Molecules 23 02334
Molecules 23 02334
Molecules 23 02334
Review
Suppressive Effects of EGCG on Cervical Cancer
Ying-Qi Wang, Jian-Liang Lu, Yue-Rong Liang and Qing-Sheng Li *
Tea Research Institute, Zhejiang University, # 866 Yuhangtang Road, Hangzhou 310058, China;
[email protected] (Y.-Q.W.); [email protected] (J.-L.L.); [email protected] (Y.-R.L.)
* Correspondence: [email protected]; Tel.: +86-571-8898-2704
Abstract: Cervical cancer is the fourth most common gynecological cancer worldwide. Although
prophylactic vaccination presents the most effective method for cervical cancer prevention,
chemotherapy is still the primary invasive intervention. It is urgent to exploit low-toxic natural
anticancer drugs on account of high cytotoxicity and side-effects of conventional agents. As a natural
product, (-)-epigallocatechingallate (EGCG) has abilities in anti-proliferation, anti-metastasis and
pro-apoptosis of cervical cancer cells. Moreover, EGCG also has pharmaceutical synergistic effects
with conventional agents such as cisplatin (CDDP) and bleomycin (BLM). The underlying mechanisms
of EGCG suppressive effects on cervical cancer are reviewed in this article. Further research directions
and ambiguous results are also discussed.
Keywords: Camellia sinensis; EGCG; cervical cancer; human papillomavirus (HPV); anticancer
1. Introduction
Cervical cancer is the fourth most common female cancer in terms of both incidence and mortality
rates worldwide, with an estimated 528,000 new cases in 2012, including 266,000 deaths (85% occurring
in developing countries) [1]. Although screening methods and early detection programs have been
established, invasive cervical cancer still represents a major concern for public health. Risk factors
of cervical cancer include human papillomavirus (HPV), sexual behavior beginning at a young age
(<16 years old), multiple sexual partners (more than four), history of genital warts, HIV positive,
and cigarette smoking or environmental tobacco smoke [2]. More than 99% of cervical cancer patients
carry at least one genotype of oncogenic HPV [3], since persistent infection with HPV is the prominent
etiological reason in the formation of cervical cancer [4]. However, more than 200 types of identified
HPVs can be classified as low-risk HPVs and high-risk HPVs [5]. Low-risk HPVs induce inconspicuous
infection or benign papilloma which could eventually be resolved by the immune system and rarely
cause neoplasia and carcinogenesis [6]. On the contrary, high-risk HPVs are related to the propensity of
malignant progression of virus-mediated lesions [7,8]. Among them, HPV 16 and HPV18 are the two
major viruses responsible for approximately 70% of all cervical carcinomas worldwide. HPV has two
vital transcriptional units, E6 and E7, that encoded oncoproteins primarily attribute to its oncogenic
function [9]. E6 protein inhibits the activity of tumor suppressor P53, and E7 protein targets other tumor
suppressors of the retinoblastoma family [10,11]. A series of human cervical cancer cell lines have been
used to study the potential anticancer ability of chemo therapeutic agents, including HPV18-positive
HeLa cell lines, HPV16-positive CaSki and SiHa cell lines, etc. Similar to other cancers, cervical
cancer harms the human body mainly due to the proliferation and metastasis of cancer cells. Current
treatments in curing cancers aim at anti-proliferation, anti-metastasis of cancer cells, and inducing
cancer cell apoptosis.
Until now, prophylactic vaccination is the primarily effective prevention strategy for cervical
malignancies [12]. Although these vaccines could prevent approximately 90% of cervical carcinoma,
the prohibitive
Molecules price
2018, 23, 2334 is incubus especially in developing countries [13,14]. Besides prophylactic 2 of 17
vaccination, cervical cancer remains curable if detected at early stage, but hard to remedy in
metastatic or recurrent carcinoma [5]. Among conventional therapies including surgery,
the prohibitive price is incubus especially in developing countries [13,14]. Besides prophylactic
radiotherapy, chemotherapy and immunotherapy [15], chemotherapy is the first option for patients
vaccination, cervical cancer remains curable if detected at early stage, but hard to remedy in
that could effectively promote the apoptosis of cancer cells. Nevertheless, due to its high
metastatic or recurrent carcinoma [5]. Among conventional therapies including surgery, radiotherapy,
chemoresistance ability and toxicity on normal cells, more effective methods using less toxic
chemotherapy and immunotherapy [15], chemotherapy is the first option for patients that could
anticancer drugs and novel therapeutic intervention strategies are required nowadays. Polyphenols
effectively promote the apoptosis of cancer cells. Nevertheless, due to its high chemoresistance
such as catechins, curcumin and ferulic acid with low side effects are potential safe anticancer
ability and toxicity on normal cells, more effective methods using less toxic anticancer drugs and
strategies for cervical cancer intervention.
novel therapeutic intervention strategies are required nowadays. Polyphenols such as catechins,
Tea is one of the three most widely consumed non-alcohol beverages in the world. The
curcumin and ferulic acid with low side effects are potential safe anticancer strategies for cervical
prominent catechins in teas are (-)-epigallocatechingallate (EGCG), (-)-epicatechingallate (ECG), (-)-
cancer intervention.
epigallocatechin (EGC), and (-)-epicatechin (EC) [16]. EGCG accounts for more than 40% of total
Tea is one of the three most widely consumed non-alcohol beverages in the world. The prominent
catechins in green tea [17], and plays a critical role in cancer chemoprevention, diabetes,
catechins in teas are (-)-epigallocatechingallate (EGCG), (-)-epicatechingallate (ECG), (-)-epigallocatechin
neurodegenerative diseases, stroke, obesity and other biochemical disorders [18]. The cancer
(EGC), and (-)-epicatechin (EC) [16]. EGCG accounts for more than 40% of total catechins in green
prevention ability of EGCG is widely supported by results from epidemiological, in vivo and in vitro
tea [17], and plays a critical role in cancer chemoprevention, diabetes, neurodegenerative diseases,
studies [19–22], especially in breast cancer [23], liver cancer [24], and prostate cancer [25,26].
stroke, obesity and other biochemical disorders [18]. The cancer prevention ability of EGCG is widely
However, the effects of EGCG on the prevention of cervical cancer are still inconclusive and
supported by results from epidemiological, in vivo and in vitro studies [19–22], especially in breast
controversial [27]. This review summarizes recent research data mainly focused on the effects of
cancer [23], liver cancer [24], and prostate cancer [25,26]. However, the effects of EGCG on the prevention
EGCG on cervical cancer, including in vivo and in vitro studies, and offers directions for further
of cervical cancer are still inconclusive and controversial [27]. This review summarizes recent research
study.
data mainly focused on the effects of EGCG on cervical cancer, including in vivo and in vitro studies,
and offers directions for further study.
2. Anti-Proliferation of Cervical Cancer Cells
2. Anti-Proliferation
The mechanism of of tumor
Cervical Cancer Cells
progression is based on the proliferation and metastasis of cancer cells
[28]. The
Onemechanism
of the characteristics
of tumor progression is malignancies
in advanced based on the isproliferation
infinite proliferation of cancer
and metastasis cells.
of cancer
Inhibiting
cells proliferation
[28]. One of cervical cancer
of the characteristics cells could
in advanced stabilize the
malignancies symptoms
is infinite of a patient
proliferation and extend
of cancer cells.
Inhibiting proliferation of cervical cancer cells could stabilize the symptoms of a patient andcancer
the treatment duration with higher curative potential. EGCG can reduce cervical extend cell
the
proliferation in various ways (Figure 1), including: (1) inducing cancer cell cycle arrest;
treatment duration with higher curative potential. EGCG can reduce cervical cancer cell proliferation (2) regulating
cancer
in variouscell ways
growth; (3) inducing
(Figure cellular
1), including: microtubule
(1) inducing depolymerization
cancer cell cycle arrest;and(2) inhibiting
regulating tubulin
cancer
assembly; (4) inhibiting angiogenesis; and (5) restraining HPV oncoproteins.
cell growth; (3) inducing cellular microtubule depolymerization and inhibiting tubulin assembly;
(4) inhibiting angiogenesis; and (5) restraining HPV oncoproteins.
cervical adenocarcinoma cells [49]. Those results suggest that EGCG may be beneficial in early
Molecules 2018, 23, 2334 4 of 17
cervical lesions and cervical adenocarcinoma prevention.
