You are on page 1of 11

BBADIS-63368; No.

of pages: 11; 4C: 4, 5


Biochimica et Biophysica Acta xxx (2011) xxx–xxx

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbadis

Review

Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects


of polyphenols☆
F. Di Domenico a, C. Foppoli b, R. Coccia a, M. Perluigi a,⁎
a
Department of Biochemical Sciences, Sapienza University of Rome, P.le A. Moro, 5-00185 Rome, Italy
b
CNR Institute of Molecular Biology and Pathology, Sapienza University of Rome, Rome, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Cervical cancer lesions are a major threat to the health of women, representing the second most common
Received 27 August 2011 cancer worldwide. The unanimously recognized etiological factor in the causation of cervical cancer is the in-
Received in revised form 5 October 2011 fection with human papilloma virus (HPV). HPV infection, although necessary, is not per se sufficient to in-
Accepted 6 October 2011
duce cancer. Other factors have to be involved in the progression of infected cells to the full neoplastic
Available online xxxx
phenotype. Oxidative stress represents an interesting and under-explored candidate as a promoting factor
Keywords:
in HPV-initiated carcinogenesis. Oxidative stress is known to perturb the cellular redox status thus leading
Cervical cancer to alteration of gene expression responses through the activation of several redox-sensitive transcription fac-
HPV tors. This signaling cascade affects both cell growth and cell death. The ability of naturally occurring antiox-
Polyphenol idants to modulate cellular signal transduction pathways, through the activation/repression of multiple
Apoptosis redox-sensitive transcription factors, has been claimed for their potential therapeutic use as chemopreven-
Oxidative stress tive agents. Among these compounds, polyphenols have been found to be promising agents toward cervical can-
cer. In addition to acting as antioxidants, polyphenols display a wide variety of biological function including
induction of apoptosis, growth arrest, inhibition of DNA synthesis and modulation of signal transduction path-
ways. They can interfere with each stage of carcinogenesis initiation, promotion and progression to prevent can-
cer development. The present review discusses current knowledge of the major molecular pathways, which are
involved in HPV-driven cancerogenesis, and the ability of polyphenols to modulate these pathways. By acting at
specific steps of viral transformation cascade, polyphenols have been demonstrated to selectively inhibit tumor
cell growth and may be a promising therapeutic tool for treatment of cervical cancer. In addition, recent results
obtained in clinical trials using polyphenols are also discussed. This article is part of a Special Issue entitled: An-
tioxidants and Antioxidant Treatment in Disease.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction: cervical cancer (SIL), graded as low (LSIL) or high (HSIL). Cervical cancers are thought
to arise from these lesions after long persistent infection. The majority
Cervical cancer lesions are a major threat to the health of the of HPV infections are subclinical and transitory (80% disappears) and
women, representing the second most common cancer worldwide. in most of the cases they are resolved spontaneously by an effective im-
Human papillomaviruses (HPVs) have been identified as the major mune response that eliminates virus or suppresses it down to unde-
etiological factor in cervical carcinogenesis. The hypothesis of a corre- tected levels [2]. In few subjects, when the virus is not suppressed, the
lation between HPV infection and cervical neoplasia was formulated infection progress to CIN 1 and evolve to CIN 2/3 but the development
first time at the beginning of the 70's by Harold zur Hausen [1]. of cervical carcinoma is a multi-step carcinogenesis, however the
HPVs can be clinically classified as “low-risk” (LR-HPV) and “high- steps in the progression from LSIL to tumor remain unknown. It has
risk” (HR-HPV) depending on the relative propensity of the HPV- been estimated that approximately 10% of LSIL and about 20–30% of
associated lesions to undergo malignant progression. HPV 16 is the HSIL actually progress to cancer [3, 4].
most prevalent HR-HPV type, followed by HPV 18 and others. LR-
HPVs generate moderate dysplasia and genital warts, while high-risk
HPVs are associated with cervical dysplasia or cervical intraepithelial 2. HPV: structure, infection and transformation activity
neoplasia (CIN), currently indicated as squamous intraepithelial lesions
HPVs are small, non-enveloped DNA viruses with a circular genome
of around 8 kb. The viral genome can be divided into three regions. The
☆ This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment
in Disease.
up-stream regulatory region is largely responsible in determining the
⁎ Corresponding author. Tel.: + 39 0649910900. host range and tissue tropism of each HPV type and regulates viral
E-mail address: marzia.perluigi@uniroma1.it (M. Perluigi). gene expression after infection. This region contains the viral origin of

0925-4439/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbadis.2011.10.005

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
2 F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx

replication and numerous binding sites for cellular transcription factors. 3. Oxidative stress and cervical cancer
The rest of the genome comprises eight open reading frames divided
between the early region, that encodes the E1, E2, E4, E5, E6 and E7 pro- Persistent infection of the cervix with a HR-HPV is necessary to
teins and the late region, that encodes the L1 and L2 proteins [5]. E6, E7 develop cervical cancer, other associated co-factors are required for
and in minor grade E5 are well-recognized oncoproteins responsible for the malignancy progression. Oxidative stress, among others, is receiv-
cell transformation events. ing great interest for its role during the progression of neoplasias.
HPVs have a unique life cycle, which is tightly linked to the differ- Several risk factors of cervical cancer development, such as exposure
entiation program of the infected host epithelium. HPV infection to cigarette smoke and chronic inflammation, are well documented to
starts in the basal epithelium (basal cells or stem cells) in prolifera- increase oxidative stress, which could account for higher risk of cervical
tion, where HPVs are in the phase of latent infection and the viral ge- cancer in these two conditions [20, 21].
nome is maintained at a low copy number without production of Oxidative stress is a condition arising from an increased production
virions. In the upper layer, as the daughter cells move towards the of reactive oxygen species (ROS) associated with a decreased antioxi-
surface and undergo differentiation, vegetative replication of the dant capability of the cell. ROS are constantly generated in aerobic
viral genome coordinates with expression of capsid proteins to form cells by the incomplete reduction of molecular O2 to H2O during mito-
viral particles. Finally, the progeny virions are released from the up- chondrial oxidative phosphorylation, as well as during a number of pro-
permost layer within terminally differentiated epithelial squamae, cesses such as inflammation, infections, mechanical and chemical
to search for new host cells. HPV infection thus results in enhanced stresses, exposure to UV and to ionizing irradiation [22–24]. The effect
proliferation of the infected cells and their lateral expansion [2, 6, 7]. of ROS on cell depends on the level at which they are present. Low levels
The failure of the immune system, in persistent HPV infection clear- provide a beneficial effect, support cell proliferation and survival path-
ance, can lead, after about 10–20 years, to the development of cervical ways. However, at high levels, ROS can oxidatively damage biological
cancer. In LSIL, most HPV genomes are in episomal state whereas, in macromolecules and cause detrimental oxidative stress that can lead
many HSIL, genomes are integrated into the host cell. Over one-half of to cell death. ROS can induce genotoxic damage, including single- and
HPV 16-positive cancers and most HPV 18-positive malignancies con- double-strand breaks, DNA–protein cross-links, basic sites and modified
tain integrated HPV genomes, suggesting that integration may, in bases [25–27].
some cases, contribute to malignant progression. In lesions containing Cells withstand and counteract oxidative stress by the use of several
episomal HPV, the viral E2 protein directly represses early gene expres- and different defense mechanisms, ranging from free radical scavengers
sion regulating copy numbers. Integration of viral DNA usually disrupts and antioxidant molecules/enzymes to sophisticated and elaborate
E2 expression, leading to the deregulated expression of early viral DNA repair mechanisms to reduce excessive levels of ROS and prevent
genes, including E6 and E7, as well as increased proliferative capacity, irreversible cellular damage [28, 29].
a crucial step in progression to cancer [6]. An increased rate of ROS production occurs in highly proliferative
In cervical carcinoma cells, the E6 and E7 proteins are continuous- cancer cells, owing to the presence of oncogenic mutations that pro-
ly expressed and this is required for transformation. Prior to viral in- mote aberrant metabolism. Increased oxidative stress is well documen-
tegration these proteins are believed to cooperate in order to ted in transformed cells [30–33] and growing evidence suggests that
overcome normal cell cycle controls. Expression of the E6 and E7 pro- ROS act as second messengers in intracellular signaling cascades
teins from HR- but not from LR-HPV types lead to cell immortaliza- which induce and maintain the oncogenic phenotype of cancer cells
tion necessary for malignant progression. [34]. During cervical carcinogenesis an increase of oxidative DNA dam-
The most characterized role of HR-HPV E6 is its ability to bind to age, as shown by progressive increase in the levels of 8-OHdG from nor-
the p53 tumor suppressor protein in conjunction with the cellular mal to SIL to invasive carcinomas, has been reported [35]. In our recent
ubiquitin ligase E6AP and target p53 for degradation through the ubi- work we demonstrated that UVB-induced oxidative stress provoked an
quitin–proteasome pathway [8, 9]. This impairs the normal cellular increased and selective protein oxidation in keratinocyte cells trans-
response to cell stress and DNA damage and allows infected cells to fected with the whole HPV-16 genome [36]. Such oxidative modifica-
proliferate. Moreover since p53 plays a key role in preserving genome tions, combined with UVB-induced modulation of protein expression,
integrity, HPV infected cells accumulate chromosomal abnormalities likely lead to protein dysfunctions that might contribute to the malig-
thus increasing their intrinsic susceptibility towards malignancy. E6 nant progression of transformed cells.
can also interfere with the apoptotic pathway via its association However, ROS regulation is crucial for transformed cells that coun-
with proteins belonging to the bcl-2 family, including Bax and Bak teract ROS accumulation by up regulating antioxidant systems, seem-
[10, 11]. Another way by which E6 confers resistance to apoptosis is ingly creating a paradox of high ROS production in the presence of
the up-regulation of inhibitors of apoptosis proteins, as c-IAP2 and high antioxidant levels [37]. We further demonstrated that UVB irra-
survivin, through a mechanism involving NF-κB [12, 13]. diation caused, in HPV-transformed cells, a general suppression of
E7 is a small nuclear phosphoprotein known to bind to the retino- viral transcription, accompanied by a moderate growth arrest, an ap-
blastoma tumor suppressor gene product, pRb, and its family members, propriated response of cellular antioxidant enzymes, the activation of
p107 and p130. HR-HPV E7 protein, by binding to pRb leads to the re- cell repair mechanisms and a mild induction of apoptosis [38]. This
lease of E2F proteins and prematurely induces cells to enter the S response was absent in transformed cells devoid of HPV sequences
phase prevailing the normal cell cycle control. E7 from HR-HPV can (HaCaT) that appeared extremely susceptible to apoptosis. We postu-
also induce the degradation of pRb via a proteasome-dependent path- lated that the suppression of viral oncogenes, restoring the cell con-
way [14]. p16INK4a, a cyclin-dependent kinase (CDK) inhibitor, is over- trol on growth and repair mechanisms, allows the damage repair,
expressed when pRb is inactivated by E7 [15] in additional E7 interact ultimately resulting in a surviving response. The fine mechanisms ac-
with other CDK inhibitors such as p27 and p21 confirming its involve- tivated by cancer cells to counteract oxidative attack, allowing them
ment in the abrogation of cell-cycle inhibition [2]. Finally, in targeting to avoid ROS-induced cytotoxicity, also make them more exposed to
pRb, E7 causes deregulated E2F activity and E2F-1 can induce apoptosis additional ROS-mediated mutagenic events and accumulation of
in the presence of p53 [5]. E2F-1 activates expression of p19ARF, which DNA damage.
inhibits Mdm2 and thereby stabilizes p53 [5]. A number of studies dem- Studies on serum of patients with CIN and carcinoma of the cervix evi-
onstrated that HR-HPV oncogenes activate MAPKs and the AP-1 family denced changes in lipid peroxidation and impairment of antioxidant sys-
of transcription factors [16]. Moreover, increased COX-2 levels were tems, either enzymatic [21, 39] or not [40]. Other blood-related studies
shown in cervical cancer cells, via the EGFR→Ras→MAPK→AP-1 path- showed that LSIL, HSIL or cervical cancer can be associated with changes
way and in correlation to E6-mediated p53 degradation [16–19]. in three indicators of oxidative stress: increase in erythrocyte TBARS,

