1799–803.
on pancreatic cancer cell death. Int J Oncol 2009;34:281–6. [103] Li X, Feng Y, Liu J, Feng X, Zhou K, Tang X. Epigallocatechin-3-gallate inhibits
[75] Thangapazham RL, Passi N, Maheshwari RK. Green tea polyphenol and IGF-I-stimulated lung cancer angiogenesis through downregulation of
epigallocatechin gallate induce apoptosis and inhibit invasion in human HIF-1alpha and VEGF expression. J Nutrigenet Nutrigenomics 2013;6:
breast cancer cells. Cancer Biol Ther 2007;6:1938–43. 169–78.
[76] Wu PP, Kuo SC, Huang WW, Yang JS, Lai KC, Chen HJ, et al. (-)- [104] He L, Zhang E, Shi J, Li X, Zhou K, Zhang Q, et al. (-)-Epigallocatechin-3-gallate
Epigallocatechin gallate induced apoptosis in human adrenal cancer NCI- inhibits human papillomavirus (HPV)-16 oncoprotein-induced angiogenesis
H295 cells through caspase-dependent and caspase-independent pathway. in non-small cell lung cancer cells by targeting HIF-1alpha. Cancer
Anticancer Res 2009;29:1435–42. Chemother Pharmacol 2013;71:713–25.
[77] Hu Q, Chang X, Yan R, Rong C, Yang C, Cheng S, et al. (-)-Epigallocatechin-3- [105] Shi J, Liu F, Zhang W, Liu X, Lin B, Tang X. Epigallocatechin-3-gallate inhibits
gallate induces cancer cell apoptosis via acetylation of amyloid precursor nicotine-induced migration and invasion by the suppression of angiogenesis
protein. Med Oncol 2015;32:390. and epithelial-mesenchymal transition in non-small cell lung cancer cells.
[78] Onoda C, Kuribayashi K, Nirasawa S, Tsuji N, Tanaka M, Kobayashi D, et al. (-)- Oncol Rep 2015;33:2972–80.
Epigallocatechin-3-gallate induces apoptosis in gastric cancer cell lines by [106] Takahashi A, Watanabe T, Mondal A, Suzuki K, Kurusu-Kanno M, Li Z, et al.
down-regulating survivin expression. Int J Oncol 2011;38:1403–8. Mechanism-based inhibition of cancer metastasis with (-)-epigallocatechin
[79] Landis-Piwowar KR, Huo CD, Chen D, Milacic V, Shi GQ, Chan TH, et al. A novel gallate. Biochem Biophys Res Commun 2014;443:1–6.
prodrug of the green tea polyphenol (-)-epigallocatechin-3-gallate as a [107] Yin Z, Henry EC, Gasiewicz TA. (-)-Epigallocatechin-3-gallate is a novel Hsp90
potential anticancer agent. Cancer Research 2007;67:4303–10. inhibitor. Biochemistry 2009;48:336–45.
[80] Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna [108] Belguise K, Guo S, Sonenshein GE. Activation of FOXO3a by the green tea
J, et al. Sustained proliferation in cancer: Mechanisms and novel therapeutic polyphenol epigallocatechin-3-gallate induces estrogen receptor alpha
targets. Semin Cancer Biol 2015;35(Suppl):S25–54. expression reversing invasive phenotype of breast cancer cells. Cancer Res
[81] Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. 2007;67:5763–70.
Development 2013;140:3079–93. [109] Zhang Y, Han G, Fan B, Zhou Y, Zhou X, Wei L, et al. Green tea (-)-
[82] Suzuki A, Hayashida M, Ito T, Kawano H, Nakano T, Miura M, et al. Survivin epigallocatechin-3-gallate down-regulates VASP expression and inhibits
initiates cell cycle entry by the competitive interaction with Cdk4/p16 breast cancer cell migration and invasion by attenuating Rac1 activity. Eur
(INK4a) and Cdk2/cyclin E complex activation. Oncogene 2000;19:3225–34. J Pharmacol 2009;606:172–9.