2.3. Inducing
2.3. Inducing Cellular
Cellular Microtubule
Microtubule Depolymerization
Depolymerization andand Inhibiting
Inhibiting Tubulin
TubulinAssembly
Assembly
Microtubules are
Microtubules are dynamic
dynamic filamentous
filamentous cytoskeletal
cytoskeletal protein
protein structures
structures [50]
[50] composed
composed of of tubulins
tubulins
including α and β subunits [50]. Some key roles of microtubules are in proliferation,
including α and β subunits [50]. Some key roles of microtubules are in proliferation, signaling and signaling and
migration in cancer cells, hence both microtubule and tubulin are crucial therapeutic
migration in cancer cells, hence both microtubule and tubulin are crucial therapeutic targets for targets for
anticancer drugs.
anticancer drugs. A A recent
recent study
study found
found that
that EGCG
EGCG could
could inhibit
inhibit proliferation
proliferation ofof HeLa
HeLa cells
cells through
through
depolymerizing cellular microtubule and restraining tubulin assembly both
depolymerizing cellular microtubule and restraining tubulin assembly both in cells and cell-free in cells and cell-free
system (IC50
system (IC50 ofof 39.6
39.6±±0.63 μM)
0.63 µM) [36]. EGCG
[36]. EGCG andand
theaflavins also also
theaflavins prevented the reformation
prevented the reformationof cold-
of
treat cellular microtubule network distortion in HeLa cells [51]. The mechanism
cold-treat cellular microtubule network distortion in HeLa cells [51]. The mechanism of microtubule of microtubule
depolymerizationby
depolymerization byEGCG
EGCG is aissimilar
a similar anti-proliferation
anti-proliferation function
function to colchicine
to colchicine (a well-known
(a well-known tubulin
tubulin drug) through bounding to the α subunit of tubulin, and ultimately leading
drug) through bounding to the α subunit of tubulin, and ultimately leading to apoptosis in cervical to apoptosis in
cervical cancer
cancer cells [36]. cells [36].
2.4. Inhibiting
2.4. Inhibiting Angiogenesis
Angiogenesis
Angiogenesis consists
Angiogenesis consists of
of vascular
vascular endothelial
endothelial degradation,
degradation, adhesion
adhesion and
and migration
migration procedures,
procedures,
thatprovide
that provide nutrients
nutrients and and
oxygenoxygen in growth
in tumor tumor andgrowth and proliferation.
proliferation. Therefore,
Therefore, inhibiting inhibiting
angiogenesis
angiogenesis could effectively suppress the growth and proliferation of tumor cells. Recent
could effectively suppress the growth and proliferation of tumor cells. Recent work revealed that EGCG work
revealed that EGCG could interfere with cell signaling pathways of angiogenesis in ovarian, lung,
could interfere with cell signaling pathways of angiogenesis in ovarian, lung, breast and cervical cancer
breast
cells and cervical
(Figure cancer cells (Figure 2) [52–54].
2) [52–54].
recruitment of tumor-infiltrating macrophages [65]), EFNA1 (ephrin A1, a prototype ligand [66]),
TGF-β2 (transforming growth factor β 2), TNFAIP2 (tumor necrosis factor α-induced protein 2),
and CXCL6 (granulocyte chemotactic protein 2); meanwhile, EGCG up-regulated 4 gene expressions
consisting of ANGPTL4 (angiopoietin-like 4, inhibiting vascular invasion [67]), IFN-β1 (interferon β 1,
reducing pro-angiogenic factors [68] and blocking endothelial cell migration [69]), IL-1β (interleukin
1 β, decreasing the secretion of matrix metalloproteinase (MMP)-2 in cervical cancer [70]), and ID1
(inhibitor of DNA binding 1). These genes are known as mediating multiple mechanisms in proliferation,
adhesion, migration and invasion of vascular endothelial cells [71]. The results suggest that EGCG may
act as an important anti-angiogenic agent in cervical cancer.
in hypoxia-mediated activation of PI3K/Akt and ERK1/2 signaling pathways, EGCG and green tea
Molecules 2018, 23, 2334 6 of 17
extract down-regulated the mRNA and protein levels of VEGF that results in inhibiting HeLa cell
migration [61].
activation of PI3K/Akt and ERK1/2 signaling pathways, EGCG and green tea extract down-regulated
4. Pro-Apoptosis
the of Cervical
mRNA and protein Cancer
levels of VEGFCells
that results in inhibiting HeLa cell migration [61].
Anti-proliferation and anti-metastasis of cancer cells stand for restraining malignancies, while
4. Pro-Apoptosis of Cervical Cancer Cells
pro-apoptosis of cancer cells could cure cancer eventually. EGCG can induce apoptosis of cervical
Anti-proliferation
cancer and anti-metastasis
cells (Figure 3) through of cancer
(1) inducing caspase cells stand
secretion, forinduced
(2) ROS restraining malignancies,
apoptosis of cancer
while pro-apoptosis of cancer cells could cure cancer
cells, and (3) inducing lysosomal proteases secretion. eventually. EGCG can induce apoptosis of
cervical cancer cells (Figure 3) through (1) inducing caspase secretion, (2) ROS induced apoptosis of
cancer cells, and (3) inducing lysosomal proteases secretion.
Figure 3. Molecular mechanisms of EGCG promoting apoptosis of cervical cancer cells. EGCG
down-regulated E6 through estrogen, then the expressions of P53 and casp8 were up-regulated. EGCG
also could
Figure increase P53
3. Molecular expression
mechanisms of by AKT/PI3K
EGCG promoting pathways. BAX
apoptosis and casp3
of cervical would
cancer cells.promote their
EGCG down-
expressions
regulated E6with up-regulation
through of P53
estrogen, then and
the casp8 respectively.
expressions of P53 andThe up-regulation
casp8 of BAX EGCG
were up-regulated. and casp3
also
could promote apoptosis of cervical cancer cells in the end (broken lines mean indirect
could increase P53 expression by AKT/PI3K pathways. BAX and casp3 would promote their approaches).
AKT: protein with
expressions kinase B; BAX: Bcl-2ofassociated
up-regulation X protein;
P53 and casp8 Casp3: caspase
respectively. 3; Casp8: caspase
The up-regulation of BAX 8; E6:
andone of
casp3
HPV oncogenes; P53: tumor suppressor protein; PI3K: phosphoinositide-3-kinase.
could promote apoptosis of cervical cancer cells in the end (broken lines mean indirect approaches).
AKT: protein kinase B; BAX: Bcl-2 associated X protein; Casp3: caspase 3; Casp8: caspase 8; E6: one of
4.1. Inducing Caspase Secretion
HPV oncogenes; P53: tumor suppressor protein; PI3K: phosphoinositide-3-kinase.
Caspases are a series of proteases which have been verified as involved in tumor cell apoptosis,
necrosis and Caspase
4.1. Inducing inflammation.
SecretionThe deficiency of caspase would lead to tumor proliferation. EGCG
induced cancer cells apoptosis at high concentration (25–50 µg/mL) in squamous cervical carcinoma
Caspases are a series of proteases which have been verified as involved in tumor cell apoptosis,
Me180 cells [29]. In the meantime, EGCG improved the expression of caspase-3 in cervical carcinoma
necrosis and inflammation. The deficiency of caspase would lead to tumor proliferation. EGCG
cells [82]. Black tea polyphenol theaflavins and green tea catechins hydrate could also induce the
induced cancer cells apoptosis at high concentration (25–50 μg/mL) in squamous cervical carcinoma
expressions of caspase-3, -8, -9 and P53 in HeLa [30] or SiHa cells [83]. Eventually, EGCG could
Me180 cells [29]. In the meantime, EGCG improved the expression of caspase-3 in cervical carcinoma
promote caspase-mediated cancer cell apoptosis.
cells [82]. Black tea polyphenol theaflavins and green tea catechins hydrate could also induce the
expressions
4.2. of caspase-3,
Reactive Oxygen Species -8, -9 and
(ROS) P53 Apoptosis
Induced in HeLa of [30] or SiHa
Cancer Cellscells [83]. Eventually, EGCG could
promote caspase-mediated cancer cell apoptosis.