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx 3

ALA-D reactivation index and a decrease in vitamin C content [41]. Nitro- widely studied to gain an insight into its role in chemoprevention. It is
sative stress was also evidenced in patients with cervical squamous cell known to be involved in the regulation of ARE-mediated gene transcrip-
carcinoma, as shown by increased plasmatic NO levels [21]. tion. Polyphenols, such as curcumin and resveratrol, are known to in-
These data support the hypothesis about the involvement of both duce ARE-mediated gene expression by increasing the levels of Nrf2
oxidative and nitrosative stress in the pathogenesis of cervical cancer. protein or suppressing its turnover by ubiquitination [53]. They activate
Nrf2-mediated gene expression either by directly modifying the cysteine
4. Antioxidants, chemoprevention and chemotherapy residues on Keap1 to disrupt the Nrf2–Keap1 complex or by activating
kinase signaling pathways such as MAPKs, protein kinase C (PKC), and
In recent years, many studies have been carried out to investigate PI3K to phosphorylate Nrf2/Keap1 complex and/or facilitate the release
the potential cancer chemopreventive activities of natural antioxi- of Nrf2 or to increase the nuclear translocation of Nrf2 and regulate the
dants, in particular dietary polyphenols [42]. Representative exam- transcriptional activity of Nrf2 nuclear co-activators.
ples of polyphenols include epigallocatechin-3-gallate (EGCG) from One of the major research focus is to solve the “redox paradigm”,
green tea, curcumin from turmeric, and resveratrol from grapes that is to understand how these compounds can differentiate be-
(Fig. 2). These compounds have been proved to block carcinogenesis tween “normal” versus “abnormal tumor” cells in terms of signaling,
and to inhibit tumor growth both in vitro and in vivo models (animal gene expression, and pharmacological effects. Oxidative stress and
or in cell culture). These effects can be attributed to non-scavenging perturbation of intracellular redox status can have a profound influ-
role of antioxidants including induction of apoptosis, growth arrest, ence on the direction of signaling between cell survival and cell
inhibition of DNA synthesis and modulation of signal transduction death. The comprehension of the mechanisms which lead to cell
pathways [43]. Intracellular signaling cascade involve several molec- death of cancerous cells and cell survival of normal cells will need
ular events, which are initiated by the specific interaction of an extra- to be better investigated.
cellular ligand with its receptor at the cell membrane [44]. After this
first event, second messengers participate to amplify signal transduc- 5. Polyphenols and cervical cancer
tion pathways, which ultimately lead to modulation of several cellu-
lar functions. Within this frame, ROS/RNS have been proposed as Polyphenols, from natural and herbal extract are raising great in-
second messengers in the activation of several signaling pathways terest as powerful and safe anticancer strategy for their broad range
leading to mitogenesis or apoptosis. The effective transmission of sig- targeting capability and low side effects. Many herbal extract are
nals occurs through the recruitment of highly specific intermediates, both able to target against viral oncogenes and to inhibit the cooper-
and how ROS signaling may be activated with specificity and without ative signaling and deregulated gene expression of the host cells [3].
oxidative damage is still poorly understood [45]. In addition, gene ex- Recently an inverse association between antioxidant nutrients and
pression is significantly regulated in response to oxidative stress and HPV persistence, SIL, and cervical cancer was shown, suggesting pro-
is necessary to ensure cell survival involving specific redox-sensitive tective effect of natural extract against HPV persistence and cervical
transcription factors such as Nrf2, NF-κB, AP-1, and MAPKs [46]. Mod- dysplasia [57, 58]. A new approach in the use of natural extract con-
ulation of their activity leads to changes in the cellular redox status cern the sensitization of cancer cells to radio- and chemo-therapy
and alteration of gene expression responses to oxidative stresses, pre- by a pretreatment before the prolonged cure [59, 60].
sumably via sulfhydryl modification of critical cysteine residues found In the following sections are described the most recent research
on these proteins and/or other upstream redox-sensitive molecular about the anti-cancerogenic activities of curcumin, resveratrol and
targets [47]. For example, AP-1 is activated by low levels of oxidants epigallocatechin-3-gallate (EGCG), three of the most commonly studied
resulting in AP-1/DNA binding and an increase in gene expression. compounds from herbal extract, for the treatment of cervical cancer
In turn, AP-1 activation leads to the induction of JNK activity resulting (Figs. 1 and 2). Moreover few preliminary data on the therapeutic use
in the phosphorylation of the c-Jun transactivation domain [48]. On of these antioxidant compounds are reported.
the contrary, high concentrations of ROS inhibited AP-1 and AP-1 in-
duced gene expression. The inhibition of AP-1/DNA interactions is 5.1. Curcumin and ferulic acid
caused by the oxidation of specific cysteine residues in c-Jun's DNA
binding region. Curcumin (1,7-bis (4-hydroxy 3-methoxy phenyl)-1,6-heptadiene-
Similarly, NF-κB contains a redox-sensitive critical cysteine resi- 3,5-dione) is a natural polyphenolic compound extracted from the rhi-
due that is involved in DNA binding [47, 49]. NF-κB is normally se- zome of the medicinal plant Curcuma longa Linn (also known as turmeric)
questered in the cytoplasm by IκB, but under oxidative conditions, found in South and Southeast tropical Asia. Curcumin has been used for
IκB is phosphorylated by IκB kinase (IKK), ubiquitinated, and subse- centuries throughout Asia as a food additive, cosmetic, and as a tradition-
quently degraded. ROS production appears to be necessary to initiate al herbal medicine for its various biological activities.
the events leading to the dissociation of the NF-κB/IκB complex [50], Over the past decade, several studies have indicated the potential
but excessive ROS production results in the oxidation of cysteine res- therapeutic value of curcumin and have clearly demonstrated that curcu-
idue which does not affect its translocation to the nucleus, but rather, min possesses anti-inflammatory, antioxidant, anticarcinogenic, throm-
interferes with DNA binding and decreases gene expression [45]. bosuppressive, cardioprotective, antiarthritic, and anti-infectious
The ability of naturally occurring antioxidants to modulate cellular properties [61–68]. Several studies reported that curcumin suppresses
signal transduction pathways, through the activation/repression of all 3 stages of carcinogenesis: initiation, promotion, and progression. In-
multiple redox-sensitive transcription factors has been claimed for hibition of nuclear factor kappa B (NF-κB) and subsequent downregula-
their potential therapeutic use as chemopreventive agents. They can tion of various NF-κB-related proinflammatory pathways is very likely
interfere with each stage of carcinogenesis—initiation, promotion the primary target accounting for its efficacy [59]. Moreover curcumin
and progression—to prevent cancer development [51]. suppresses a number of key elements in cellular signal transduction
Decreased mutagenic risk could result from the induction of several pathways, including c-Jun/AP-1 activation [69, 70] and phosphorylation
phase 2 detoxifying and antioxidant enzymes, such as glutathione-S- reactions catalyzed by protein kinases [71]. Further, curcumin suppresses
transferase, NAD(P)H quinone reductase and heme oxygenase among angiogenesis in vivo, abrogates FGF-2-induced angiogenic response, ma-
others [46]. This process is known to involve the antioxidant response trix metalloprotease expression and cyclooxygenase-2 [72–75] (Table 1).
element (ARE), which is present in the promoter region of all the afore- The varied biologic properties of curcumin and lack of toxicity, even
mentioned genes [52–56]. Nuclear factor-E2-related factor-2 (Nrf2) is a when administered at doses as high as 8 mg/d [76], make curcumin an
basic leucine zipper, redox-sensitive transcription factor that has been ideal and very attractive compound in the attempt of therapeutic agents

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
4 F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx

against various tumors. Curcumin has been shown to prevent cancer in binding activity and reverse the expression dynamics of c-fos and
the colon, skin, stomach, duodenum, soft palate, and breasts of rodents fra-1 in HeLa cells. Subsequently, Divya et al. [82] indicated that cur-
after oral administration [77, 78]. Recent data indicate that in addition cumin is cytotoxic to cervical cancer cells in a concentration-
to its chemopreventive role, curcumin has great potential as a chemo- dependent and time-dependent manner and the cytotoxicity was
and radio-sensitizer [59]. Considering the toxic side effects of radio higher in HPV infected cells. The authors confirmed the inhibitory ac-
and chemotherapy, achieving radio and chemosensitization with mini- tion of curcumin on NF-κB activation, through the degradation of IκB,
mal toxicity represents a goal for the treatment of cancer patients. In the and on AP-1 binding that results in the downregulation of COX-2 and
other hand, one concern regarding curcumin therapeutic use is its low increased tumor cell apoptosis. Moreover, it was shown that curcu-
bioavailability and poor pharmacokinetics [76]. min downregulates the expression of HPV-16 and -18 E6 and E7
In regard of curcumin antitumoral properties, several studies were resulting in loss of the transforming phenotype and the cessation of
conducted on cervical cancer. Cervical cancer cell lines were often used cellular growth. Current studies by Singh and Singh [83, 84] explored
as a preferred carcinogenic model to understand molecular targets and other molecular mechanisms exerted by curcumin in cervical cancer
mechanism of curcumin action. Here are reported all the in vitro and in cells, showing that it inhibits telomerase activity, Ras and ERK signal-
vivo advances for the use of curcumin in the treatment of cervical cancer. ing pathway, cyclin D1, COX-2 and iNOS activity. Moreover, curcumin
Initial studies about the effect of curcumin on NF-κB activity in cervi- decreases c-Myc transcriptional factor and Hsp 70 chaperone protein,
cal cancer cells were conducted by Venkatraman et al. in 2005 [79]. They activates AIF, releases cytochrome c and enhances apoptosis via the
demonstrated in SiHa cell line, normally resistant to cisplatin, that inhi- mitochondrial pathway with the upregulation of BAX and the down-
bition of NF-κB activity induced by curcumin determined increased sen- regulation of Bcl-2 and Bcl-XL. The authors further demonstrated that
sitization to cisplatin treatment evaluated as increased cell-death. the activity of curcumin on these multiple targets was able to revert
According with this study Bava et al. [80] showed that curcumin sensi- the proliferative action due to a pretreatment with estradiol on cervi-
tizes cervical cancer cells to the therapeutic effect of taxol, acting in the cal cancer cell. A different approach was used by Madden et al. [85]
down-regulation of both NF-κB and serine/threonine kinase AKT path- that with a proteomics approach analyzed the effect of curcumin on
way, a survival signal related to NF-κB. HeLa cells, reporting significant changes in tumor-related proteins
A further step in the investigation of curcumin molecular target linked to cell metabolism, cell cycle, and carcinogenic process.
was made by the study of Prusty et al. [81], which showed that curcu- Other recent mechanistic studies confirmed the suppression of
min can downregulate HPV-18 transcription, selectively inhibit AP-1 cancer cell growth by curcumin also in a three-dimensional raft

Fig. 1. Molecular mechanisms of human papillomavirus-induced carcinogenesis. Oncogenic action of high-risk HPV E6 and E7 oncoproteins is exerted through the inactivation of
tumor suppressors p53 and pRb, respectively. Abrogation of p53 by E6 ensures cell survival by preventing apoptosis inactivation while pRb inactivation by E7 forces infected cells to
remain in a proliferative state. The dark red box represent the target of action of the polyphenols considered: C = curcumin; E = EGCG; R = resveratrol.