[83] Di Agostino S, Strano S, Emiliozzi V, Zerbini V, Mottolese M, Sacchi A, et al. [110] Sen T, Chatterjee A. Epigallocatechin-3-gallate (EGCG) downregulates EGF-
Gain of function of mutant p53: the mutant p53/NF-Y protein complex induced MMP-9 in breast cancer cells: involvement of integrin receptor
reveals an aberrant transcriptional mechanism of cell cycle regulation. Cancer alpha5beta1 in the process. Eur J Nutr 2011;50:465–78.
Cell 2006;10:191–202. [111] Chen PN, Chu SC, Kuo WH, Chou MY, Lin JK, Hsieh YS. Epigallocatechin-3
[84] Huang CH, Tsai SJ, Wang YJ, Pan MH, Kao JY, Way TD. EGCG inhibits protein gallate inhibits invasion, epithelial-mesenchymal transition, and tumor
synthesis, lipogenesis, and cell cycle progression through activation of AMPK growth in oral cancer cells. J Agric Food Chem 2011;59:3836–44.
in p53 positive and negative human hepatoma cells. Mol Nutr Food Res [112] Koh YW, Choi EC, Kang SU, Hwang HS, Lee MH, Pyun J, et al. Green tea (-)-
2009;53:1156–65. epigallocatechin-3-gallate inhibits HGF-induced progression in oral cavity
[85] Sharma C, Nusri Qel A, Begum S, Javed E, Rizvi TA, Hussain A. (-)- cancer through suppression of HGF/c-Met. J Nutr Biochem 2011;22:1074–83.
Epigallocatechin-3-gallate induces apoptosis and inhibits invasion and [113] Hwang YS, Park KK, Chung WY. Epigallocatechin-3 gallate inhibits cancer
migration of human cervical cancer cells. Asian Pac J Cancer Prev invasion by repressing functional invadopodia formation in oral squamous
2012;13:4815–22. cell carcinoma. Eur J Pharmacol 2013;715:286–95.
[86] Ma YC, Li C, Gao F, Xu Y, Jiang ZB, Liu JX, et al. Epigallocatechin gallate inhibits [114] Zhu BH, Chen HY, Zhan WH, Wang CY, Cai SR, Wang Z, et al. (-)-
the growth of human lung cancer by directly targeting the EGFR signaling Epigallocatechin-3-gallate inhibits VEGF expression induced by IL-6 via
pathway. Oncol Rep 2014;31:1343–9. Stat3 in gastric cancer. World J Gastroenterol 2011;17:2315–25.
22 I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23
[115] Zhu BH, Zhan WH, Li ZR, Wang Z, He YL, Peng JS, et al. (-)-Epigallocatechin-3- [142] Shafiei SS, Solati-Hashjin M, Samadikuchaksaraei A, Kalantarinejad R, Asadi-
gallate inhibits growth of gastric cancer by reducing VEGF production and Eydivand M, Abu Osman NA. pigallocatechin gallate/layered double
angiogenesis. World J Gastroenterol 2007;13:1162–9. hydroxide nanohybrids: preparation, characterization, and in vitro anti-
[116] Shimizu M, Shirakami Y, Sakai H, Yasuda Y, Kubota M, Adachi S, et al. (-)- tumor study. PLoS One 2015;10:e0136530.
Epigallocatechin gallate inhibits growth and activation of the VEGF/VEGFR [143] Khan N, Bharali DJ, Adhami VM, Siddiqui IA, Cui H, Shabana SM, et al. Oral
axis in human colorectal cancer cells. Chem Biol Interact 2010;185:247–52. administration of naturally occurring chitosan-based nanoformulated green
[117] Zhou F, Zhou H, Wang T, Mu Y, Wu B, Guo DL, et al. Epigallocatechin-3-gallate tea polyphenol EGCG effectively inhibits prostate cancer cell growth in a
inhibits proliferation and migration of human colon cancer SW620 cells xenograft model. Carcinogenesis 2014;35:415–23.
in vitro. Acta Pharmacol Sin 2012;33:120–6. [144] Siddiqui IA, Bharali DJ, Nihal M, Adhami VM, Khan N, Chamcheu JC, et al.