Reactive oxygen species (ROS) are a series of chemical-reactive molecules, containing hydrogen
peroxide, superoxide
4.2. Reactive anion (ROS)
Oxygen Species radical,Induced
singletApoptosis
oxygen, and hydroxyl
of Cancer Cells radical, which are associated with
the decreased antioxidant capability of cells and multiple stages of carcinogenesis [84]. In cancer cells,
a lowReactive
ROS level oxygen species
supports (ROS)pathways
survival are a series
thatofmakes
chemical-reactive molecules,
a positive impact in cellcontaining hydrogen
proliferation, but a
peroxide,orsuperoxide
medium high level anion
of ROS radical,
causessinglet oxygen,
oxidative stressandthathydroxyl
could leadradical,
to cellwhich are associated
apoptosis, necrosis with
and
the decreased
genotoxic damageantioxidant
[85]. capability of cells and multiple stages of carcinogenesis [84]. In cancer cells,
a low ROS level
EGCG is notsupports survival
only a natural pathwaysagent
antioxidant that makes a positive
in normal impact
cells but alsoin cell proliferation,
exhibits prooxidantbut anda
medium or high level
apoptosis-inducing of ROS in
properties causes
canceroxidative stressHigh
cells [86,87]. that concentrations
could lead to cell apoptosis,
of EGCG (50 µgnecrosis
to 200and
µg
genotoxic damage [85].
gallic acid equivalents GAE/mL) induced formation of intracellular superoxide anion radical, H2 O2 and
EGCG
hydroxyl is notasonly
radical, a natural antioxidant
a consequence of increasingagent in normal
oxidative stress cells but cells
in HeLa also [88].
exhibits prooxidant and
High-concentration
apoptosis-inducing properties in cancer cells [86,87]. High concentrations
(100 µg gallic acid equivalents GAE/mL) green tea induced the formation of intracellular of EGCG (50 μg ROS
to 200 μg
and
gallic acid equivalents GAE/mL) induced formation of intracellular superoxide
inhibited the activity of catalase, while increasing the production of superoxide anion radicals and anion radical, H2O2
and hydroxyl radical, as a consequence of increasing oxidative stress in HeLa cells [88]. High-
reducing glutathione. Thioredoxin (Trx) and thioredoxin reductase (TrxR) presenting in all living cells
concentration (100 μg gallic acid equivalents GAE/mL) green tea induced the formation of
act as antioxidant and apoptotic resistance proteins, which are often overexpressed in drug-resistant
intracellular ROS and inhibited the activity of catalase, while increasing the production of superoxide
cancer cells. Both of them are critical regulators of cellular redox homeostasis. The increased level
anion radicals and reducing glutathione. Thioredoxin (Trx) and thioredoxin reductase (TrxR)
of ROS induced by EGCG has proven to be an effective regulator for both Trx and TrxR [89].
presenting in all living cells act as antioxidant and apoptotic resistance proteins, which are often
The inactivation of Trx/TrxR by a high concentration of EGCG was linked to elevation of ROS
overexpressed in drug-resistant cancer cells. Both of them are critical regulators of cellular redox
levels in HeLa cells. By autoxidation, EGCG oxidized into EGCG (semi) quinones and released H2 O2 ,
Molecules 2018, 23, 2334 7 of 17
then inactivated Trx/TrxR through binding with them separately and formed irreversible EGCG-Trx
and EGCG-TrxR conjugates, thus inducing prooxidant cytotoxicity and apoptosis of HeLa cells.
Mitochondria, which play an important role in energy production, are major sites of ROS
generation [90]. Excessive generation of ROS could lead to the opening of a mitochondrial permeability
transition pore with decline in mitochondrial membrane potential (∆Ψm) and consequent release
of cytochrome-C from the inter-membrane space into the cytosol, culminating in activation of the
caspase cascade and apoptotic cell death pathways [91]. Treating SiHa cells with green tea polyphenols
consisting of EGCG decreased ∆Ψm of mitochondria and disrupted mitochondrial function, finally
inducing apoptosis of SiHa cells [92]. The mitochondrial perturbation ability of EGCG and tea
polyphenols may because they can induce the excess hydrogen peroxide.
7. Further Suggestions
Based on its anti-proliferation, anti-metastasis, pro-apoptosis and pharmaceutical synergistic
effects, EGCG shows suppressive effects on cervical cancer. An overview of suppressive effects of
Molecules 2018, 23, 2334 9 of 17
Molecules 2018, 23, x FOR PEER REVIEW 9 of 17
EGCG
EGCG on on cervical
cervical cancer
cancer isis illustrated
illustrated in
in Figure
Figure 4. 4. Meanwhile,
Meanwhile, the the cytotoxic
cytotoxic activities
activities of
of EGCG
EGCG on on
human cervical cancer cell lines are summarized
human cervical cancer cell lines are summarized in Table 2. in Table 2.
Among
Among anti-proliferation
anti-proliferationof ofcervical
cervicalcancer
cancercells,
cells,EGCG
EGCG depolymerizes
depolymerizes the the cellular
cellular microtubule
microtubule
and
and binds
binds on
on tubulin
tubulin identical
identical to to colchicine.
colchicine. However,
However, the the IC50
IC50 ofof EGCG
EGCG is is 1000-fold
1000-fold higher
higher than
than
colchicine. This indicates that
colchicine. This indicates that EGCG EGCG may not only target on tubulin of microtubule but also
on tubulin of microtubule but also other other
targets
targetsinincellular
cellular or extracellular
or extracellular regions [36]. Therefore,
regions further studies
[36]. Therefore, further should
studiesfocus on the
should mechanism
focus on the
of EGCG on depolymerizing
mechanism cellular microtubule.
of EGCG on depolymerizing cellularIn addition, EGCGIncan
microtubule. inhibit telomerase
addition, EGCG canin inhibit
breast
cancer [117],inesophageal
telomerase carcinoma
breast cancer [118], endocervical
[117], esophageal carcinoma cells andendocervical
[118], ectocervical cells
cells [42], but shows much
and ectocervical cells
milder
[42], butcytotoxic
shows much effects in cervical
milder cancer
cytotoxic cells
effects in [119].
cervicalThe activity
cancer cellsof[119].
telomerase is related
The activity to DNA
of telomerase
methylation
is related to DNA[120],methylation
and thus whether EGCG
[120], and thuscan inhibitEGCG
whether telomerase activity
can inhibit throughactivity
telomerase reducing DNA
through
methylation
reducing DNA is amethylation
promising research project. research project.
is a promising
effects of
Figure 4. Overview of suppressive effects of EGCG
EGCG on
on cervical
cervical cancer.
cancer.
EGCG
EGCG can can induce
induce non-apoptotic
non-apoptotic cell cell death
death through
through LMP LMP in in cervical
cervical cancer
cancer cells.
cells. But
But the
the
potential mechanism of cervical cancer cell death through upregulating ROS formation
potential mechanism of cervical cancer cell death through upregulating ROS formation has not been has not been
certified
certified in
in HeLa
HeLa cells
cells [88].
[88]. There
There isis only
only oneone study
study focused
focused on on the
the potency
potency of of tea
tea components
components in in
lysosome-associated
lysosome-associatedcell celldeath
deathpathways
pathways[121].
[121]. Further
Further studies
studies could
could work
work on on the
the mechanism
mechanism of of LMP
LMP
induced
induced byby EGCG
EGCG in in cervical
cervical cancer
cancer cells.
cells.
DNMT
DNMT and andHDAC
HDAC areare
twotwo
promising
promisingtargets for anticancer
targets drugs. Adrugs.
for anticancer series of
A effective
series ofepigenetic
effective
targeting
epigeneticdrugs in clinical
targeting drugstrials has been
in clinical proved,
trials but proved,
has been adverse butreactions
adversesuch as marrow
reactions suchsuppression
as marrow
and gastrointestinal
suppression symptoms also
and gastrointestinal appeared also
symptoms during treatment
appeared [122].treatment
during Thus, a low toxicity
[122]. Thus,natural
a low
inhibitor for epigenetic
toxicity natural inhibitor modification
for epigeneticis an attractive research
modification direction.
is an attractive EGCGdirection.
research as a dietary agentasina
EGCG
cancer
dietarytreatment has the
agent in cancer ability tohas
treatment inhibit the enzymatic
the ability to inhibitactivity of DNMT
the enzymatic and of
activity HDACDNMT in and
HeLa cells.
HDAC
However, the inhibition
in HeLa cells. However, mechanism
the inhibition needs more research
mechanism needs moreto support and
research tothe efficacy
support andofthe
EGCG for
efficacy
therapeutic
of EGCG forpurpose as anpurpose
therapeutic epigenetic drug
as an should be
epigenetic tested
drug in the
should befuture
tested[108].
in the future [108].