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx 5

Fig. 2. Polyphenolic antioxidants. Chemical structure, properties and sources of curcumin (1,7-bis (4-hydroxy 3-methoxy phenyl)-1,6-heptadiene-3,5-dione), ferulic acid (3-methoxy-4-
hydroxycinnamic acid), EGCG (epigallocatechin-3-gallate) and resveratrol (trans-3,5,4′-trihydroxy-trans-stilbene).

culture system [86]. This effect was exerted through the induction of prooxidant activity of FA was observed in different experimental
apoptosis, the inhibition of cell motility, the decrease in expression models that might help to enhance radiation effects in cancer cells, as
of HPV oncoproteins, and the restoration of expression of tumor sup- was previously proven for other phenolic compounds [97].
pressor proteins. Interestingly they also showed that curcumin was A recent study from Karthikeyan et al. [98] investigated the radio-
able to inhibit benzo[a]-pyrene (BaP) induced increase in HPV sensitizing effect of FA on two human cervical carcinoma cell lines.
oncoproteins. The authors showed that FA enhances radiation effects by decreasing
Several studies proposed curcumin as a powerful radio- and cancer cell viability through the influence on mitochondrial activity,
chemo-sensitizer. Javvadi et al. [87] showed that curcumin, with ion- cell survival, and antioxidant status. Further was showed that FA
izing radiation (IR), produced a significant increase of ROS, which fur- and IR increase lipid peroxidative markers, ROS levels, oxidative
ther led to sustained ERK 1/2 activation, favoring the radiotherapic DNA damage and apoptotic morphological changes, corroborating
effect on cervical cancer cells. Moreover Javvadi et al. showed later that treatment of cancer cells with FA may help in augmenting the an-
the involvement thioredoxin reductase enzyme in the radiosensitiz- ticancer effect of radiation.
ing action of curcumin in cervical cancer cells [88]. Very recently the From the studies aforementioned in this review, it is clear that
group of Anto R.J. showed that curcumin given during paclitaxel curcumin and its product FA holds great potential in the prevention
treatment act as a down-regulator of paclitaxel pro-survival path- and therapy of cervical cancer. Researchers suggest that curcumin
ways linked to NF-κB and Akt activation in cervical cancer cells and represents an ideal compound for cancer patient treatment, consider-
mouse models of cervical cancer [89, 90]. These studies, both in ing the encouraging data as a safe and effective antitumoral and
vitro and in vivo, indicate that NF-κB is one of the central players in chemo- and radio-sensitizer agent, and considering also its ability to
the synergism of paclitaxel and curcumin and act as a regulator of prevent depression, fatigue, lack of appetite and most of the symp-
Cox-2, Cyclin D1, XIAP and cIAP1. AKT instead is the regulator of toms induced by cancer.
NF-κB, which through the phosphorylation of MAPKs regulates a set
of survival signals. 5.2. Epigallocatechin-3-gallate
The degradation of curcumin brings the formation of two products:
vanillic acid and ferulic acid (FA) and FA represent the major compo- Green tea is one of the most widely consumed beverages. It has
nent of curcumin. been proven that it possesses beneficial anticarcinogenic and antipro-
FA (3-methoxy-4-hydroxycinnamic acid) is a dietary phytochemical liferative properties attributed to the biological properties of green
that occurs primarily in the seeds and leaves of most plants such as rice, tea polyphenolic compounds. Consumption of green tea has been
wheat, barley, oat, roasted coffee, tomatoes, and citrus fruits. FA ethyl reported to lower the risk of developing gastric, pancreatic, and colo-
ester, FAEE, has been largely investigated in the protection against OS rectal cancers in human populations [99, 100]. The polyphenols ac-
during neurodegeneration, driven by Alzheimer disease, for its ability count for up to 30% of the dry weight of green tea and include
to cross the brain blood barrier and exert its activity directly on OS chal- flavanols, flavandiols, flavonoids, and phenolic acids [101]. In particu-
lenged brain cells [91–94]. Moreover FA and FAEE are well-known anti- lar, flavanols, known as catechins, are a major component of most
oxidant agents used to prevent damage to cells caused by ultraviolet green tea. Generally, a typical cup of green tea contains 100 to
light [95]. For this property FA is used in a wide range of cosmetics, 150 mg catechins, including 50% of epigallocatechin-3-gallate, 15%
such as skin lighteners, sunscreens, anti-aging creams and moisturizers. of epigallocatechin, 15% of epicatechin-3-gallate, and 8% of epicate-
However several studies show that FA exhibits chemotherapic proper- chin [102]. Epigallocatechin-3-gallate (EGCG) is the major bioactive
ty. It was described as a specific inhibitor of the anti-apoptotic proteins polyphenol present in green tea and displays antiproliferative, anti-
Bcl-XL and Bcl-2, thereby inducing apoptosis [96]. However, a angiogenic, antimetastatic, proapoptotic, and cell cycle perturbation

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
6 F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx

Table 1
Biochemical effects and molecular targets of chemopreventive action of polyphenols (curcumin, ferulic acid, EGCG, and resveratrol) in cervical cancer models.

Effects in cervical Compound Molecular targets References


cancer models

Cell growth Curcumin WAF-1/p21; Cdc2; p16; Venkatraman et al. (2005); Prusty et al. (2005); Bava et al. (2005); Divya et al. (2006); Javvadi et al. (2008);
inhibition F.A. PCNA; cyclin D1 Singh et al. (2009); Maher et al. (2011); Singh et al. (2011); Sreekanth et al. (2011); Bava et al. (2011);
Karthikeyan et al. (2011)
EGCG G1 phase arrest; WAF-1/ Ahn et al. (2003); Sah et al. (2003); Yokoyama et al. (2004); Noguchi et al. (2006); Yokoyama et al. (2008);
p21; KIP-1/p27; cyclin; Qiao et al. (2009); Zou et al. (2010)
CDK
Resveratrol S phase arrest Zoberi et al. (2002); Roy et al. (2002); Kramer et al. (2009)
Apoptosis induction Curcumin Caspases 3, 8, 9; DNA Venkatraman et al. (2005); Prusty et al. (2005); Bava et al. (2005); Divya et al. (2006); Singh et al. (2009);
F.A. damage; p53; PARP; Bava et al. (2011); Maher et al. (2011); Karthikeyan et al. (2011)
cytocrome c; Bax, AIF;
Bcl-2; Bcl-XL; annexin V
EGCG p53; BAD; caspase 3 Ahn et al. (2003); Sah et al. (2003); Yokoyama et al. (2004); Yokoyama et al. (2008);
Qiao et al. (2009); Siddiqui et al. (2010); Zou et al. (2010)
Resveratrol DNA damage; caspase 3; Roy et al. (2002); Hsu et al. (2009)
cathepsin L
Angiogenesis Curcumin VEGF Sreekanth et al. (2011)
inhibition F.A.
EGCG VEGF; HIF-1α Zhang et al. (2007)
Resveratrol VEGF; HIF-1α Tang et al. (2007)
Transcriptional Curcumin AKT; NF-κB; AP-1; Venkatraman et al. (2005); Bava et al. (2005); Prusty et al. (2005); Divya et al. (2006); Javvadi et al. (2008);
activity inhibition F.A. telomerase; c-fos; fra-1 Singh et al. (2009); Singh et al. (2011); Sreekanth et al. (2011);
Bava et al. (2011); Maher et al. (2011)
EGCG AKT; RNA pol. III; Sah et al. (2003); Yokoyama et al. (2004); Noguchi et al. (2006); Li et al. (2007);
telomerase Zhang et al. (2007); Jacob et al. (2007); Yokoyama et al. (2008)
Resveratrol AKT Sexton et al. (2006)
Signal transduction Curcumin ERK 1/2; JNK; p38; c-Jun Javvadi et al. (2008); Singh et al. (2009); Sreekanth et al. (2011); Bava et al. (2011)
pathways regulation F.A.
EGCG ERK 1/2; p90RSK Sah et al. (2003); Li et al. (2007); Zhang et al. (2007)
Resveratrol ERK; JNK; PKC-δ Woo et al. (2004)
HPV suppression Curcumin E6/E7; HPV-18 mRNA Prysty et al. (2005); Divya et al. (2006); Maher et al. (2011)
F.A.
EGCG E6/E7 Qiao et al. (2009); Zou et al. (2010)
Resveratrol
Cyclooxygenase Curcumin COX-2; Divya et al. (2006); Singh et al. (2009); Madden et al. (2009); Sreekanth et al. (2011);
inhibition F.A. Bava et al. (2011)
EGCG
Resveratrol COX-1; COX-2 Zoberi et al. (2002); Sexton et al. (2006)
Cell redox status/cell Curcumin iNOS; HSP7; TRX-R; ROS Javvadi et al. (2008); Singh et al. (2009); Javvadi et al. (2010); Karthikeyan et al. (2011)
stress response F.A. production
EGCG
Resveratrol
Other effects Curcumin MMP-9, -2; ICAM-1; Sreekanth et al. (2011); Bava et al. (2011)
F.A. survivin
EGCG IGF-IR; EGFR; aromatase; Sah et al. (2003); Li et al. (2007); Qiao et al. (2009); Bonfili et al. (2011)
ER α, β; proteasome
inhibition
Resveratrol MMP-9; autophagy Woo et al. (2004); Hsu et al. (2009)

activities in various in vitro and in vivo tumor models [103–109] ubiquitinated proteins and three natural proteasome targets (p27,
(Fig. 2). It has also been reported that there exists a threshold level IκB-α and Bax). Sah et al. [116] investigated the effect of EGCG on
of EGCG that induces apoptosis and growth inhibition in cancer cell EGFR signaling in several cervical cells lines, demonstrating that
lines, but not in normal cells [110, 111]. EGCG acts as a potent antiox- EGCG inhibits EGFR, the initial kinase in the EGF signaling cascade,
idant and can scavenge ROS, such as lipid free radicals, superoxide while activating the ERK1/2 and AKT activity. This inhibition is associ-
radicals, hydroxyl radicals, hydrogen peroxide and singlet oxygen ated with reduced phosphorylation, levels and activity of ERK1/2 and
[112–115]. AKT downstream substrates, which lead again to G1 arrest and in-
EGCG has been shown to inhibit several critical signal transduc- creased apoptosis. Yokoyama et al. and Noguchi et al. [122, 123] indi-
tion pathways as well the activation of the redox-sensitive transcrip- cated that EGCG, beyond cell growth and apoptosis, target also
tion factors, NF-κB and AP-1 in cultured cells [116–118]. telomerase, inhibiting its activity required for the initiation and pro-
One of the first study about the effect of EGCG on the HPV-16 as- gression of cervical lesions. It is well recognize that angiogenesis is a
sociated cervical cancer cell line CaSki, by Ahn et al. [119] demon- key step in the progression cervical cancer. Zhang et al. [124] reported
strated that an EGCG antiproliferative effect depends by the arrest that EGCG exhibit anti-angiogenic activities for the suppression of
of cell cycle in G1 phase that precedes in time the consequent pro- HIF-1a protein accumulation and the decrease of VEGF expression at
grammed cell death by apoptosis. Furthermore, antitumor effects of both mRNA and protein levels, through the enhancement of protea-
EGCG were also observed in nude mice challenged with CaSki cells some system activity and the blocking of PI3K/AKT and ERK1/2 sig-
and then treated orally with EGCG. Siddiqui et al. [120], in a recent naling pathways. The anti-angiogenic effect of EGCG was shown
study on cervical cancer cells extracted from fresh tissue, observed recently also by Tudoran et al. [125] through the analysis on the alter-
apoptosis induction by EGCG, confirming results previously obtained. ations of the expression pattern of 84 genes known to be involved in
Afterwards, Khun et al. [121] showed that EGCG potently inhibits the the angiogenesis process. Li et al. [126] showed that EGCG abrogates
proteasomal activity in HeLa cells, as evident by accumulation of anchorage-independent growth and inhibits cell proliferation and