[118] Maruyama T, Murata S, Nakayama K, Sano N, Ogawa K, Nowatari T, et al. (-)- Excellent anti-proliferative and pro-apoptotic effects of (-)-epigallocatechin-
Epigallocatechin-3-gallate suppresses liver metastasis of human colorectal 3-gallate encapsulated in chitosan nanoparticles on human melanoma cell
cancer. Oncol Rep 2014;31:625–33. growth both in vitro and in vivo. Nanomedicine 2014;10:1619–26.
[119] Larsen CA, Dashwood RH. (-)-Epigallocatechin-3-gallate inhibits Met [145] Ray L, Kumar P, Gupta KC. The activity against Ehrlich’s ascites tumors of
signaling, proliferation, and invasiveness in human colon cancer cells. Arch doxorubicin contained in self assembled, cell receptor targeted nanoparticle
Biochem Biophys 2010;501:52–7. with simultaneous oral delivery of the green tea polyphenol
[120] Larsen CA, Dashwood RH. Suppression of Met activation in human colon epigallocatechin-3-gallate. Biomaterials 2013;34:3064–76.
cancer cells treated with (-)-epigallocatechin-3-gallate: minor role of [146] Ranzato E, Magnelli V, Martinotti S, Waheed Z, Cain SM, Snutch TP, et al.
hydrogen peroxide. Biochem Biophys Res Commun 2009;389:527–30. Epigallocatechin-3-gallate elicits Ca2+ spike in MCF-7 breast cancer cells:
[121] Qin J, Wang Y, Bai Y, Yang K, Mao Q, Lin Y, et al. Epigallocatechin-3-gallate essential role of Cav3.2 channels. Cell Calcium 2014;56:285–95.
inhibits bladder cancer cell invasion via suppression of NF-kappaBmediated [147] Gupta S, Ahmad N, Mohan RR, Husain MM, Mukhtar H. Prostate cancer
matrix metalloproteinase-9 expression. Mol Med Rep 2012;6:1040–4. chemoprevention by green tea: in vitro and in vivo inhibition of testosterone-
[122] Duhon D, Bigelow RL, Coleman DT, Steffan JJ, Yu C, Langston W, et al. The mediated induction of ornithine decarboxylase. Cancer Res
polyphenol epigallocatechin-3-gallate affects lipid rafts to block activation of 1999;59:2115–20.
the c-Met receptor in prostate cancer cells. Mol Carcinog 2010;49:739–49. [148] Fu H, He J, Mei F, Zhang Q, Hara Y, Ryota S, et al. Lung cancer inhibitory effect
[123] Lee SC, Chan WK, Lee TW, Lam WH, Wang X, Chan TH, et al. Effect of a of epigallocatechin-3-gallate is dependent on its presence in a complex
prodrug of the green tea polyphenol (-)-epigallocatechin-3-gallate on the mixture (polyphenon E). Cancer Prev Res (Phila) 2009;2:531–7.
growth of androgen-independent prostate cancer in vivo. Nutr Cancer [149] Kim J, Zhang X, Rieger-Christ KM, Summerhayes IC, Wazer DE, Paulson KE,
2008;60:483–91. et al. Suppression of Wnt signaling by the green tea compound (-)-
[124] Moses MA, Henry EC, Ricke WA, Gasiewicz TA. The heat shock protein 90 epigallocatechin 3-gallate (EGCG) in invasive breast cancer cells.
inhibitor, (-)-epigallocatechin gallate, has anticancer activity in a novel Requirement of the transcriptional repressor HBP1. J Biol Chem
human prostate cancer progression model. Cancer Prev Res (Phila) 2006;281:10865–75.