Molecules 2018, 23, 2334 10 of 17
Although chemoprevention effects and molecular mechanisms of EGCG have been illustrated
in cervical cancer, there is only one piece of epidemiological evidence and only one clinical study
has been undertaken until now. A case control study in central China showed that green tea intake
was identified as a protective factor against cervical cancer or cervical intraepithelial neoplasia [123].
Compared with other cancers [18–20], epidemiological research on the suppressive effects of EGCG in
cervical cancer is extremely rare.
8. Conclusions
EGCG has abilities in the anti-proliferation, anti-metastasis and pro-apoptosis of cervical cancer
cells. Although little epidemiological and clinical research supports the suppressive effects of EGCG
on cervical cancer, molecular evidence shows positive results of EGCG in inhibiting cervical cancer.
Moreover, EGCG could reduce the side-effects of traditional chemotherapy agents, such as CDDP and
BLM. Knowledge of more precise and deeper mechanisms of how EGCG restrains cervical cancer cells
is required. Overall, EGCG shows a potential role in suppressing cervical cancer.
Author Contributions: Y.-Q.W.: Figures 1–4; Tables 1 and 2; Section 4: Pro-apoptosis of cervical cancer
cells; Section 5: Pharmaceutical synergistic effect; J.-L.L.: Section 1: Introduction; Section 6: Ambiguous
potential functions of EGCG on cervical cancer; Y.-R.L.: Section 7: Further suggestions; Section 8: Conclusions;
Q.-S.L.: Section 2: Anti-proliferation of cervical cancer cells; Section 3: Anti-metastasis of cervical cancer cells;
modifying manuscript.
Funding: This work was financially supported by National Natural Science Foundation of China (No. 31470687).
Acknowledgments: The authors appreciate the advice of Xiao-Chang Wang, Xin-Qiang Zheng and Jian-Hui Ye.
Conflicts of Interest: The authors declare no conflict of interest.
Abbreviations
AKT: protein kinase B; ANGPTL4: angiopoietin-like 4; BAX: Bcl-2 associated X protein; BLM: bleomycin;
Casp3: caspase 3; Casp8: caspase 8; CCL2: chemoattractant protein 1; CDDP: cisdiamminedichloroplatinum II;
CDK2: cyclin-dependent kinase 2; CKI: cyclin-dependent kinases inhibitor; CXCL6: granulocyte chemotactic
protein 2; DNMT: DNA methyltransferase; EC: epicatechin; ECG: epicatechingallate; EFNA1: ephrin A1; EGC:
epigallocatechin; EGCG: epigallocatechingallate; EGF: epidermal growth factor; EGFR: epidermal growth factor
receptor; ER: estrogen receptor; ERK: extracellular signal-regulated kinase; ER-α: estrogen receptor-α; HDAC:
histone deacetylase; HIF-1: hypoxia-inducible factor 1; HIV: human immunodeficiency virus; HPV: human
papillomavirus; ID1: inhibitor of DNA binding 1; IFN-β1: interferon β 1; IGF-1: insulin-like growth factor
1; IGF-1R: insulin-like growth factor receptor; LMP: lysosomal membrane permeabilization; MMPs: matrix
metalloproteinases; mTOR: mammalian target of rapamycin; PBD: polo-box domain; PDGFA: platelet-derived
growth factor α; PI3K: phosphoinositide-3-kinase; PLK1: Polo-like kinase 1; RNA pol III: RNA polymerase III;
ROS: reactive oxygen species; TGF-β2: transforming growth factor β 2; THBS-1: thrombospondin 1; TIMP-1:
tissue inhibitor of metalloproteinase-1; TNFAIP2: tumor necrosis factor α-induced protein 2; Trx: thioredoxin;
TrxR: thioredoxin reductase; VEGF: vascular endothelial growth factor; 67LR: 67KD laminin receptor.
References
1. World Health Organization. Globocan2012: Estimated Cancer Incidence, Mortality and Prevalence
worldwide in 2012. Available online: http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx (accessed on
12 December 2013).
2. Waggoner, S.E. Cervical cancer. Lancet 2003, 361, 2217–2225. [CrossRef]
3. Moody, C.A.; Laimins, L.A. Human papillomavirus oncoproteins: Pathways to transformation. Nat. Rev. Cancer
2010, 10, 550–560. [CrossRef] [PubMed]
4. De Sanjose, S.; Quint, W.G.V.; Alemany, L.; Geraets, D.T.; Klaustermeier, J.E.; Lloveras, B.; Tous, S.; Felix, A.;
Bravo, L.E.; Shin, H.R.; et al. Human papillomavirus genotype attribution in invasive cervical cancer:
A retrospective cross-sectional worldwide study. Lancet Oncol. 2010, 11, 1048–1056. [CrossRef]
5. Doorbar, J.; Quint, W.; Banks, L.; Bravo, I.G.; Stoler, M.; Broker, T.R.; Stanley, M.A. The biology and life-cycle
of human papillomaviruses. Vaccine 2012, 30, F55–F70. [CrossRef] [PubMed]
Molecules 2018, 23, 2334 12 of 17
6. Bernard, H.U.; Burk, R.D.; Chen, Z.G.; Van Doorslaer, K.; Zur Hausen, H.; Ze Villiers, E.M. Classification of
papillomaviruses (pvs) based on 189 pv types and proposal of taxonomic amendments. Virology 2010, 401,
70–79. [CrossRef] [PubMed]
7. Hirchaud, F.; Hermetet, F.; Ablise, M.; Fauconnet, S.; Vuitton, D.A.; Pretet, J.L.; Mougin, C. Isoliquiritigenin
induces caspase-dependent apoptosis via downregulation of hpv16 e6 expression in cervical cancer caski
cells. Planta Med. 2013, 79, 1628–1635. [PubMed]
8. Bosch, F.X.; Burchell, A.N.; Schiffman, M.; Giuliano, A.R.; De Sanjose, S.; Bruni, L.; Tortolero-Luna, G.;
Kjaer, S.K.; Munoz, N. Epidemiology and natural history of human papillomavirus infections and
type-specific implications in cervical neoplasia. Vaccine 2008, 26, K1–K16. [CrossRef] [PubMed]
9. Melsheimer, P.; Vinokurova, S.; Wentzensen, N.; Bastert, G.; Doeberitz, M.V. DNA aneuploidy and integration
of human papillomavirus type 16 e6/e7 oncogenes in intraepithelial neoplasia and invasive squamous cell
carcinoma of the cervix uteri. Clin. Cancer Res. 2004, 10, 3059–3063. [CrossRef] [PubMed]
10. Ziegert, C.; Wentzensen, N.; Vinokurova, S.; Kisseljov, F.; Einenkel, J.; Hoeckel, M.; Doeberitz, M.V.
A comprehensive analysis of hpv integration loci in anogenital lesions combining transcript and
genome-based amplification techniques. Oncogene 2003, 22, 3977–3984. [CrossRef] [PubMed]
11. Wilting, S.M.; Steenbergen, R.D.M.; Tijssen, M.; Van Wieringen, W.N.; Helmerhorst, T.J.M.;
Van Kemenade, F.J.; Bleeker, M.C.G.; Van de Wiel, M.A.; Carvalho, B.; Meijer, G.A.; et al. Chromosomal
signatures of a subset of high-grade premalignant cervical lesions closely resemble invasive carcinomas.