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx 7

transformation induced by IGF-IR overexpression by competing with division inducing an early S-phase arrest, suggesting a role for resver-
ATP, that result in a prevention of IGF-IR downstream signaling and atrol in the regulation of cell cycle progression. Kramer et al. [131]
expression of cervical cancer cell phenotype. Another target of EGCG using a double dose of resveratrol confirmed this result adding that
anti-proliferative activity in cervical cancer cells is RNA pol III tran- resveratrol-induced S-phase block was transient and reversible. Cy-
scription, as reported by Jacob et al. [127]. The authors found that clooxygenase (COX) is well recognized as one of the main targets of
EGCG treatment inhibits transcription by RNA pol III, from both the resveratrol mechanism of action [142] [143]; in their study Zoberi
gene internal (tRNA) and gene external (U6 snRNA) promoters. et al. showed that COX-1 is overexpressed in cervical cancer cells
Moreover EGCG inhibits expression of the TFIIIB subunits Brf1 and while resveratrol treatment inhibits COX-1 activity. PGs are implicat-
Brf2, and inhibits the activity of the human Brf2 promoter. An essen- ed in the response of tumor cells to ionizing radiation and other cyto-
tial effect of EGCG on cervical cancer cell lines is represented by the toxic insults; COX-1 inhibition suppresses PGs biosynthesis,
inhibition of HPV oncogenes E6/E7 expression. Quiao et al. [128] ob- suggesting that COX-1 inhibition by resveratrol might alter how
served that EGCG, in cervical cancer cells, could suppress mRNA and tumor cells respond to the cytotoxicity of ionizing radiation. Zoberi
protein expression of ERα and aromatase limiting in this way the ex- et al. supported this hypothesis demonstrating that HeLa and SiHa
pression of E6/E7, and indirectly inhibiting cervical cancer cell growth cell line cytotoxicity was enhanced by pretreatment with resveratrol
and inducing apoptosis. Zou et al. [129] achieved a similar result prior to ionizing radiation exposure. Sexton et al. [144] showed, in
showing decreased expression of E7 oncogene, increased apoptosis six different human uterine cancer cell lines, that treatment with res-
coupled with increased expression levels and increased cell growth veratrol decrease endogenous COX-2 protein levels, concomitant
inhibition during treatment of cervical cancer cells with EGCG. Re- with a decrease in production of COX metabolites PGE2 and PGF2α.
cently, Bonfili et al. [130] sustained that the observed EGCG effects They suggest in their study that resveratrol regulates COX expression
on proteasome functionality and on pro-apoptotic protein intracellu- not directly but possibly thru other enzymes involved in prostaglan-
lar concentrations in cervical cancer cells may be the result of a com- din synthesis that act downstream of the COXs. Moreover they also
bined action of EGCG and its oxidation product. In fact the authors showed that resveratrol reduced cellular levels of the phosphorylat-
observed that EGCG, under a common experimental condition un- ed/active form of anti-apoptotic kinase AKT. Lin et al. obtained similar
dergoes modification, involving the hydrolysis of the gallate ester, results, about COX-2 suppression by resveratrol, in ovarian and breast
and the disruption of the catechin A ring, with the formation of a bi- cancer cells supporting the role of resveratrol on cervical cancer cells
ologically active product with inhibitory activity towards cellular [145–147]. Another recognized mechanism of resveratrol chemopre-
proteasomes. vention is represented by the inhibition of metalloproteinases
Green tea represents, for its common use, one of the most studied (MMPs). A positive correlation between the expression of MMP-9
antioxidant compounds. From the aforementioned studies, it is clear and tumor metastasis for several types of cancer was recently
that EGCG, a green tea major constituent, holds great potential in shown [148, 149]. Woo et al. [150] demonstrated, in a CaSki cervical
the prevention and therapy of cervical cancer. Taken together, these cancer cell line, a decreased expression of MMP-9 during treatment
data support the idea that EGCG provides an additional option for a with resveratrol that exerts its actions through a tight regulation of
new and potential drug approach for cervical cancer patients. MMP-9 transcription. The authors also show that reduced MMP-9
levels depend by reduced PKC-δ activity, JNK activation as well as
5.3. Resveratrol diminished NF-κB and AP-1 activity during treatment with
resveratrol.
Resveratrol (trans-3,5,4′-trihydroxy-trans-stilbene) is a natural The progression of cervical cancer relies on tumor angiogenic ac-
polyphenolic phytoalexin most commonly found in nuts, berries, tivity and several studies showed that HPV-16 oncoproteins, E5, E6,
grapes, and red wine. In several plants, including grapevine, resvera- and E7, induce VEGF expression in several established cervical cancer
trol is synthesized in response to stress, injury, ultraviolet irradiation, cells lines [151–153], suggesting that high-risk HPV-16 oncoproteins
and fungal infection. can promote tumor angiogenesis possibly via HIF-1α. Tang et al.
After the discovery of resveratrol pronounced pharmacological ef- [154] revealed that overexpression of HPV-16 E6 and E7 oncoproteins
fects, it became a topic of intense investigations. It has been proposed significantly promoted HIF-1α protein accumulation and VEGF ex-
to prevent or counteract cardiovascular diseases (result from a mod- pression in human cervical cancer cells. However activation of HIF-
erate wine drinking known as “The French Paradox”) [131, 132] and 1α and VEGF can be reverted by resveratrol through the inhibition
inhibit cellular events associated with tumor initiation, promotion of AKT and ERK1/2 in cervical cancer cells, conceivably via the PI3K/
and progression as a result of anti-inflammatory, proapoptotic, anti- AKT and ERK1/2 signaling pathways. These results suggest that res-
angiogenic, chemo-, and radio-sensitizing properties [132–139] veratrol can suppress tumor angiogenic activity in vitro and thereby
(Fig. 2). Numerous reports also indicate that resveratrol has antiviral tumor progression and proliferation. Recently Hsu et al. [155] indicat-
effects against HIV-1 and the herpes simplex virus [140, 141]. The ed that resveratrol induces autophagy, inhibits proliferation and in-
high concentration of resveratrol in grape skin (50–100 mg/g) and duces apoptotic death in HeLa and SiHa cervical cancer cell lines.
thereby the high amounts in red wine (from 0.2 mg/l to 7.7 mg/l) The authors showed that resveratrol destabilize lysosomes, which
suggests that wine consumption might be protective against several cause lysosomal leakage, increased cytosol translocation, and enzymatic
pathologies. However, the amount of resveratrol consumed daily by activity of cathepsin L (cat L). The increased cat L activity in cytosol in-
a person drinking one glass of red wine is considerably lower than duces cytochrome c release from mitochondria, and subsequently re-
the dose needed to benefit of resveratrol activity as antitumoral sults in apoptotic cell death. An essential event in the pathway is the
agent [131, 142]. Studies focused on resveratrol anti-tumor activity lysosomal membrane permeabilization led by resveratrol induced
showed that this multi-hit compound exerts its action interacting autophagy and the release of lysosomal proteases to the cytosol. The re-
with numerous molecular targets involved in tumor progression, leased cathepsins may trigger caspase-dependent cell death through
such as Fas pathway, Rb-E2F/DP pathway, NF-κB and AP-1 transcrip- cleavage of bid and antiapoptotic Bcl-2 homologues, and induce cyto-
tional factors, MAPK and many others [142]. However just few of chrome c release.
them were investigated in cervical cancer (Table 1). Recently a num- Studies by Rezk et al. [156] on the chemotherapic use of resvera-
ber of studies were conducted to investigate the effects of resveratrol trol showed that this compound used with cisplatin or doxorubicin
intake during cervical cancer development and the results are demonstrated an additive growth-inhibitory anticancer effect on
reviewed below. Zoberi et al. [60] showed that resveratrol pretreat- uterine cancer cells supporting its possible use in the treatment of
ment, in cervical cancer cell lines, has the ability to inhibit cell cervical cancer.

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
8 F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx

In conclusion, all these studies demonstrated that resveratrol is ca- infection and its oncogenic expression. However, apart from the cur-
pable of: regulating cell cycle, progression, suppressing angiogenesis, rent pharmacological intervention it becomes essential to explore al-
inhibiting MMP activities, inducing autophagy with consequent pro- ternative approach. In recent years there has been increasing interest
grammed cell death and increasing IR-induced cytotoxicity (Table 1). in the potential cancer chemopreventive properties of diet-derived
These properties strongly support the beneficial effects of resveratrol agents. Among these, polyphenolic compounds, such as curcumin,
as a chemopreventive, a chemotherapeutic and a radiosensitizing com- resveratrol and ECGC, received much attention for prevention and
pound in the treatment of high-risk HPV-associated human cervical treatment of cervical cancer.
cancers as well as other tumors [157]. However supplementary studies Polyphenols were demonstrated to inhibit the proliferation of
to elucidate all the molecular mechanisms activated by resveratrol HPV-immortalized and HPV-positive cancer cells, through induction
treatment during cervical cancer are needed. Moreover, the lack of spe- of apoptosis, growth arrest, inhibition of DNA synthesis and modula-
cific clinical trials to prove the efficiency of resveratrol for the treatment tion of signal transduction pathways. They are able to act at several
of cervical cancer still raises the question about its real suitability. steps in the cascade of cell transformation promoted by HPV infec-
tion. However, further studies will likely provide additional insights
6. Clinical trial in cervical cancer into the mechanism of redox control of cancerogenesis events.
From the studies reviewed in this paper, it is clear that dietary
The effects and molecular mechanisms of action of curcumin, polyphenols hold a great potential in the prevention and therapy of
EGCG and resveratrol clearly sustain the chemopreventive and che- cervical cancer. Encouraging data as a safe and effective antitumoral
motherapic properties of these phenolic compounds, as well as their and chemo- and radiosensitizer agent have been demonstrated in
chemosensitizer and radiosensitizer abilities in cervical cancer clinical trials. Further, considering that curcumins, EGCG and resvera-
models. However, despite the confirmed beneficial effect of these an- trol present peculiar targets of action the synergistic use of these
tioxidant compounds on cervical cancer progression, only few clinical polyphenols might favor their anticarcinogenic effect, however no
studies have been conducted until now. such studies has been conducted yet on cervical cancer models. In ad-
Cheng et al. in 2001 [76] piloted a prospective phase-I study to dition, they also prevent depression, fatigue, lack of appetite and most
evaluate anticancer properties and also toxicology, pharmacokinetics of the symptoms induced by cancer.
and biologically effective dose of curcumin in patients with uterine
CIN. The study demonstrated that curcumin is not toxic to humans
up to 8 mg/d when taken by mouth for 3 months and suggested a bi- References
ologic effect of curcumin in the chemoprevention of cancer lesions at
[1] H. zur Hausen, W. Meinhof, W. Scheiber, G.W. Bornkamm, Attempts to detect
least in 25% of the cases under treatment. A curcumin testing started
virus-specific DNA in human tumors. I. Nucleic acid hybridizations with
in 2008 for the investigation of its safety and efficacy against HPV in- complementary RNA of human wart virus, Int. J. Cancer 13 (1974) 650–656.
fection, in a Phase II multicentric clinical trial on 280 women enrolled [2] M. Narisawa-Saito, T. Kiyono, Basic mechanisms of high-risk human
on the basis of clinical examination, cytology, histo-pathology and papillomavirus-induced carcinogenesis: roles of E6 and E7 proteins, Cancer
Sci. 98 (2007) 1505–1511.
presence of HPV [158]. [3] A.C. Bharti, S. Shukla, S. Mahata, S. Hedau, B.C. Das, Anti-human papillomavirus
Ahn et al. in 2003 [159] investigated clinical efficacy of EGCG and an- therapeutics: facts & future, Indian J. Med. Res. 130 (2009) 296–310.
other green tea component, poly E, delivered vaginally, orally, or both, [4] J.P. Baak, A.J. Kruse, S.J. Robboy, E.A. Janssen, B. van Diermen, I. Skaland, Dynamic
behavioural interpretation of cervical intraepithelial neoplasia with molecular
in patients with HPV-infected cervical lesions. Fifty-one patients with biomarkers, J. Clin. Pathol. 59 (2006) 1017–1028.
such lesions, ranging from chronic cervicitis through mild and moderate [5] N.A. Hamid, C. Brown, K. Gaston, The regulation of cell proliferation by the pap-
dysplasia to severe dysplasia, were divided into four groups and com- illomavirus early proteins, Cell Mol. Life Sci. 66 (2009) 1700–1717.
[6] C.A. Moody, L.A. Laimins, Human papillomavirus oncoproteins: pathways to
pared with 39 controls. For oral delivery, a 200 mg of EGCG or Poly E transformation, Nat. Rev. Cancer 10 (2010) 550–560.
capsules was taken every day for eight to 12 weeks while Poly E oint- [7] R.S. Jayshree, A. Sreenivas, M. Tessy, S. Krishna, Cell intrinsic & extrinsic factors
ment was applied locally to 27 patients twice a week. The 60% of sub- in cervical carcinogenesis, Indian J. Med. Res. 130 (2009) 286–295.
[8] M. Scheffner, J.M. Huibregtse, R.D. Vierstra, P.M. Howley, The HPV-16 E6 and E6-AP
jects under EGCG capsule therapy, the 50% under poly E capsule
complex functions as a ubiquitin-protein ligase in the ubiquitination of p53, Cell 75
therapy, the 74% under poly E ointment therapy and the 75% under (1993) 495–505.
poly E ointment plus poly E capsule therapy showed a response. Overall, [9] P. Massimi, A. Shai, P. Lambert, L. Banks, HPV E6 degradation of p53 and PDZ
containing substrates in an E6AP null background, Oncogene 27 (2008)
a 69% response rate was noted for treatment with green tea extracts, as
1800–1804.
compared with a 10% response rate in untreated controls. These results [10] M. Vogt, K. Butz, S. Dymalla, J. Semzow, F. Hoppe-Seyler, Inhibition of Bax activity is
indicate that green tea extracts used orally and/or vaginally are effective crucial for the antiapoptotic function of the human papillomavirus E6 oncoprotein,
for treating HPV-related cervical lesions suggesting that green tea ex- Oncogene 25 (2006) 4009–4015.
[11] M. Thomas, L. Banks, Inhibition of Bak-induced apoptosis by HPV-18 E6, Oncogene
tracts can be a potential therapy routine for patients with HPV infected 17 (1998) 2943–2954.
cervical lesions [12] M.A. James, J.H. Lee, A.J. Klingelhutz, Human papillomavirus type 16 E6 activates
As previously stated, despite the promising results in preclinical NF-kappaB, induces cIAP-2 expression, and protects against apoptosis in a PDZ
binding motif-dependent manner, J. Virol. 80 (2006) 5301–5307.
models, clinical trials, focusing on the health promoting effect of res- [13] H. Yuan, F. Fu, J. Zhuo, W. Wang, J. Nishitani, D.S. An, I.S. Chen, X. Liu, Human
veratrol, in subjects with cervical neoplasia are currently missing papillomavirus type 16 E6 and E7 oncoproteins upregulate c-IAP2 gene expres-
[160]. To date most of the available clinical studies in humans are fo- sion and confer resistance to apoptosis, Oncogene 24 (2005) 5069–5078.
[14] N. Dyson, The regulation of E2F by pRB-family proteins, Genes Dev. 12 (1998)
cused on resveratrol bioavailability, pharmacokinetics and metabolism 2245–2262.
[161] and so far only two recently reports showed that resveratrol [15] M. Giarre, S. Caldeira, I. Malanchi, F. Ciccolini, M.J. Leao, M. Tommasino, Induc-
could represent a promising agent in the treatment of cancerogenesis tion of pRb degradation by the human papillomavirus type 16 E7 protein is es-
sential to efficiently overcome p16INK4a-imposed G1 cell cycle arrest, J. Virol.
[162, 163]. 75 (2001) 4705–4712.
[16] R. Balan, C. Amalinei, S.E. Giusca, D. Ditescu, V. Gheorghita, E. Crauciuc, I.D.
7. Conclusions Caruntu, Immunohistochemical evaluation of COX-2 expression in HPV-
positive cervical squamous intraepithelial lesions, Rom. J. Morphol. Embryol.
52 (2011) 39–43.
Cervical cancer is the second most common cancer among women [17] K. Subbaramaiah, A.J. Dannenberg, Cyclooxygenase-2 transcription is regulated
worldwide. Thus, it still represents a major concern for public health by human papillomavirus 16 E6 and E7 oncoproteins: evidence of a corepres-
although current screening methods are already available to allow sor/coactivator exchange, Cancer Res. 67 (2007) 3976–3985.
[18] S.H. Kim, J.M. Oh, J.H. No, Y.J. Bang, Y.S. Juhnn, Y.S. Song, Involvement of NF-kappaB
early diagnosis and treatment. Several therapeutic strategies are and AP-1 in COX-2 upregulation by human papillomavirus 16 E5 oncoprotein, Carci-
being explored which can effectively target various phases of viral nogenesis 30 (2009) 753–757.