2015;8:249–57. [150] Meeran SM, Patel SN, Chan TH, Tollefsbol TO. A novel prodrug of
[125] Wang S, Chen R, Zhong Z, Shi Z, Chen M, Wang Y. Epigallocatechin-3-gallate epigallocatechin-3-gallate: differential epigenetic hTERT repression in
potentiates the effect of curcumin in inducing growth inhibition and human breast cancer cells. Cancer Prev Res (Phila) 2011;4:1243–54.
apoptosis of resistant breast cancer cells. Am J Chin Med 2014;42:1279–300. [151] Moreira L, Araujo I, Costa T, Correia-Branco A, Faria A, et al. Quercetin and
[126] Lambert JD, Sang S, Yang CS. N-Acetylcysteine enhances the lung cancer epigallocatechin gallate inhibit glucose uptake and metabolism by breast
inhibitory effect of epigallocatechin-3-gallate and forms a new adduct. Free cancer cells by an estrogen receptor-independent mechanism. Exp Cell Res
Radic Biol Med 2008;44:1069–74. 2013;319:1784–95.
[127] Gupta S, Hastak K, Ahmad N, Lewin JS, Mukhtar H. Inhibition of prostate [152] Shutava TG, Balkundi SS, Vangala P, Steffan JJ, Bigelow RL, Cardelli JA, et al.
carcinogenesis in TRAMP mice by oral infusion of green tea polyphenols. Proc Layer-by-layer-coated gelatin nanoparticles as a vehicle for delivery of
Natl Acad Sci U S A 2001;98:10350–5. natural polyphenols. ACS Nano 2009;3:1877–85.
[128] D’Angelo L, Piazzi G, Pacilli A, Prossomariti A, Fazio C, Montanaro L, et al. A [153] Tu SH, Ku CY, Ho CT, Chen CS, Huang CS, Lee CH, et al. Tea polyphenol (-)-
combination of eicosapentaenoic acid-free fatty acid, epigallocatechin-3- epigallocatechin-3-gallate inhibits nicotine- and estrogen-induced alpha9-
gallate and proanthocyanidins has a strong effect on mTOR signaling in nicotinic acetylcholine receptor upregulation in human breast cancer cells.
colorectal cancer cells. Carcinogenesis 2014;35:2314–20. Mol Nutr Food Res 2011;55:455–66.
[129] Kale A, Gawande S, Kotwal S, Netke S, Roomi W, Ivanov V, et al. Studies on the [154] De Amicis F, Russo A, Avena P, Santoro M, Vivacqua A, Bonofiglio D, et al. In
effects of oral administration of nutrient mixture, quercetin and red onions vitro mechanism for downregulation of ER-alpha expression by
on the bioavailability of epigallocatechin gallate from green tea extract. epigallocatechin gallate in ER+/PR+ human breast cancer cells. Mol Nutr
Phytother Res 2010;24(Suppl 1):S48–55. Food Res 2013;57:840–53.
[130] Dawood S, Austin L, Cristofanilli M. Cancer stem cells: implications for cancer [155] Braicu C, Gherman CD, Irimie A, Berindan-Neagoe I. Epigallocatechin-3-
therapy. Oncology 2014;28:1101–7 [1110]. Gallate (EGCG) inhibits cell proliferation and migratory behaviour of triple
[131] Ajani JA, Song S, Hochster HS, Steinberg IB. Cancer stem cells: the promise negative breast cancer cells. J Nanosci Nanotechnol 2013;13:632–7.
and the potential. Semin Oncol 2015;42(Suppl 1):S3–17. [156] Pianetti S, Guo S, Kavanagh KT, Sonenshein GE. Green tea polyphenol
[132] Tang SN, Singh C, Nall D, Meeker D, Shankar S, Srivastava RK. The dietary epigallocatechin-3 gallate inhibits Her-2/neu signaling, proliferation, and
bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to transformed phenotype of breast cancer cells. Cancer Res 2002;62:652–5.