Cancer Res. 2009, 69, 647–655. [CrossRef] [PubMed]
12. Poljak, M. Prophylactic human papillomavirus vaccination and primary prevention of cervical cancer: Issues
and challenges. Clin. Microbiol. Infect. 2012, 18, 64–69. [CrossRef] [PubMed]
13. Alhamlan, F.S.; Al-Zahrani, A.S.; Almatrrouk, S.A.; Al-Ahdal, M.N. Human papillomaviruses: The cervical
cancer saga in developing countries. J. Infect. Dev. Ctries. 2017, 11, 819–825. [CrossRef]
14. Yuan, C.H.; Filippova, M.; Tungteakkhun, S.S.; Duerksen-Hughes, P.J.; Krstenansky, J.L. Small molecule
inhibitors of the hpv16-e6 interaction with caspase 8. Bioorg. Med. Chem. Lett. 2012, 22, 2125–2129. [CrossRef]
[PubMed]
15. Vici, P.; Pizzuti, L.; Mariani, L.; Zampa, G.; Santini, D.; Di Lauro, L.; Gamucci, T.; Natoli, C.; Marchetti, P.;
Barba, M.; et al. Targeting immune response with therapeutic vaccines in premalignant lesions and cervical
cancer: Hope or reality from clinical studies. Expert Rev. Vaccines 2016, 15, 1327–1336. [CrossRef] [PubMed]
16. Liang, Y.R.; Ye, Q.; Jin, J.; Liang, H.; Lu, J.L.; Du, Y.Y.; Dong, J.J. Chemical and instrumental assessment of
green tea sensory preference. Int. J. Food Prop. 2008, 11, 258–272. [CrossRef]
17. Dong, J.J.; Ye, J.H.; Lu, J.L.; Zheng, X.Q.; Liang, Y.R. Isolation of antioxidant catechins from green tea and its
decaffeination. Food Bioprod. Process. 2011, 89, 62–66. [CrossRef]
18. Xiang, L.P.; Wang, A.; Ye, J.H.; Zheng, X.Q.; Polito, C.A.; Lu, J.L.; Li, Q.S.; Liang, Y.R. Suppressive effects of
tea catechins on breast cancer. Nutrients 2016, 8, 458. [CrossRef] [PubMed]
19. Hou, I.C.; Amarnani, S.; Chong, M.T.; Bishayee, A. Green tea and the risk of gastric cancer: Epidemiological
evidence. World J. Gastroenterol. 2013, 19, 3713–3722. [CrossRef] [PubMed]
20. Arts, I.C. A review of the epidemiological evidence on tea, flavonoids, and lung cancer. J. Nutr. 2008, 138,
1561S–1566S. [CrossRef] [PubMed]
21. Adhami, V.M.; Malik, A.; Zaman, N.; Sarfaraz, S.; Siddiqui, I.A.; Syed, D.N.; Afaq, F.; Pasha, F.S.; Saleem, M.;
Mukhtar, H. Combined inhibitory effects of green tea polyphenols and selective cyclooxygenase-2 inhibitors
on the growth of human prostate cancer cells both in vitro and in vivo. Clin. Cancer Res. 2007, 13, 1611–1619.
[CrossRef] [PubMed]
22. Lee, S.C.; Chan, W.K.; Lee, T.W.; Lam, W.H.; Wang, X.; Chan, T.H.; Wong, Y.C. Effect of a prodrug of the
green tea polyphenol (-)-epigallocatechin-3-gallate on the growth of androgen-independent prostate cancer
in vivo. Nutr. Cancer 2008, 60, 483–491. [CrossRef] [PubMed]
23. Chen, X.; Li, Y.; Lin, Q.; Wang, Y.; Sun, H.; Wang, J.; Cui, G.; Cai, L.; Dong, X. Tea polyphenols induced
apoptosis of breast cancer cells by suppressing the expression of survivin. Sci. Rep. 2014, 4, 4416. [CrossRef]
[PubMed]
24. Ni, C.X.; Gong, H.; Liu, Y.; Qi, Y.; Jiang, C.L.; Zhang, J.P. Green tea consumption and the risk of liver cancer:
A meta-analysis. Nutr. Cancer 2017, 69, 211–220. [CrossRef] [PubMed]
25. Connors, S.K.; Chornokur, G.; Kumar, N.B. New insights into the mechanisms of green tea catechins in the
chemoprevention of prostate cancer. Nutr. Cancer 2012, 64, 4–22. [CrossRef] [PubMed]
Molecules 2018, 23, 2334 13 of 17
26. Pandey, M.; Shukla, S.; Gupta, S. Promoter demethylation and chromatin remodeling by green tea
polyphenols leads to re-expression of GSTP1 in human prostate cancer cells. Int. J. Cancer 2010, 126,
2520–2533. [CrossRef] [PubMed]
27. Garcia, F.A.R.; Cornelison, T.; Nuno, T.; Greenspan, D.L.; Byron, J.W.; Hsu, C.H.; Alberts, D.S.; Chow, H.H.S.
Results of a phase II randomized, double-blind, placebo-controlled trial of polyphenon E in women with
persistent high-risk hpv infection and low-grade cervical intraepithelial neoplasia. Gynecol. Oncol. 2014, 132,
377–382. [CrossRef] [PubMed]
28. Small, W., Jr.; Bacon, M.A.; Bajaj, A.; Chuang, L.T.; Fisher, B.J.; Harkenrider, M.M.; Jhingran, A.;
Kitchener, H.C.; Mileshkin, L.R.; Viswanathan, A.N.; et al. Cervical cancer: A global health crisis. Cancer
2017, 123, 2404–2412. [CrossRef] [PubMed]
29. Zou, C.; Liu, H.; Feugang, J.M.; Hao, Z.; Chow, H.H.; Garcia, F. Green tea compound in chemoprevention of
cervical cancer. Int. J. Gynecol. Cancer 2010, 20, 617–624. [CrossRef] [PubMed]
30. Singh, M.; Singh, R.; Bhui, K.; Tyagi, S.; Mahmood, Z.; Shukla, Y. Tea polyphenols induce apoptosis through
mitochondrial pathway and by inhibiting nuclear factor-kappaB and Akt activation in human cervical cancer
cells. Oncol. Res. 2011, 19, 245–257. [CrossRef] [PubMed]
31. Ahn, W.S.; Huh, S.W.; Bae, S.M.; Lee, I.P.; Lee, J.M.; Namkoong, S.E.; Kim, C.K.; Sin, J.I. A major constituent
of green tea, EGCG, inhibits the growth of a human cervical cancer cell line, caski cells, through apoptosis,
G1 arrest, and regulation of gene expression. DNA Cell Biol. 2003, 22, 217–224. [CrossRef] [PubMed]
32. Zhang, L.F.; Wu, J.H.; Ling, M.T.; Zhao, L.; Zhao, K.N. The role of the PI3K/Akt/mTOR signalling pathway
in human cancers induced by infection with human papillomaviruses. Mol. Cancer 2015, 14, 1–13. [CrossRef]
[PubMed]
33. Sah, J.F.; Balasubramanian, S.; Eckert, R.L.; Rorke, E.A. Epigallocatechin-3-gallate inhibits epidermal growth
factor receptor signaling pathway. Evidence for direct inhibition of ERK1/2 and Akt kinases. J. Biol. Chem.
2004, 279, 12755–12762. [CrossRef] [PubMed]
34. Pollak, M.N.; Schernhammer, E.S.; Hankinson, S.E. Insulin-like growth factors and neoplasia. Nat. rev. Cancer
2004, 4, 505–518. [CrossRef] [PubMed]
35. Li, M.; He, Z.; Ermakova, S.; Zheng, D.; Tang, F.; Cho, Y.Y.; Zhu, F.; Ma, W.Y.; Sham, Y.; Rogozin, E.A.; et al.
Direct inhibition of insulin-like growth factor-I receptor kinase activity by (-)-epigallocatechin-3-gallate
regulates cell transformation. Cancer Epidemiol. Biomark. Prev. 2007, 16, 598–605. [CrossRef] [PubMed]
36. Chakrabarty, S.; Ganguli, A.; Das, A.; Nag, D.; Chakrabarti, G. Epigallocatechin-3-gallate shows
anti-proliferative activity in hela cells targeting tubulin-microtubule equilibrium. Chem. Biol. Interact.
2015, 242, 380–389. [CrossRef] [PubMed]
37. Liu, X.Q.; Erikson, R.L. Polo-like kinase 1 in the life and death of cancer cells. Cell Cycle 2003, 2, 424–425.
[CrossRef] [PubMed]
38. Goh, K.C.; Wang, H.S.; Yu, N.F.; Zhou, Y.F.; Zheng, Y.; Lim, Z.Y.; Sangthongpitag, K.; Fang, L.J.; Du, M.;
Wang, X.K.; et al. PLK1 as a potential drug target in cancer therapy. Drug Dev. Res. 2004, 62, 349–361.