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx 9

[19] S.R. Sudarshan, R. Schlegel, X. Liu, The HPV-16 E5 protein represses expression [51] M.B. Sporn, Carcinogenesis and cancer: different perspectives on the same dis-
of stress pathway genes XBP-1 and COX-2 in genital keratinocytes, Biochem. ease, Cancer Res. 51 (1991) 6215–6218.
Biophys. Res. Commun. 399 (2010) 617–622. [52] R. Hu, S. Zhou, X. Li, Altered Bcl-2 and Bax expression is associated with cultured
[20] A. Moktar, R. Singh, M.V. Vadhanam, S. Ravoori, J.W. Lillard, C.G. Gairola, R.C. first trimester human cytotrophoblasts apoptosis induced by hypoxia, Life Sci.
Gupta, Cigarette smoke condensate-induced oxidative DNA damage and its re- 79 (2006) 351–355.
moval in human cervical cancer cells, Int. J. Oncol. 39 (2011) 941–947. [53] W.S. Jeong, Y.S. Keum, C. Chen, M.R. Jain, G. Shen, J.H. Kim, W. Li, A.N. Kong, Dif-
[21] S.S. Beevi, M.H. Rasheed, A. Geetha, Evidence of oxidative and nitrosative stress ferential expression and stability of endogenous nuclear factor E2-related factor
in patients with cervical squamous cell carcinoma, Clin. Chim. Acta 375 (2007) 2 (Nrf2) by natural chemopreventive compounds in HepG2 human hepatoma
119–123. cells, J. Biochem. Mol. Biol. 38 (2005) 167–176.
[22] T. Finkel, Oxidant signals and oxidative stress, Curr. Opin. Cell Biol. 15 (2003) [54] S. Nair, C. Xu, G. Shen, V. Hebbar, A. Gopalakrishnan, R. Hu, M.R. Jain, W. Lin, Y.S.
247–254. Keum, C. Liew, J.Y. Chan, A.N. Kong, Pharmacogenomics of phenolic antioxidant
[23] D. Darr, I. Fridovich, Free radicals in cutaneous biology, J. Invest. Dermatol. 102 butylated hydroxyanisole (BHA) in the small intestine and liver of Nrf2 knock-
(1994) 671–675. out and C57BL/6J mice, Pharm. Res. 23 (2006) 2621–2637.
[24] B.A. Jurkiewicz, G.R. Buettner, Ultraviolet light-induced free radical formation in skin: [55] G. Shen, W.S. Jeong, R. Hu, A.N. Kong, Regulation of Nrf2, NF-kappaB, and AP-1
an electron paramagnetic resonance study, Photochem. Photobiol. 59 (1994) 1–4. signaling pathways by chemopreventive agents, Antioxid. Redox Signal. 7
[25] K.B. Beckman, B.N. Ames, Oxidative decay of DNA, J. Biol. Chem. 272 (1997) (2005) 1648–1663.
19633–19636. [56] G. Shen, C. Xu, R. Hu, M.R. Jain, A. Gopalkrishnan, S. Nair, M.T. Huang, J.Y. Chan,
[26] J. Cadet, T. Delatour, T. Douki, D. Gasparutto, J.P. Pouget, J.L. Ravanat, S. Sauvaigo, A.N. Kong, Modulation of nuclear factor E2-related factor 2-mediated gene ex-
Hydroxyl radicals and DNA base damage, Mutat. Res. 424 (1999) 9–21. pression in mice liver and small intestine by cancer chemopreventive agent cur-
[27] M.M. Vilenchik, A.G. Knudson, Endogenous DNA double-strand breaks: produc- cumin, Mol. Cancer Ther. 5 (2006) 39–51.
tion, fidelity of repair, and induction of cancer, Proc. Natl. Acad. Sci. U. S. A. 100 [57] E.M. Siegel, J.L. Salemi, L.L. Villa, A. Ferenczy, E.L. Franco, A.R. Giuliano, Dietary
(2003) 12871–12876. consumption of antioxidant nutrients and risk of incident cervical intraepithelial
[28] J.M. Mates, C. Perez-Gomez, I. Nunez de Castro, Antioxidant enzymes and human neoplasia, Gynecol. Oncol. 118 (2010) 289–294.
diseases, Clin. Biochem. 32 (1999) 595–603. [58] L.Y. Tomita, A. Longatto Filho, M.C. Costa, M.A. Andreoli, L.L. Villa, E.L. Franco, M.A.
[29] S. Maynard, S.H. Schurman, C. Harboe, N.C. de Souza-Pinto, V.A. Bohr, Base exci- Cardoso, Brazilian Investigation into Nutrition and Cervical Cancer Prevention
sion repair of oxidative DNA damage and association with cancer and aging, Car- (BRINCA) Study Team, Diet and serum micronutrients in relation to cervical neoplasia
cinogenesis 30 (2009) 2–10. and cancer among low-income Brazilian women, Int. J. Cancer 126 (2010) 703–714.
[30] P. Storz, Reactive oxygen species in tumor progression, Front. Biosci. 10 (2005) [59] A. Goel, B.B. Aggarwal, Curcumin, the golden spice from Indian saffron, is a che-
1881–1896. mosensitizer and radiosensitizer for tumors and chemoprotector and radiopro-
[31] R.H. Burdon, Superoxide and hydrogen peroxide in relation to mammalian cell tector for normal organs, Nutr. Cancer 62 (2010) 919–930.
proliferation, Free Radic. Biol. Med. 18 (1995) 775–794. [60] I. Zoberi, C.M. Bradbury, H.A. Curry, K.S. Bisht, P.C. Goswami, J.L. Roti Roti, D.
[32] S. Toyokuni, K. Okamoto, J. Yodoi, H. Hiai, Persistent oxidative stress in cancer, Gius, Radiosensitizing and anti-proliferative effects of resveratrol in two
FEBS Lett. 358 (1995) 1–3. human cervical tumor cell lines, Cancer Lett. 175 (2002) 165–173.
[33] T.B. Kryston, A.B. Georgiev, P. Pissis, A.G. Georgakilas, Role of oxidative stress [61] I. Brouet, H. Ohshima, Curcumin, an anti-tumour promoter and anti-inflammatory
and DNA damage in human carcinogenesis, Mutat. Res. 711 (2011) 193–201. agent, inhibits induction of nitric oxide synthase in activated macrophages, Bio-
[34] G.Y. Liou, P. Storz, Reactive oxygen species in cancer, Free. Radic. Res. 44 (2010) chem. Biophys. Res. Commun. 206 (1995) 533–540.
479–496. [62] Sreejayan, M.N. Rao, Nitric oxide scavenging bCBEy curcuminoids, J. Pharm.
[35] A. Sgambato, G.F. Zannoni, B. Faraglia, A. Camerini, E. Tarquini, D. Spada, A. Pharmacol. 49 (1997) 105–107.
Cittadini, Decreased expression of the CDK inhibitor p27Kip1 and increased [63] R. Srivastava, M. Dikshit, R.C. Srimal, B.N. Dhawan, Anti-thrombotic effect of cur-
oxidative DNA damage in the multistep process of cervical carcinogenesis, cumin, Thromb. Res. 40 (1985) 413–417.
Gynecol. Oncol. 92 (2004) 776–783. [64] N. Venkatesan, Curcumin attenuation of acute adriamycin myocardial toxicity in
[36] M. Perluigi, A. Giorgi, C. Blarzino, F. De Marco, C. Foppoli, F. Di Domenico, D.A. rats, Br. J. Pharmacol. 124 (1998) 425–427.
Butterfield, M.E. Schinina, C. Cini, R. Coccia, Proteomics analysis of protein ex- [65] S.D. Deodhar, R. Sethi, R.C. Srimal, Preliminary study on antirheumatic activity of
pression and specific protein oxidation in human papillomavirus transformed curcumin (diferuloyl methane), Indian J. Med. Res. 71 (1980) 632–634.
keratinocytes upon UVB irradiation, J. Cell. Mol. Med. 13 (2009) 1809–1822. [66] M.M. Chan, N.S. Adapala, D. Fong, Curcumin overcomes the inhibitory effect of
[37] R.A. Cairns, I.S. Harris, T.W. Mak, Regulation of cancer cell metabolism, Nat. Rev. nitric oxide on Leishmania, Parasitol. Res. 96 (2005) 49–56.
Cancer 11 (2011) 85–95. [67] A. Khafif, S. Lev-Ari, A. Vexler, I. Barnea, A. Starr, V. Karaush, S. Haif, R. Ben-Yosef,
[38] F. De Marco, M. Perluigi, C. Foppoli, C. Blarzino, C. Cini, R. Coccia, A. Venuti, UVB Curcumin: a potential radio-enhancer in head and neck cancer, Laryngoscope
irradiation down-regulates HPV-16 RNA expression: implications for malignant 119 (2009) 2019–2026.
progression of transformed cells, Virus Res. 130 (2007) 249–259. [68] J.K. Lin, S.Y. Lin-Shiau, Mechanisms of cancer chemoprevention by curcumin,
[39] M.L. Looi, A.Z. Mohd Dali, S.A. Md Ali, W.Z. Wan Ngah, Y.A. Mohd Yusof, Oxida- Proc. Natl. Sci. Counc. Repub. China B 25 (2001) 59–66.
tive damage and antioxidant status in patients with cervical intraepithelial neo- [69] G. Kang, P.J. Kong, Y.J. Yuh, S.Y. Lim, S.V. Yim, W. Chun, S.S. Kim, Curcumin sup-
plasia and carcinoma of the cervix, Eur. J. Cancer Prev. 17 (2008) 555–560. presses lipopolysaccharide-induced cyclooxygenase-2 expression by inhibiting
[40] S.Y. Kim, J.W. Kim, Y.S. Ko, J.E. Koo, H.Y. Chung, Y.C. Lee-Kim, Changes in lipid activator protein 1 and nuclear factor kappab bindings in BV2 microglial cells,
peroxidation and antioxidant trace elements in serum of women with cervical J. Pharmacol. Sci. 94 (2004) 325–328.
intraepithelial neoplasia and invasive cancer, Nutr. Cancer 47 (2003) 126–130. [70] S.S. Han, Y.S. Keum, H.J. Seo, Y.J. Surh, Curcumin suppresses activation of NF-
[41] T.L. Goncalves, F. Erthal, C.L. Corte, L.G. Muller, C.M. Piovezan, C.W. Nogueira, J.B. kappaB and AP-1 induced by phorbol ester in cultured human promyelocytic
Rocha, Involvement of oxidative stress in the pre-malignant and malignant leukemia cells, J. Biochem. Mol. Biol. 35 (2002) 337–342.
states of cervical cancer in women, Clin. Biochem. 38 (2005) 1071–1075. [71] J.Y. Liu, S.J. Lin, J.K. Lin, Inhibitory effects of curcumin on protein kinase C activity
[42] S.C. Thomasset, D.P. Berry, G. Garcea, T. Marczylo, W.P. Steward, A.J. Gescher, induced by 12-O-tetradecanoyl-phorbol-13-acetate in NIH 3T3 cells, Carcino-
Dietary polyphenolic phytochemicals—promising cancer chemopreventive agents genesis 14 (1993) 857–861.
in humans? A review of their clinical properties, Int. J. Cancer 120 (2007) 451–458. [72] S.M. Plummer, K.A. Holloway, M.M. Manson, R.J. Munks, A. Kaptein, S. Farrow, L.
[43] M. D'Archivio, C. Santangelo, B. Scazzocchio, R. Vari, C. Filesi, R. Masella, C. Howells, Inhibition of cyclo-oxygenase 2 expression in colon cells by the chemo-
Giovannini, Modulatory effects of polyphenols on apoptosis induction: preventive agent curcumin involves inhibition of NF-kappaB activation via the
relevance for cancer prevention, Int. J. Mol. Sci. 9 (2008) 213–228. NIK/IKK signalling complex, Oncogene 18 (1999) 6013–6020.
[44] H.J. Forman, M. Torres, J. Fukuto, Redox signaling, Mol. Cell. Biochem. 234–235 [73] S.M. Plummer, K.A. Hill, M.F. Festing, W.P. Steward, A.J. Gescher, R.A. Sharma,
(2002) 49–62. Clinical development of leukocyte cyclooxygenase 2 activity as a systemic bio-
[45] S. Nair, W. Li, A.N. Kong, Natural dietary anti-cancer chemopreventive com- marker for cancer chemopreventive agents, Cancer Epidemiol. Biomarkers
pounds: redox-mediated differential signaling mechanisms in cytoprotection Prev. 10 (2001) 1295–1299.
of normal cells versus cytotoxicity in tumor cells, Acta Pharmacol. Sin. 28 [74] L.I. Lin, Y.F. Ke, Y.C. Ko, J.K. Lin, Curcumin inhibits SK-Hep-1 hepatocellular car-
(2007) 459–472. cinoma cell invasion in vitro and suppresses matrix metalloproteinase-9 secre-
[46] A. Gopalakrishnan, A.N. Tony Kong, Anticarcinogenesis by dietary phytochemi- tion, Oncology 55 (1998) 349–353.
cals: cytoprotection by Nrf2 in normal cells and cytotoxicity by modulation of [75] R. Mohan, J. Sivak, P. Ashton, L.A. Russo, B.Q. Pham, N. Kasahara, M.B. Raizman,
transcription factors NF-kappa B and AP-1 in abnormal cancer cells, Food M.E. Fini, Curcuminoids inhibit the angiogenic response stimulated by fibroblast
Chem. Toxicol. 46 (2008) 1257–1270. growth factor-2, including expression of matrix metalloproteinase gelatinase B,
[47] J.M. Hansen, W.H. Watson, D.P. Jones, Compartmentation of Nrf-2 redox control: J. Biol. Chem. 275 (2000) 10405–10412.
regulation of cytoplasmic activation by glutathione and DNA binding by [76] A.L. Cheng, C.H. Hsu, J.K. Lin, M.M. Hsu, Y.F. Ho, T.S. Shen, J.Y. Ko, J.T. Lin, B.R. Lin,
thioredoxin-1, Toxicol. Sci. 82 (2004) 308–317. W. Ming-Shiang, H.S. Yu, S.H. Jee, G.S. Chen, T.M. Chen, C.A. Chen, M.K. Lai, Y.S.
[48] M. Karin, The regulation of AP-1 activity by mitogen-activated protein kinases, Pu, M.H. Pan, Y.J. Wang, C.C. Tsai, C.Y. Hsieh, Phase I clinical trial of curcumin,
J. Biol. Chem. 270 (1995) 16483–16486. a chemopreventive agent, in patients with high-risk or pre-malignant lesions,
[49] J.R. Matthews, W. Kaszubska, G. Turcatti, T.N. Wells, R.T. Hay, Role of cysteine62 Anticancer. Res. 21 (2001) 2895–2900.
in DNA recognition by the P50 subunit of NF-kappa B, Nucleic Acids Res. 21 [77] C.V. Rao, A. Rivenson, B. Simi, B.S. Reddy, Chemoprevention of colon carcinogen-
(1993) 1727–1734. esis by dietary curcumin, a naturally occurring plant phenolic compound, Can-
[50] J.M. Hansen, Y.M. Go, D.P. Jones, Nuclear and mitochondrial compartmentation cer Res. 55 (1995) 259–266.
of oxidative stress and redox signaling, Annu. Rev. Pharmacol. Toxicol. 46 [78] T. Kawamori, R. Lubet, V.E. Steele, G.J. Kelloff, R.B. Kaskey, C.V. Rao, B.S. Reddy,
(2006) 215–234. Chemopreventive effect of curcumin, a naturally occurring anti-inflammatory

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
10 F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx

agent, during the promotion/progression stages of colon cancer, Cancer Res. 59 [104] M.S. Baliga, S. Meleth, S.K. Katiyar, Growth inhibitory and antimetastatic effect of
(1999) 597–601. green tea polyphenols on metastasis-specific mouse mammary carcinoma 4T1
[79] M. Venkatraman, R.J. Anto, A. Nair, M. Varghese, D. Karunagaran, Biological and cells in vitro and in vivo systems, Clin. Cancer Res. 11 (2005) 1918–1927.
chemical inhibitors of NF-kappaB sensitize SiHa cells to cisplatin-induced apo- [105] T. Kondo, T. Ohta, K. Igura, Y. Hara, K. Kaji, Tea catechins inhibit angiogenesis in
ptosis, Mol. Carcinog. 44 (2005) 51–59. vitro, measured by human endothelial cell growth, migration and tube forma-
[80] S.V. Bava, V.T. Puliappadamba, A. Deepti, A. Nair, D. Karunagaran, R.J. Anto, Sen- tion, through inhibition of VEGF receptor binding, Cancer Lett. 180 (2002)
sitization of taxol-induced apoptosis by curcumin involves down-regulation of 139–144.
nuclear factor-kappaB and the serine/threonine kinase Akt and is independent [106] G. Fassina, R. Vene, M. Morini, S. Minghelli, R. Benelli, D.M. Noonan, A. Albini,
of tubulin polymerization, J. Biol. Chem. 280 (2005) 6301–6308. Mechanisms of inhibition of tumor angiogenesis and vascular tumor growth
[81] B.K. Prusty, B.C. Das, Constitutive activation of transcription factor AP-1 in cervi- by epigallocatechin-3-gallate, Clin. Cancer Res. 10 (2004) 4865–4873.
cal cancer and suppression of human papillomavirus (HPV) transcription and [107] S. Qanungo, M. Das, S. Haldar, A. Basu, Epigallocatechin-3-gallate induces mito-
AP-1 activity in HeLa cells by curcumin, Int. J. Cancer 113 (2005) 951–960. chondrial membrane depolarization and caspase-dependent apoptosis in pan-
[82] C.S. Divya, M.R. Pillai, Antitumor action of curcumin in human papillomavirus creatic cancer cells, Carcinogenesis 26 (2005) 958–967.
associated cells involves downregulation of viral oncogenes, prevention of [108] N. Ahmad, P. Cheng, H. Mukhtar, Cell cycle dysregulation by green tea polyphe-
NFkB and AP-1 translocation, and modulation of apoptosis, Mol. Carcinog. 45 nol epigallocatechin-3-gallate, Biochem. Biophys. Res. Commun. 275 (2000)
(2006) 320–332. 328–334.
[83] M. Singh, N. Singh, Molecular mechanism of curcumin induced cytotoxicity in [109] Y.C. Liang, S.Y. Lin-Shiau, C.F. Chen, J.K. Lin, Inhibition of cyclin-dependent ki-
human cervical carcinoma cells, Mol. Cell. Biochem. 325 (2009) 107–119. nases 2 and 4 activities as well as induction of Cdk inhibitors p21 and p27 during
[84] M. Singh, N. Singh, Curcumin counteracts the proliferative effect of estradiol and growth arrest of human breast carcinoma cells by (−)-epigallocatechin-3-gal-
induces apoptosis in cervical cancer cells, Mol. Cell. Biochem. 347 (2011) 1–11. late, J. Cell. Biochem. 75 (1999) 1–12.
[85] K. Madden, L. Flowers, R. Salani, I. Horowitz, S. Logan, K. Kowalski, J. Xie, S.I. [110] N. Ahmad, D.K. Feyes, A.L. Nieminen, R. Agarwal, H. Mukhtar, Green tea constit-
Mohammed, Proteomics-based approach to elucidate the mechanism of antitu- uent epigallocatechin-3-gallate and induction of apoptosis and cell cycle arrest
mor effect of curcumin in cervical cancer, Prostaglandins Leukot. Essent. Fatty in human carcinoma cells, J. Natl. Cancer Inst. 89 (1997) 1881–1886.
Acids 80 (2009) 9–18. [111] Z.P. Chen, J.B. Schell, C.T. Ho, K.Y. Chen, Green tea epigallocatechin gallate shows
[86] D.M. Maher, M.C. Bell, E.A. O'Donnell, B.K. Gupta, M. Jaggi, S.C. Chauhan, Curcu- a pronounced growth inhibitory effect on cancerous cells but not on their nor-
min suppresses human papillomavirus oncoproteins, restores p53, Rb, and mal counterparts, Cancer Lett. 129 (1998) 173–179.
PTPN13 proteins and inhibits benzo[a]pyrene-induced upregulation of HPV E7, [112] F. Nanjo, M. Honda, K. Okushio, N. Matsumoto, F. Ishigaki, T. Ishigami, Y. Hara,
Mol. Carcinog. 50 (2011) 47–57. Effects of dietary tea catechins on alpha-tocopherol levels, lipid peroxidation,
[87] P. Javvadi, A.T. Segan, S.W. Tuttle, C. Koumenis, The chemopreventive agent cur- and erythrocyte deformability in rats fed on high palm oil and perilla oil diets,
cumin is a potent radiosensitizer of human cervical tumor cells via increased re- Biol. Pharm. Bull. 16 (1993) 1156–1159.
active oxygen species production and overactivation of the mitogen-activated [113] T. Nakagawa, T. Yokozawa, Direct scavenging of nitric oxide and superoxide by
protein kinase pathway, Mol. Pharmacol. 73 (2008) 1491–1501. green tea, Food Chem. Toxicol. 40 (2002) 1745–1750.
[88] P. Javvadi, L. Hertan, R. Kosoff, T. Datta, J. Kolev, R. Mick, S.W. Tuttle, C. Koumenis, [114] G.R. Haenen, J.B. Paquay, R.E. Korthouwer, A. Bast, Peroxynitrite scavenging by
Thioredoxin reductase-1 mediates curcumin-induced radiosensitization of flavonoids, Biochem. Biophys. Res. Commun. 236 (1997) 591–593.
squamous carcinoma cells, Cancer Res. 70 (2010) 1941–1950. [115] B.C. Scott, J. Butler, B. Halliwell, O.I. Aruoma, Evaluation of the antioxidant ac-
[89] C.N. Sreekanth, S.V. Bava, E. Sreekumar, R.J. Anto, Molecular evidences for the tions of ferulic acid and catechins, Free. Radic. Res. Commun. 19 (1993)
chemosensitizing efficacy of liposomal curcumin in paclitaxel chemotherapy in 241–253.
mouse models of cervical cancer, Oncogene 30 (2011) 3139–3152. [116] J.F. Sah, S. Balasubramanian, R.L. Eckert, E.A. Rorke, Epigallocatechin-3-gallate
[90] S.V. Bava, C.N. Sreekanth, A.K. Thulasidasan, N.P. Anto, V.T. Cheriyan, V.T. inhibits epidermal growth factor receptor signaling pathway. Evidence for direct
Puliyappadamba, S.G. Menon, S.D. Ravichandran, R.J. Anto, Akt is upstream and inhibition of ERK1/2 and AKT kinases, J. Biol. Chem. 279 (2004) 12755–12762.
MAPKs are downstream of NF-kappaB in paclitaxel-induced survival signaling [117] N. Khan, F. Afaq, M. Saleem, N. Ahmad, H. Mukhtar, Targeting multiple signaling
events, which are down-regulated by curcumin contributing to their synergism, pathways by green tea polyphenol (−)-epigallocatechin-3-gallate, Cancer Res.
Int. J. Biochem. Cell Biol. 43 (2011) 331–341. 66 (2006) 2500–2505.
[91] R. Sultana, A. Ravagna, H. Mohmmad-Abdul, V. Calabrese, D.A. Butterfield, Feru- [118] H.K. Na, Y.J. Surh, Intracellular signaling network as a prime chemopreventive
lic acid ethyl ester protects neurons against amyloid beta-peptide(1–42)-in- target of (−)-epigallocatechin gallate, Mol. Nutr. Food Res. 50 (2006)
duced oxidative stress and neurotoxicity: relationship to antioxidant activity, J. 152–159.
Neurochem. 92 (2005) 749–758. [119] W.S. Ahn, S.W. Huh, S.M. Bae, I.P. Lee, J.M. Lee, S.E. Namkoong, C.K. Kim, J.I. Sin, A
[92] H. Mohmmad Abdul, D.A. Butterfield, Protection against amyloid beta-peptide major constituent of green tea, EGCG, inhibits the growth of a human cervical
(1–42)-induced loss of phospholipid asymmetry in synaptosomal membranes cancer cell line, CaSki cells, through apoptosis, G(1) arrest, and regulation of
by tricyclodecan-9-xanthogenate (D609) and ferulic acid ethyl ester: implica- gene expression, DNA Cell Biol. 22 (2003) 217–224.
tions for Alzheimer's disease, Biochim. Biophys. Acta 1741 (2005) 140–148. [120] F. Asif Siddiqui, M. Naim, N. Islam, Apoptotic effect of green tea polyphenol
[93] G. Joshi, M. Perluigi, R. Sultana, R. Agrippino, V. Calabrese, D.A. Butterfield, In (EGCG) on cervical carcinoma cells, Diagn. Cytopathol. 39 (2011) 482–488.
vivo protection of synaptosomes by ferulic acid ethyl ester (FAEE) from oxida- [121] D.J. Kuhn, A.C. Burns, A. Kazi, Q.P. Dou, Direct inhibition of the ubiquitin-
tive stress mediated by 2,2-azobis(2-amidino-propane) dihydrochloride proteasome pathway by ester bond-containing green tea polyphenols is associ-
(AAPH) or Fe(2+)/H(2)O(2): insight into mechanisms of neuroprotection and ated with increased expression of sterol regulatory element-binding protein 2
relevance to oxidative stress-related neurodegenerative disorders, Neurochem. and LDL receptor, Biochim. Biophys. Acta 1682 (2004) 1–10.
Int. 48 (2006) 318–327. [122] M. Noguchi, M. Yokoyama, S. Watanabe, M. Uchiyama, Y. Nakao, K. Hara, T.
[94] M. Perluigi, G. Joshi, R. Sultana, V. Calabrese, C. De Marco, R. Coccia, C. Cini, D.A. Iwasaka, Inhibitory effect of the tea polyphenol, (−)-epigallocatechin gallate,
Butterfield, In vivo protective effects of ferulic acid ethyl ester against amyloid- on growth of cervical adenocarcinoma cell lines, Cancer Lett. 234 (2006)
beta peptide 1–42-induced oxidative stress, J. Neurosci. Res. 84 (2006) 418–426. 135–142.
[95] F. Di Domenico, M. Perluigi, C. Foppoli, C. Blarzino, R. Coccia, F. De Marco, D.A. [123] M. Yokoyama, M. Noguchi, Y. Nakao, A. Pater, T. Iwasaka, The tea polyphenol,
Butterfield, C. Cini, Protective effect of ferulic acid ethyl ester against oxidative (−)-epigallocatechin gallate effects on growth, apoptosis, and telomerase activ-
stress mediated by UVB irradiation in human epidermal melanocytes, Free. ity in cervical cell lines, Gynecol. Oncol. 92 (2004) 197–204.
Radic. Res. 43 (2009) 365–375. [124] Q. Zhang, X. Tang, Q. Lu, Z. Zhang, J. Rao, A.D. Le, Green tea extract and (−)-epi-
[96] B. Jayaprakasam, M. Vanisree, Y. Zhang, D.L. Dewitt, M.G. Nair, Impact of alkyl es- gallocatechin-3-gallate inhibit hypoxia- and serum-induced HIF-1alpha protein
ters of caffeic and ferulic acids on tumor cell proliferation, cyclooxygenase en- accumulation and VEGF expression in human cervical carcinoma and hepatoma
zyme, and lipid peroxidation, J. Agric. Food Chem. 54 (2006) 5375–5381. cells, Mol. Cancer Ther. 5 (2006) 1227–1238.
[97] L.F. Zheng, F. Dai, B. Zhou, L. Yang, Z.L. Liu, Prooxidant activity of hydroxycin- [125] O. Tudoran, O. Soritau, O. Balacescu, L. Balacescu, C. Braicu, M. Rus, C. Gherman,
namic acids on DNA damage in the presence of Cu (II) ions: mechanism and P. Virag, F. Irimie, I. Berindan-Neagoe, Early transcriptional pattern of angio-
structure–activity relationship, Food Chem. Toxicol. 46 (2008) 149–156. genesis induced by EGCG treatment in cervical tumor cells, J. Cell. Mol. Med.
[98] S. Karthikeyan, G. Kanimozhi, N.R. Prasad, R. Mahalakshmi, Radiosensitizing effect (2011).
of ferulic acid on human cervical carcinoma cells in vitro, Toxicol. In Vitro (2011). [126] M. Li, Z. He, S. Ermakova, D. Zheng, F. Tang, Y.Y. Cho, F. Zhu, W.Y. Ma, Y. Sham,
[99] G.P. Yu, C.C. Hsieh, L.Y. Wang, S.Z. Yu, X.L. Li, T.H. Jin, Green-tea consumption and E.A. Rogozin, A.M. Bode, Y. Cao, Z. Dong, Direct inhibition of insulin-like growth
risk of stomach cancer: a population-based case–control study in Shanghai, factor-I receptor kinase activity by (−)-epigallocatechin-3-gallate regulates cell
China, Cancer Causes Control 6 (1995) 532–538. transformation, Cancer Epidemiol. Biomarkers Prev. 16 (2007) 598–605.
[100] B.T. Ji, W.H. Chow, A.W. Hsing, J.K. McLaughlin, Q. Dai, Y.T. Gao, W.J. Blot, J.F. [127] J. Jacob, S. Cabarcas, I. Veras, N. Zaveri, L. Schramm, The green tea component
Fraumeni Jr., Green tea consumption and the risk of pancreatic and colorectal EGCG inhibits RNA polymerase III transcription, Biochem. Biophys. Res. Com-
cancers, Int. J. Cancer 70 (1997) 255–258. mun. 360 (2007) 778–783.
[101] H.N. Graham, Green tea composition, consumption, and polyphenol chemistry, [128] Y. Qiao, J. Cao, L. Xie, X. Shi, Cell growth inhibition and gene expression regula-
Prev. Med. 21 (1992) 334–350. tion by (−)-epigallocatechin-3-gallate in human cervical cancer cells, Arch.
[102] N. Horie, N. Hirabayashi, Y. Takahashi, Y. Miyauchi, H. Taguchi, K. Takeishi, Syn- Pharm. Res. 32 (2009) 1309–1315.
ergistic effect of green tea catechins on cell growth and apoptosis induction in [129] C. Zou, H. Liu, J.M. Feugang, Z. Hao, H.H. Chow, F. Garcia, Green tea compound in
gastric carcinoma cells, Biol. Pharm. Bull. 28 (2005) 574–579. chemoprevention of cervical cancer, Int. J. Gynecol. Cancer 20 (2010) 617–624.
[103] M. Nihal, N. Ahmad, H. Mukhtar, G.S. Wood, Anti-proliferative and proapoptotic ef- [130] L. Bonfili, M. Cuccioloni, M. Mozzicafreddo, V. Cecarini, M. Angeletti, A.M. Eleu-
fects of (−)-epigallocatechin-3-gallate on human melanoma: possible implica- teri, Identification of an EGCG oxidation derivative with proteasome modulatory
tions for the chemoprevention of melanoma, Int. J. Cancer 114 (2005) 513–521. activity, Biochimie 93 (2011) 931–940.