inhibit prostate cancer stem cell characteristics, invasion, migration and [157] Jang JY, Lee JK, Jeon YK, Kim CW. Exosome derived from epigallocatechin
epithelial-mesenchymal transition. J Mol Signal 2010;5:14. gallate treated breast cancer cells suppresses tumor growth by inhibiting
[133] Mineva ND, Paulson KE, Naber SP, Yee AS, Sonenshein GE. Epigallocatechin-3- tumor-associated macrophage infiltration and M2 polarization. BMC Cancer
gallate inhibits stem-like inflammatory breast cancer cells. PLoS One 2013;8: 2013;13:421.
e73464. [158] Pan X, Zhao B, Song Z, Han S, Wang M. Estrogen receptor-alpha36 is involved
[134] Nishimura N, Hartomo TB, Pham TV, Lee MJ, Yamamoto T, Morikawa S, in epigallocatechin-3-gallate induced growth inhibition of ER-negative breast
et alTakeshima Y, Iijima K, Matsuo M, Nishio H. Epigallocatechin gallate cancer stem/progenitor cells. J Pharmacol Sci 2016;130:85–93.
inhibits sphere formation of neuroblastoma BE(2)-C cells. Environ Health [159] Jin L, Li C, Xu Y, Wang L, Liu J, Wang D, et al. Epigallocatechin gallate promotes
Prev Med 2012;17:246–51. p53 accumulation and activity via the inhibition of MDM2-mediated p53
[135] Eom DW, Lee JH, Kim YJ, Hwang GS, Kim SN, Kwak JH, et al. Synergistic effect ubiquitination in human lung cancer cells. Oncol Rep 2013;29:1983–90.
of curcumin on epigallocatechin gallate-induced anticancer action in PC3 [160] Kim KC, Lee C. Reversal of Cisplatin resistance by epigallocatechin gallate is
prostate cancer cells. BMB Rep 2015;48:461–6. mediated by downregulation of axl and tyro 3 expression in human lung
[136] Wei DZ, Yang JY, Liu JW, Tong WY. Inhibition of liver cancer cell proliferation cancer cells. Korean J Physiol Pharmacol 2014;18:61–6.
and migration by a combination of (-)-epigallocatechin-3-gallate and [161] Sonoda JI, Ikeda R, Baba Y, Narumi K, Kawachi A, Tomishige E, et al. Green tea
ascorbic acid. J Chemother 2003;15:591–5. catechin, epigallocatechin-3-gallate, attenuates the cell viability of human
[137] Roomi MW, Ivanov V, Kalinovsky T, Niedzwiecki A, Rath M. Antitumor effect non-small-cell lung cancer A549 cells via reducing Bcl-xL expression. Exp
of ascorbic acid, lysine, proline, arginine, and green tea extract on bladder Ther Med 2014;8:59–63.
cancer cell line T-24. Int J Urol 2006;13:415–9. [162] Wang H, Bian S, Yang CS. Green tea polyphenol EGCG suppresses lung cancer
[138] Li YJ, Wu SL, Lu SM, Chen F, Guo Y, Gan SM, et al. (-)-Epigallocatechin-3- cell growth through upregulating miR-210 expression caused by stabilizing
gallate inhibits nasopharyngeal cancer stem cell self-renewal and migration HIF-1alpha. Carcinogenesis 2011;32:1881–9.
and reverses the epithelial-mesenchymal transition via NF-kappaB p65 [163] Li GX, Chen YK, Hou Z, Xiao H, Jin H, Lu G, et al. Pro-oxidative activities and
inactivation. Tumour Biol 2015;36:2747–61. dose-response relationship of (-)-epigallocatechin-3-gallate in the inhibition
[139] Siddiqui IA, Mukhtar H. Nanochemoprevention by bioactive food of lung cancer cell growth: a comparative study in vivo and in vitro.
components: a perspective. Pharm Res 2010;27:1054–60. Carcinogenesis 2010;31:902–10.