[CrossRef]
39. Shan, H.M.; Shi, Y.; Quan, J. Identification of green tea catechins as potent inhibitors of the polo-box domain
of polo-like kinase 1. ChemMedChem 2015, 10, 158–163. [CrossRef] [PubMed]
40. Blackburn, E.H. Telomerases. Annu. Rev. Biochem. 1992, 61, 113–129. [CrossRef] [PubMed]
41. Shay, J.W. Role of telomeres and telomerase in aging and cancer. Cancer Discov. 2016, 6, 584–593. [CrossRef]
[PubMed]
42. Yokoyama, M.; Noguchi, M.; Nakao, Y.; Pater, A.; Iwasaka, T. The tea polyphenol, (-)-epigallocatechin gallate
effects on growth, apoptosis, and telomerase activity in cervical cell lines. Gynecol. Oncol. 2004, 92, 197–204.
[CrossRef] [PubMed]
43. Noguchi, M.; Yokoyama, M.; Watanabe, S.; Uchiyama, M.; Nakao, Y.; Hara, K.; Iwasaka, T. Inhibitory effect
of the tea polyphenol, (-)-epigallocatechin gallate, on growth of cervical adenocarcinoma cell lines. Cancer
Lett. 2006, 234, 135–142. [CrossRef] [PubMed]
44. Schramm, L.; Hernandez, N. Recruitment of RNA polymerase III to its target promoters. Gene Dev. 2002, 16,
2593–2620. [CrossRef] [PubMed]
45. White, R.J. RNA polymerase III transcription and cancer. Oncogene 2004, 23, 3208–3216. [CrossRef] [PubMed]
Molecules 2018, 23, 2334 14 of 17
46. Mital, R.; Kobayashi, R.; Hernandez, N. RNA polymerase III transcription from the human U6 and
adenovirus type 2 VAI promoters has different requirements for human BRF, a subunit of human TFIIIB.
Mol. Cell. Biol. 1996, 16, 7031–7042. [CrossRef] [PubMed]
47. Teichmann, M.; Wang, Z.X.; Roeder, R.G. A stable complex of a novel transcription factor IIB-related factor,
human TFIIIB50, and associated proteins mediate selective transcription by RNA polymerase III of genes
with upstream promoter elements. Proc. Natl. Acad. Sci. USA 2000, 97, 14200–14205. [CrossRef] [PubMed]
48. Jacob, J.; Cabarcas, S.; Veras, I.; Zaveri, N.; Schramm, L. The green tea component EGCG inhibits RNA
polymerase III transcription. Biochem. Biophys. Res. Commun. 2007, 360, 778–783. [CrossRef] [PubMed]
49. Yokoyama, M.; Noguchi, M.; Nakao, Y.; Ysunaga, M.; Yamasaki, F.; Iwasaka, T. Antiproliferative effects
of the major tea polyphenol, (-)-epigallocatechin gallate and retinoic acid in cervical adenocarcinoma.
Gynecol. Oncol. 2008, 108, 326–331. [CrossRef] [PubMed]
50. Kavallaris, M. Microtubules and resistance to tubulin-binding agents. Nat. Rev. Cancer 2010, 10, 194–204.
[CrossRef] [PubMed]
51. Chakrabarty, S.; Das, A.; Bhattacharya, A.; Chakrabarti, G. Theaflavins depolymerize microtubule network
through tubulin binding and cause apoptosis of cervical carcinoma hela cells. J. Agric. Food Chem. 2011, 59,
2040–2048. [CrossRef] [PubMed]
52. Ji, H.; Liu, N.; Yin, Y.; Wang, X.; Chen, X.; Li, J.; Li, J. Oxytocin inhibits ovarian cancer metastasis by repressing
the expression of MMP-2 and VEGF. J. Cancer 2018, 9, 1379–1384. [CrossRef] [PubMed]
53. Luo, H.Q.; Xu, M.; Zhong, W.T.; Cui, Z.Y.; Liu, F.M.; Zhou, K.Y.; Li, X.Y. EGCG decreases the expression of
HIF-1α and VEGF and cell growth in MCF-7 breast cancer cells. J. Buon 2014, 19, 435–439. [PubMed]
54. Li, X.Y.; Feng, Y.; Liu, J.H.; Feng, X.W.; Zhou, K.Y.; Tang, X.D. Epigallocatechin-3-gallate inhibits
IGF-I-stimulated lung cancer angiogenesis through downregulation of HIF-l α and VEGF expression.
J. Nutrigenet. Nutr. 2013, 6, 169–178. [CrossRef] [PubMed]
55. Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676.
[CrossRef] [PubMed]
56. De Francesco, E.M.; Sims, A.H.; Maggiolini, M.; Sotgia, F.; Lisanti, M.P.; Clarke, R.B. GPER mediates
the angiocrine actions induced by IGF1 through the HIF-1 α/VEGF pathway in the breast tumor
microenvironment. Breast Cancer Res. 2017, 19, 129. [CrossRef] [PubMed]
57. Wang, G.L.; Jiang, B.H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS
heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [CrossRef]
[PubMed]
58. Carmeliet, P.; Dor, Y.; Herbert, J.M.; Fukumura, D.; Brusselmans, K.; Dewerchin, M.; Neeman, M.; Bono, F.;
Abramovitch, R.; Maxwell, P.; et al. Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and
tumour angiogenesis. Nature 1998, 394, 485–490. [CrossRef] [PubMed]
59. Krishnamachary, B.; Berg-Dixon, S.; Kelly, B.; Agani, F.; Feldser, D.; Ferreira, G.; Iyer, N.; LaRusch, J.; Pak, B.;
Taghavi, P.; et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res. 2003,
63, 1138–1143. [PubMed]
60. Frezza, M.; Schmitt, S.; Dou, Q.P. Targeting the ubiquitin-proteasome pathway: An emerging concept in
cancer therapy. Curr. Top. Med. Chem. 2011, 11, 2888–2905. [CrossRef] [PubMed]
61. Zhang, Q.; Tang, X.; Lu, Q.; Zhang, Z.; Rao, J.; Le, A.D. Green tea extract and (-)-epigallocatechin-3-gallate
inhibit hypoxia- and serum-induced HIF-1α protein accumulation and VEGF expression in human cervical
carcinoma and hepatoma cells. Mol. Cancer Ther. 2006, 5, 1227–1238. [CrossRef] [PubMed]
62. Joyce, J.A. Therapeutic targeting of the tumor microenvironment. Cancer Cell 2005, 7, 513–520. [CrossRef]
[PubMed]
63. Park, C.C.; Bissell, M.J.; Barcellos-Hoff, M.H. The influence of the microenvironment on the malignant
phenotype. Mol. Med. Today 2000, 6, 324–329. [CrossRef]
64. Wilson, K.E.; Li, Z.Q.; Kara, M.; Gardner, K.L.; Roberts, D.D. β(1) integrin- and proteoglycan-mediated
stimulation of T lymphoma cell adhesion and mitogen-activated protein kinase signaling by
thrombospondin-1 and thrombospondin-1 peptides. J. Immunol. 1999, 163, 3621–3628. [PubMed]
65. Kleine-Lowinski, K.; Gillitzer, R.; Kuhne-Heid, R.; Rosl, F. Monocyte-chemo-attractant-protein-1
(MCP-1)-gene expression in cervical intra-epithelial neoplasias and cervical carcinomas. Int. J. Cancer
1999, 82, 6–11. [CrossRef]
Molecules 2018, 23, 2334 15 of 17
66. Cui, X.D.; Lee, M.J.; Yu, G.R.; Kim, I.H.; Yu, H.C.; Song, E.Y.; Kim, D.G. EFNA1 ligand and its receptor
EphA2: Potential biomarkers for hepatocellular carcinoma. Int. J. Cancer 2010, 126, 940–949. [CrossRef]
[PubMed]
67. Le Jan, S.; Amy, C.; Cazes, A.; Monnot, C.; Lamande, N.; Favier, J.; Philippe, J.; Sibony, M.; Gasc, J.M.;
Corvol, P.; et al. Angiopoietin-like 4 is a proangiogenic factor produced during ischemia and in conventional
renal cell carcinoma. Am. J. Pathol. 2003, 162, 1521–1528. [CrossRef]
68. Kaido, T.; Bandu, M.T.; Maury, C.; Ferrantini, M.; Belardelli, F.; Gresser, I. IFN-α(1) gene transfection
completely abolishes the tumorigenicity of murine B16 melanoma-cells in allogeneic DBA/2 mice and
decreases their tumorigenicity in syngeneic C57BL/6 mice. Int. J. Cancer 1995, 60, 221–229. [CrossRef]
[PubMed]
69. Sidky, Y.A.; Borden, E.C. Inhibition of angiogenesis by interferons-effects on tumor-induced and