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005
F. Di Domenico et al. / Biochimica et Biophysica Acta xxx (2011) xxx–xxx 11

[131] M.P. Kramer, J. Wesierska-Gadek, Monitoring of long-term effects of resveratrol [149] E.T. Waas, R.M. Lomme, J. DeGroot, T. Wobbes, T. Hendriks, Tissue levels of active
on cell cycle progression of human HeLa cells after administration of a single matrix metalloproteinase-2 and − 9 in colorectal cancer, Br. J. Cancer 86 (2002)
dose, Ann. N. Y. Acad. Sci. 1171 (2009) 257–263. 1876–1883.
[132] A.Y. Sun, A. Simonyi, G.Y. Sun, The “French Paradox” and beyond: neuroprotec- [150] J.H. Woo, J.H. Lim, Y.H. Kim, S.I. Suh, D.S. Min, J.S. Chang, Y.H. Lee, J.W. Park, T.K.
tive effects of polyphenols, Free Radic. Biol. Med. 32 (2002) 314–318. Kwon, Resveratrol inhibits phorbol myristate acetate-induced matrix
[133] M. Jang, L. Cai, G.O. Udeani, K.V. Slowing, C.F. Thomas, C.W. Beecher, H.H. Fong, metalloproteinase-9 expression by inhibiting JNK and PKC delta signal transduc-
N.R. Farnsworth, A.D. Kinghorn, R.G. Mehta, R.C. Moon, J.M. Pezzuto, Cancer che- tion, Oncogene 23 (2004) 1845–1853.
mopreventive activity of resveratrol, a natural product derived from grapes, Sci- [151] E. Toussaint-Smith, D.B. Donner, A. Roman, Expression of human papillomavi-
ence 275 (1997) 218–220. rus type 16 E6 and E7 oncoproteins in primary foreskin keratinocytes is suffi-
[134] M. Alkhalaf, A. El-Mowafy, W. Renno, O. Rachid, A. Ali, R. Al-Attyiah, Resveratrol- cient to alter the expression of angiogenic factors, Oncogene 23 (2004)
induced apoptosis in human breast cancer cells is mediated primarily through 2988–2995.
the caspase-3-dependent pathway, Arch. Med. Res. 39 (2008) 162–168. [152] O. Lopez-Ocejo, A. Viloria-Petit, M. Bequet-Romero, D. Mukhopadhyay, J. Rak,
[135] M. Alkhalaf, Resveratrol-induced apoptosis is associated with activation of p53 R.S. Kerbel, Oncogenes and tumor angiogenesis: the HPV-16 E6 oncoprotein ac-
and inhibition of protein translation in T47D human breast cancer cells, Pharma- tivates the vascular endothelial growth factor (VEGF) gene promoter in a p53
cology 80 (2007) 134–143. independent manner, Oncogene 19 (2000) 4611–4620.
[136] E. Pozo-Guisado, J.M. Merino, S. Mulero-Navarro, M.J. Lorenzo-Benayas, F. [153] S.H. Kim, Y.S. Juhnn, S. Kang, S.W. Park, M.W. Sung, Y.J. Bang, Y.S. Song, Human
Centeno, A. Alvarez-Barrientos, P.M. Fernandez-Salguero, Resveratrol-induced papillomavirus 16 E5 up-regulates the expression of vascular endothelial
apoptosis in MCF-7 human breast cancer cells involves a caspase-independent growth factor through the activation of epidermal growth factor receptor,
mechanism with downregulation of Bcl-2 and NF-kappaB, Int. J. Cancer 115 MEK/ERK1,2 and PI3K/Akt, Cell. Mol. Life Sci. 63 (2006) 930–938.
(2005) 74–84. [154] X. Tang, Q. Zhang, J. Nishitani, J. Brown, S. Shi, A.D. Le, Overexpression of human
[137] H.Y. Tang, A. Shih, H.J. Cao, F.B. Davis, P.J. Davis, H.Y. Lin, Resveratrol-induced papillomavirus type 16 oncoproteins enhances hypoxia-inducible factor 1 alpha
cyclooxygenase-2 facilitates p53-dependent apoptosis in human breast cancer protein accumulation and vascular endothelial growth factor expression in
cells, Mol. Cancer Ther. 5 (2006) 2034–2042. human cervical carcinoma cells, Clin. Cancer Res. 13 (2007) 2568–2576.
[138] Y. Hu, C.Y. Sun, J. Huang, L. Hong, L. Zhang, Z.B. Chu, Antimyeloma effects of resvera- [155] K.F. Hsu, C.L. Wu, S.C. Huang, C.M. Wu, J.R. Hsiao, Y.T. Yo, Y.H. Chen, A.L. Shiau,
trol through inhibition of angiogenesis, Chin. Med. J. (Engl) 120 (2007) 1672–1677. C.Y. Chou, Cathepsin L mediates resveratrol-induced autophagy and apoptotic
[139] N. Ahmad, V.M. Adhami, F. Afaq, D.K. Feyes, H. Mukhtar, Resveratrol causes cell death in cervical cancer cells, Autophagy 5 (2009) 451–460.
WAF-1/p21-mediated G(1)-phase arrest of cell cycle and induction of apoptosis [156] Y.A. Rezk, S.S. Balulad, R.S. Keller, J.A. Bennett, Use of resveratrol to improve the
in human epidermoid carcinoma A431 cells, Clin. Cancer Res. 7 (2001) effectiveness of cisplatin and doxorubicin: study in human gynecologic cancer
1466–1473. cell lines and in rodent heart, Am. J. Obstet. Gynecol. 194 (2006) e23–e26.
[140] A. Heredia, C. Davis, R. Redfield, Synergistic inhibition of HIV-1 in activated and [157] S.C. Gupta, R. Kannappan, S. Reuter, J.H. Kim, B.B. Aggarwal, Chemosensitization
resting peripheral blood mononuclear cells, monocyte-derived macrophages, and of tumors by resveratrol, Ann. N. Y. Acad. Sci. 1215 (2011) 150–160.
selected drug-resistant isolates with nucleoside analogues combined with a natu- [158] G.P. Talwar, A clinical study on the clearance of human papilloma virus (HPV)
ral product, resveratrol, J. Acquir. Immune Defic. Syndr. 25 (2000) 246–255. infection in uterine cervix by Basant (a polyherbal cream) and curcumin soft
[141] J.J. Docherty, M.M. Fu, B.S. Stiffler, R.J. Limperos, C.M. Pokabla, A.L. DeLucia, Resvera- gelatin capsule in females infected with HPV Clinical Trial Registry — India, Na-
trol inhibition of herpes simplex virus replication, Antiviral Res. 43 (1999) 145–155. tional Institute of Medical Statistics (ICMR, India, 2008 CTRI/2008/091/000095).
[142] B.B. Aggarwal, A. Bhardwaj, R.S. Aggarwal, N.P. Seeram, S. Shishodia, Y. Takada, [159] W.S. Ahn, J. Yoo, S.W. Huh, C.K. Kim, J.M. Lee, S.E. Namkoong, S.M. Bae, I.P. Lee,
Role of resveratrol in prevention and therapy of cancer: preclinical and clinical Protective effects of green tea extracts (polyphenon E and EGCG) on human cer-
studies, Anticancer. Res. 24 (2004) 2783–2840. vical lesions, Eur. J. Cancer Prev. 12 (2003) 383–390.
[143] K.B. Harikumar, B.B. Aggarwal, Resveratrol: a multitargeted agent for age- [160] O. Vang, N. Ahmad, C.A. Baile, J.A. Baur, K. Brown, A. Csiszar, D.K. Das, D. Delmas,
associated chronic diseases, Cell Cycle 7 (2008) 1020–1035. C. Gottfried, H.Y. Lin, Q.Y. Ma, P. Mukhopadhyay, N. Nalini, J.M. Pezzuto, T. Rich-
[144] E. Sexton, C. Van Themsche, K. LeBlanc, S. Parent, P. Lemoine, E. Asselin, Resver- ard, Y. Shukla, Y.J. Surh, T. Szekeres, T. Szkudelski, T. Walle, J.M. Wu, What is new
atrol interferes with AKT activity and triggers apoptosis in human uterine cancer for an old molecule? Systematic review and recommendations on the use of res-
cells, Mol. Cancer 5 (2006) 45. veratrol, PLoS One 6 (2011) e19881.
[145] H.Y. Lin, M. Sun, H.Y. Tang, T.M. Simone, Y.H. Wu, J.R. Grandis, H.J. Cao, P.J. Davis, [161] K.R. Patel, E. Scott, V.A. Brown, A.J. Gescher, W.P. Steward, K. Brown, Clinical tri-
F.B. Davis, Resveratrol causes COX-2- and p53-dependent apoptosis in head and als of resveratrol, Ann. N. Y. Acad. Sci. 1215 (2011) 161–169.
neck squamous cell cancer cells, J. Cell. Biochem. 104 (2008) 2131–2142. [162] K.R. Patel, V.A. Brown, D.J. Jones, R.G. Britton, D. Hemingway, A.S. Miller, K.P.
[146] C. Lin, D.R. Crawford, S. Lin, J. Hwang, A. Sebuyira, R. Meng, J.E. Westfall, H.Y. West, T.D. Booth, M. Perloff, J.A. Crowell, D.E. Brenner, W.P. Steward, A.J.
Tang, P.Y. Yu, P.J. Davis, H.Y. Lin, Inducible COX-2-dependent apoptosis in Gescher, K. Brown, Clinical pharmacology of resveratrol and its metabolites in
human ovarian cancer cells, Carcinogenesis 32 (2011) 19–26. colorectal cancer patients, Cancer Res. 70 (2010) 7392–7399.
[147] H.Y. Lin, H.Y. Tang, F.B. Davis, P.J. Davis, Resveratrol and apoptosis, Ann. N. Y. [163] V.A. Brown, K.R. Patel, M. Viskaduraki, J.A. Crowell, M. Perloff, T.D. Booth, G.
Acad. Sci. 1215 (2011) 79–88. Vasilinin, A. Sen, A.M. Schinas, G. Piccirilli, K. Brown, W.P. Steward, A.J. Gescher,
[148] P.P. Lee, J.J. Hwang, G. Murphy, M.M. Ip, Functional significance of MMP-9 in D.E. Brenner, Repeat dose study of the cancer chemopreventive agent resvera-
tumor necrosis factor-induced proliferation and branching morphogenesis of trol in healthy volunteers: safety, pharmacokinetics, and effect on the insulin-
mammary epithelial cells, Endocrinology 141 (2000) 3764–3773. like growth factor axis, Cancer Res. 70 (2010) 9003–9011.

Please cite this article as: F. Di Domenico, et al., Antioxidants in cervical cancer: Chemopreventive and chemotherapeutic effects of polyphe-
nols, Biochim. Biophys. Acta (2011), doi:10.1016/j.bbadis.2011.10.005

You might also like