[140] Siddiqui IA, Adhami VM, Chamcheu JC, Mukhtar H. Impact of nanotechnology [164] Sadava D, Whitlock E, Kane SE. The green tea polyphenol, epigallocatechin-3-
in cancer: emphasis on nanochemoprevention. Int J Nanomedicine gallate inhibits telomerase and induces apoptosis in drug-resistant lung
2012;7:591–605. cancer cells. Biochem Biophys Res Commun 2007;360:233–7.
[141] Narayanan S, Mony U, Vijaykumar DK, Koyakutty M, Paul-Prasanth B, Menon [165] Li Y, Zhang T, Jiang Y, Lee HF, Schwartz SJ, Sun D. (-)-Epigallocatechin-3-
D. Sequential release of epigallocatechin gallate and paclitaxel from PLGA- gallate inhibits Hsp90 function by impairing Hsp90 association with
casein core/shell nanoparticles sensitizes drug-resistant breast cancer cells. cochaperones in pancreatic cancer cell line Mia Paca-2. Mol Pharm
Nanomedicine 2015;11:1399–406. 2009;6:1152–9.
I. Rady et al. / Egyptian Journal of Basic and Applied Sciences 5 (2018) 1–23 23
[166] Rao SD, Pagidas K. Epigallocatechin-3-gallate, a natural polyphenol, inhibits epigallocatechin-3-gallate in different progression stages of LNCaP prostate
cell proliferation and induces apoptosis in human ovarian cancer cells. cancer cells. Cancer Lett 2009;275:86–92.
Anticancer Res 2010;30:2519–23. [179] Harper CE, Patel BB, Wang J, Eltoum IA, Lamartiniere CA. Epigallocatechin-3-
[167] Yan C, Yang J, Shen L, Chen X. Inhibitory effect of Epigallocatechin gallate on Gallate suppresses early stage, but not late stage prostate cancer in TRAMP
ovarian cancer cell proliferation associated with aquaporin 5 expression. mice: mechanisms of action. Prostate 2007;67:1576–89.
Arch Gynecol Obstet 2012;285:459–67. [180] Li J, Wu S, Wu C, Qiu L, Zhu G, Cui C, et al. Versatile surface engineering of
[168] Cheng CW, Shieh PC, Lin YC, Chen YJ, Lin YH, Kuo DH, et al. Indoleamine 2,3- porous nanomaterials with bioinspired polyphenol coatings for targeted and
dioxygenase, an immunomodulatory protein, is suppressed by (-)- controlled drug delivery. Nanoscale 2016;8:8600–6.
epigallocatechin-3-gallate via blocking of gamma-interferon-induced JAK- [181] Min NY, Kim JH, Choi JH, Liang W, Ko YJ, Rhee S, et al. Selective death of
PKC-delta-STAT1 signaling in human oral cancer cells. J Agric Food Chem cancer cells by preferential induction of reactive oxygen species in response
2010;58:887–94. to (-)-epigallocatechin-3-gallate. Biochem Biophys Res Commun
[169] Irimie AI, Braicu C, Zanoaga O, Pileczki V, Gherman C, Berindan-Neagoe I, 2012;421:91–7.
et al. Epigallocatechin-3-gallate suppresses cell proliferation and promotes [182] Jin H, Gong W, Zhang C, Wang S. Epigallocatechin gallate inhibits the
apoptosis and autophagy in oral cancer SSC-4 cells. Onco Targets Ther proliferation of colorectal cancer cells by regulating Notch signaling. Onco
2015;8:461–70. Targets Ther 2013;6:145–53.
[170] Lin CH, Chao LK, Hung PH, Chen YJ. EGCG inhibits the growth and [183] Umeda D, Yano S, Yamada K, Tachibana H. Involvement of 67-kDa laminin
tumorigenicity of nasopharyngeal tumor-initiating cells through receptor-mediated myosin phosphatase activation in antiproliferative effect
attenuation of STAT3 activation. Int J Clin Exp Pathol 2014;7:2372–81. of epigallocatechin-3-O-gallate at a physiological concentration on Caco-2
[171] Ma J, Shi M, Li G, Wang N, Wei J, Wang T, et al. Regulation of Id1 expression by colon cancer cells. Biochem Biophys Res Commun 2008;371:172–6.