lymphocyte-induced vascular-responses. Cancer Res. 1987, 47, 5155–5161. [PubMed]
70. Jovanovic, M.; Stefanoska, I.; Radojcic, L.; Vicovac, L. Interleukin-8 (CXCL8) stimulates trophoblast cell
migration and invasion by increasing levels of matrix metalloproteinase (MMP)2 and MMP9 and integrins
α(5) and β(1). Reproduction 2010, 139, 789–798. [CrossRef] [PubMed]
71. Tudoran, O.; Soritau, O.; Balacescu, O.; Balacescu, L.; Braicu, C.; Rus, M.; Gherman, C.; Virag, P.; Irimie, F.;
Berindan-Neagoe, I. Early transcriptional pattern of angiogenesis induced by EGCG treatment in cervical
tumour cells. J. Cell. Mol. Med. 2012, 16, 520–530. [CrossRef] [PubMed]
72. Gonzalez, S.L.; Stremlau, M.; He, X.; Basile, J.R.; Munger, K. Degradation of the retinoblastoma tumor
suppressor by the human papillomavirus type 16 E7 oncoprotein is important for functional inactivation
and is separable from proteasomal degradation of E7. J. Virol. 2001, 75, 7583–7591. [CrossRef] [PubMed]
73. Yokoyama, M.; Tsutsumi, K.; Pater, A.; Pater, M.M. Human papillomavirus 18-immortalized endocervical
cells with in-vitro cytokeratin expression characteristics of adenocarcinoma. Obstet. Gynecol. 1994, 83,
197–204. [PubMed]
74. Kuhn, D.J.; Burns, A.C.; Kazi, A.; Dou, Q.P. Direct inhibition of the ubiquitin-proteasome pathway by
ester bond-containing green tea polyphenols is associated with increased expression of sterol regulatory
element-binding protein 2 and LDL receptor. Biochim. Biophys. Acta 2004, 1682, 1–10. [CrossRef] [PubMed]
75. Wang, J.; Sampath, A.; Raychaudhuri, P.; Bagchi, S. Both Rb and E7 are regulated by the ubiquitin proteasome
pathway in HPV-containing cervical tumor cells. Oncogene 2001, 20, 4740–4749. [CrossRef] [PubMed]
76. Bonfili, L.; Cuccioloni, M.; Mozzicafreddo, M.; Cecarini, V.; Angeletti, M.; Eleuteri, A.M. Identification of an
EGCG oxidation derivative with proteasome modulatory activity. Biochimie 2011, 93, 931–940. [CrossRef]
[PubMed]
77. Qiao, Y.Y.; Cao, J.Y.; Xie, L.Q.; Shi, X.L. Cell growth inhibition and gene expression regulation by
(-)-epigallocatechin-3-gallate in human cervical cancer cells. Arch. Pharm. Res. 2009, 32, 1309–1315. [CrossRef]
[PubMed]
78. Giannelli, G.; Antonaci, S. MMP and TIMP assay in cancer: Biological and clinical significance. Int. J. Cancer
2005, 116, 1002–1003. [CrossRef]
79. GomisRuth, F.X.; Maskos, K.; Betz, M.; Bergner, A.; Huber, R.; Suzuki, K.; Yoshida, N.; Nagase, H.; Brew, K.;
Bourenkov, G.P.; et al. Mechanism of inhibition of the human matrix metalloproteinase stromelysin-1 by
TIMP-1. Nature 1997, 389, 77–81. [CrossRef] [PubMed]
80. Sharma, C.; Nusri, Q.E.A.; Begum, S.; Javed, E.; Rizvi, T.A.; Hussain, A. (-)-Epigallocatechin-3-gallate induces
apoptosis and inhibits invasion and migration of human cervical cancer cells. Asian Pac. J. Cancer 2012, 13,
4815–4822. [CrossRef]
81. Roomi, M.W.; Monterrey, J.C.; Kalinovsky, T.; Rath, M.; Niedzwiecki, A. In vitro modulation of MMP-2 and
MMP-9 in human cervical and ovarian cancer cell lines by cytokines, inducers and inhibitors. Oncol. Rep.
2010, 23, 605–614. [PubMed]
82. Siddiqui, F.A.; Naim, M.; Islam, N. Apoptotic effect of green tea polyphenol (EGCG) on cervical carcinoma
cells. Diagn. Cytopathol. 2011, 39, 500–504. [CrossRef] [PubMed]
83. Al-Hazzani, A.A.; Alshatwi, A.A. Catechin hydrate inhibits proliferation and mediates apoptosis of siha
human cervical cancer cells. Food Chem. Toxicol. 2011, 49, 3281–3286. [CrossRef] [PubMed]
84. Finkel, T. Oxidant signals and oxidative stress. Curr. Opin. Cell Biol. 2003, 15, 247–254. [CrossRef]
85. Beckman, K.B.; Ames, B.N. Oxidative decay of DNA. J. Biol. Chem. 1997, 272, 19633–19636. [CrossRef]
[PubMed]
Molecules 2018, 23, 2334 16 of 17
86. Azam, S.; Hadi, N.; Khan, N.U.; Hadi, S.M. Prooxidant property of green tea polyphenols epicatechin
and epigallocatechin-3-gallate: Implications for anticancer properties. Toxicol. In Vitro 2004, 18, 555–561.
[CrossRef] [PubMed]
87. Li, G.X.; Chen, Y.K.; Hou, Z.; Xiao, H.; Jin, H.Y.; Lu, G.; Lee, M.J.; Liu, B.; Guan, F.; Yang, Z.H.; et al.
Pro-oxidative activities and dose-response relationship of (-)-epigallocatechin-3-gallate in the inhibition
of lung cancer cell growth: A comparative study in vivo and in vitro. Carcinogenesis 2010, 31, 902–910.
[CrossRef] [PubMed]
88. Krstic, M.; Stojadinovic, M.; Smiljanic, K.; Stanic-Vucinic, D.; Velickovic, T.C. The anti-cancer activity of green
tea, coffee and cocoa extracts on human cervical adenocarcinoma hela cells depends on both pro-oxidant
and anti-proliferative activities of polyphenols. RSC Adv. 2015, 5, 3260–3268. [CrossRef]
89. Zhang, H.; Cao, D.; Cui, W.; Ji, M.; Qian, X.; Zhong, L. Molecular bases of thioredoxin and thioredoxin
reductase-mediated prooxidant actions of (-)-epigallocatechin-3-gallate. Free Radic. Biol. Med. 2010, 49,
2010–2018. [CrossRef] [PubMed]
90. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS
release. Physiol. Rev. 2014, 94, 909–950. [CrossRef] [PubMed]
91. Chung, Y.M.; Bae, Y.S.; Lee, S.Y. Molecular ordering of ROS production, mitochondrial changes, and caspase
activation during sodium salicylate-induced apoptosis. Free Radic. Biol. Med. 2003, 34, 434–442. [CrossRef]
92. Singh, M.; Tyagi, S.; Bhui, K.; Prasad, S.; Shukla, Y. Regulation of cell growth through cell cycle arrest and
apoptosis in HPV 16 positive human cervical cancer cells by tea polyphenols. Investig. New Drug. 2010, 28,
216–224. [CrossRef] [PubMed]
93. Luzio, J.P.; Pryor, P.R.; Bright, N.A. Lysosomes: Fusion and function. Nat. Rev. Cell Biol. 2007, 8, 622–632.
[CrossRef] [PubMed]
94. Zhang, Y.; Yang, N.D.; Zhou, F.; Shen, T.; Duan, T.; Zhou, J.; Shi, Y.; Zhu, X.Q.; Shen, H.M.
(-)-Epigallocatechin-3-gallate induces non-apoptotic cell death in human cancer cells via ROS-mediated
lysosomal membrane permeabilization. PLoS ONE 2012, 7, e46749. [CrossRef] [PubMed]
95. Ho, Y.P.; Au-Yeung, S.C.F.; To, K.K.W. Platinum-based anticancer agents: Innovative design strategies and
biological perspectives. Med. Res. Rev. 2003, 23, 633–655. [CrossRef] [PubMed]
96. Arany, I.; Safirstein, R.L. Cisplatin nephrotoxicity. Semin. Nephrol. 2003, 23, 460–464. [CrossRef]
97. Matsushima, H.; Yonemura, K.; Ohishi, K.; Hishida, A. The role of oxygen free radicals in cisplatin-induced
acute renal failure in rats. J. Lab. Clin. Med. 1998, 131, 518–526. [CrossRef]
98. Kilic, U.; Sahin, K.; Tuzcu, M.; Basak, N.; Orhan, C.; Elibol-Can, B.; Kilic, E.; Sahin, F.; Kucuk, O. Enhancement
of cisplatin sensitivity in human cervical cancer: Epigallocatechin-3-gallate. Front. Nutr. 2014, 1, 1–6.