epigallocatechin3gallate and its effect on the proliferation and apoptosis of [184] Hu B, Ting Y, Yang X, Tang W, Zeng X, Huang Q. Nanochemoprevention by
poorly differentiated AGS gastric cancer cells. Int J Oncol 2013;43:1052–8. encapsulation of (-)-epigallocatechin-3-gallate with bioactive
[172] Tanaka T, Ishii T, Mizuno D, Mori T, Yamaji R, Nakamura Y, et al. (-)- peptides/chitosan nanoparticles for enhancement of its bioavailability.
Epigallocatechin-3-gallate suppresses growth of AZ521 human gastric cancer Chem Commun (Camb) 2012;48:2421–3.
cells by targeting the DEAD-box RNA helicase p68. Free Radic Biol Med [185] Yuan JH, Li YQ, Yang XY. Inhibition of epigallocatechin gallate on orthotopic
2011;50:1324–35. colon cancer by upregulating the Nrf2-UGT1A signal pathway in nude mice.
[173] Zhang Y, Yang ND, Zhou F, Shen T, Duan T, Zhou J, et al. (-)-Epigallocatechin- Pharmacology 2007;80:269–78.
3-gallate induces non-apoptotic cell death in human cancer cells via ROS- [186] Ogawa K, Hara T, Shimizu M, Nagano J, Ohno T, Hoshi M, et al. (-)-
mediated lysosomal membrane permeabilization. PLoS One 2012;7:e46749. Epigallocatechin gallate inhibits the expression of indoleamine 2,3-
[174] Wubetu GY, Shimada M, Morine Y, Ikemoto T, Ishikawa D, Iwahashi S, et al. dioxygenase in human colorectal cancer cells. Oncol Lett 2012;4:546–50.
Epigallocatechin gallate hinders human hepatoma and colon cancer sphere [187] Alotaibi A, Bhatnagar P, Najafzadeh M, Gupta KC, Anderson D. Tea phenols in
formation. J Gastroenterol Hepatol 2016;31:256–64. bulk and nanoparticle form modify DNA damage in human lymphocytes from
[175] Albrecht DS, Clubbs EA, Ferruzzi M, Bomser JA. Epigallocatechin-3-gallate colon cancer patients and healthy individuals treated in vitro with platinum-
(EGCG) inhibits PC-3 prostate cancer cell proliferation via MEK-independent based chemotherapeutic drugs. Nanomedicine (Lond) 2013;8:389–401.
ERK1/2 activation. Chem Biol Interact 2008;171:89–95. [188] Singh T, Katiyar SK. Green tea polyphenol, (-)-epigallocatechin-3-gallate,
[176] Shukla R, Chanda N, Zambre A, Upendran A, Katti K, Kulkarni RR, et al. induces toxicity in human skin cancer cells by targeting beta-catenin
Laminin receptor specific therapeutic gold nanoparticles (198AuNP-EGCg) signaling. Toxicol Appl Pharmacol 2013;273:418–24.
show efficacy in treating prostate cancer. Proc Natl Acad Sci U S A [189] Balasubramanian S, Adhikary G, Eckert RL. The Bmi-1 polycomb protein
2012;109:12426–31. antagonizes the (-)-epigallocatechin-3-gallate-dependent suppression of skin
[177] Wang SI, Mukhtar H. Gene expression profile in human prostate LNCaP cancer cell survival. Carcinogenesis 2010;31:496–503.
cancer cells by (-) epigallocatechin-3-gallate. Cancer Lett 2002;182:43–51. [190] Mukhtar H, Matsui MS, Maes D, Marenus K, Tubesing K, Elmets CA.
[178] Chuu CP, Chen RY, Kokontis JM, Hiipakka RA, Liao S. Suppression of androgen Prevention by green tea polyphenols against ultraviolet-induced erythema
receptor signaling and prostate specific antigen expression by (-)- in humans. J Invest Dermatol 1996;106:243.