[CrossRef] [PubMed]
99. Singh, M.; Bhui, K.; Singh, R.; Shukla, Y. Tea polyphenols enhance cisplatin chemosensitivity in cervical
cancer cells via induction of apoptosis. Life Sci. 2013, 93, 7–16. [CrossRef] [PubMed]
100. Singh, M.; Bhatnagar, P.; Srivastava, A.K.; Kumar, P.; Shukla, Y.; Gupta, K.C. Enhancement of cancer
chemosensitization potential of cisplatin by tea polyphenols poly(lactide-co-glycolide) nanoparticles.
J. Biomed. Nanotechnol. 2011, 7, 202. [CrossRef] [PubMed]
101. Alshatwi, A.A.; Athinarayanan, J.; Subbarayan, P.V. Green synthesis of platinum nanoparticles that induce cell
death and G2/m-phase cell cycle arrest in human cervical cancer cells. J. Mater. Sci. Mater. Med. 2015, 26, 7.
[CrossRef] [PubMed]
102. Burger, R.M.; Peisach, J.; Horwitz, S.B. Activated bleomycin-a transient complex of drug, iron, and oxygen
that degrades DNA. J. Biol. Chem. 1981, 256, 1636–1644.
103. Sleijfer, S. Bleomycin-induced pneumonitis. Chest 2001, 120, 617–624. [CrossRef] [PubMed]
104. Hay, J.; Shahzeidi, S.; Laurent, G. Mechanisms of bleomycin-induced lung damage. Arch. Toxicol. 1991, 65,
81–94. [CrossRef] [PubMed]
105. Vorechovsky, I.; Munzarova, M.; Lokaj, J. Increased bleomycin-induced chromosome-damage in lymphocytes
of patients with common variable immunodeficiency indicates an involvement of chromosomal instability
in their cancer predisposition. Cancer Immunol. Immunothera. 1989, 29, 303–306. [CrossRef]
106. Alshatwi, A.A.; Periasamy, V.S.; Athinarayanan, J.; Elango, R. Synergistic anticancer activity of dietary
tea polyphenols and bleomycin hydrochloride in human cervical cancer cell: Caspase-dependent and
independent apoptotic pathways. Chem. Biol. Interact. 2016, 247, 1–10. [CrossRef] [PubMed]
Molecules 2018, 23, 2334 17 of 17
107. Tallen, G.; Riabowol, K. Keep-ING balance: Tumor suppression by epigenetic regulation. FEBS Lett. 2014,
588, 2728–2742. [CrossRef] [PubMed]
108. Khan, M.A.; Hussain, A.; Sundaram, M.K.; Alalami, U.; Gunasekera, D.; Ramesh, L.; Hamza, A.; Quraishi, U.
(-)-Epigallocatechin-3-gallate reverses the expression of various tumor-suppressor genes by inhibiting DNA
methyltransferases and histone deacetylases in human cervical cancer cells. Oncol. Rep. 2015, 33, 1976–1984.
[CrossRef] [PubMed]
109. Virksaite, A.; Bakutyte, S.; Navakauskiene, R. Assessment of apoptosis and senescence in acute myeloid
leukemia NB-4 cells treated with epigenetic modifiers EGCG and BIX-01294. Eur. J Cancer 2016, 69, S85.
[CrossRef]
110. Subramaniam, D.; Thombre, R.; Dhar, A.; Anant, S. DNA methyltransferases: A novel target for prevention
and therapy. Front. Oncol. 2014, 4, 80. [CrossRef] [PubMed]
111. Sobel, M.E. Differential expression of the 67 kDa laminin receptor in cancer. Semin. Cancer Biol. 1993, 4,
311–317. [PubMed]
112. Kumazoe, M.; Sugihara, K.; Tsukamoto, S.; Huang, Y.H.; Tsurudome, Y.; Suzuki, T.; Suemasu, Y.; Ueda, N.;
Yamashita, S.; Kim, Y.; et al. 67-kDa laminin receptor increases cGMP to induce cancer-selective apoptosis.
J. Clin. Investig. 2013, 123, 787–799. [CrossRef] [PubMed]
113. Gundimeda, U.; McNeill, T.H.; Fan, T.K.; Deng, R.; Rayudu, D.; Chen, Z.; Cadenas, E.; Gopalakrishna, R.
Green tea catechins potentiate the neuritogenic action of brain-derived neurotrophic factor: Role of 67-kDa
laminin receptor and hydrogen peroxide. Biochem. Biophys. Res. Commun. 2014, 445, 218–224. [CrossRef]
[PubMed]
114. Umeda, D.; Tachibana, H.; Yamada, K. Epigallocatechin-3-O-gallate disrupts stress fibers and the contractile
ring by reducing myosin regulatory light chain phosphorylation mediated through the target molecule
67 kDa laminin receptor. Biochem. Biophys. Res. Commun. 2005, 333, 628–635. [CrossRef] [PubMed]
115. Umeda, D.; Yano, S.; Yamada, K.; Tachibana, H. Green tea polyphenol epigallocatechin-3-gallate signaling
pathway through 67-kDa laminin receptor. J. Biol. Chem. 2008, 283, 3050–3058. [CrossRef] [PubMed]
116. Tsukamoto, S.; Yamashita, S.; Kim, Y.H.; Kumazoe, M.; Huang, Y.; Yamada, K.; Tachibana, H. Oxygen
partial pressure modulates 67-kDa laminin receptor expression, leading to altered activity of the green tea
polyphenol, EGCG. FEBS Lett. 2012, 586, 3441–3447. [CrossRef] [PubMed]
117. Moradzadeh, M.; Hosseini, A.; Erfanian, S.; Rezaei, H. Epigallocatechin-3-gallate promotes apoptosis in
human breast cancer T47D cells through down-regulation of PI3K/AKT and telomerase. Pharmacol. Rep.
2017, 69, 924–928. [CrossRef] [PubMed]
118. Liu, L.; Zuo, J.; Wang, G.D. Epigallocatechin-3-gallate suppresses cell proliferation and promotes apoptosis
in Ec9706 and Eca109 esophageal carcinoma cells. Oncol. Lett. 2017, 14, 4391–4395. [CrossRef] [PubMed]
119. Wang, J.; Liu, L.; Ma, H.W. Label-free real-time investigation of the effect of telomerase inhibitors based on
quartz crystal microbalance measurement. Sens. Actuators B Chem. 2017, 239, 943–950. [CrossRef]
120. Stern, J.L.; Theodorescu, D.; Vogelstein, B.; Papadopoulos, N.; Cech, T.R. Mutation of the TERT promoter,
switch to active chromatin, and monoallelic TERT expression in multiple cancers. Genes Dev. 2015, 29,
2219–2224. [CrossRef] [PubMed]
121. Circu, M.; Cardelli, J.; Barr, M.P.; O’Byrne, K.; Mills, G.; El-Osta, H. Modulating lysosomal function through
lysosome membrane permeabilization or autophagy suppression restores sensitivity to cisplatin in refractory
non-small-cell lung cancer cells (vol 12, e0184922, 2017). PLoS ONE 2018, 13, e0197016. [CrossRef] [PubMed]
122. Kaminskas, E.; Farrell, A.T.; Wang, Y.C.; Sridhara, R.; Pazdur, R. FDA drug approval summary: Azacitidine
(5-azacytidine, vidazaTM ) for injectable suspension. Oncologist 2005, 10, 176–182. [CrossRef] [PubMed]
123. Jia, Y.; Hu, T.; Hang, C.Y.; Yang, R.; Li, X.; Chen, Z.L.; Mei, Y.D.; Zhang, Q.H.; Huang, K.C.; Xiang, Q.Y.; et al.
Case-control study of diet in patients with cervical cancer or precancerosis in Wufeng, a high incidence
region in China. Asian Pac. J. Cancer Prev. 2012, 13, 5299–5302. [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are not available from the authors.
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).