REVIEW - Manzo-Merino Et Al 2014 - The Role of Signaling Pathways in Cervical Cancer and Molecular Therapeutic Targets

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Archives of Medical Research 45 (2014) 525e539

REVIEW ARTICLE
The Role of Signaling Pathways in Cervical Cancer and Molecular
Therapeutic Targets
Joaquın Manzo-Merino,a Adriana Contreras-Paredes,a Elenae Vazquez-Ulloa,a Leticia Rocha-Zavaleta,b
Alma M. Fuentes-Gonzalez,a and Marcela Lizanoa
a
Unidad de Investigacion Biomedica en Cancer, Instituto Nacional de Cancerologıa-Instituto de Investigaciones Biomedicas, Universidad Nacional
Autonoma de Mexico, Mexico City, Mexico
b
Departamento de Biologıa Molecular y Biotecnologıa, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico,
Mexico City, Mexico
Received for publication August 8, 2014; accepted October 29, 2014 (ARCMED-D-14-00436).

Cervical cancer is a public health issue in developing countries. Although the Pap smear
and colposcopy remain the major strategies for detection, most cases are diagnosed in the
late stages. Therefore, a major concern has been to develop early diagnostic approaches
and more effective treatments. Molecular pathways that participate in cervical malignant
transformation have emerged as promising directed therapeutic targets. In this review, we
explore some of the major pathways implicated in cervical cancer development, including
RAF/MEK/ERK, phosphatidylinositol-3 kinase (PI3K/AKT), Wnt/b-catenin, apoptosis
and coupled membrane receptor signaling. We focus on the role of these pathways in
cervical carcinogenesis, their alterations and the consequences of these abnormalities.
In addition, the most recent preclinical and clinical data on the rationally designed
target-based agents that are currently being tested against elements of these pathways
are reviewed. Ó 2014 IMSS. Published by Elsevier Inc.
Key Words: Cervical cancer, Carcinogenesis, Signaling pathways, Inhibitors.

Introduction important cell cycle regulators such as p53 and PDZ domain
containing proteins (4), whereas E7 promotes the inactivation
Cervical cancer is the fourth most common neoplasm in
of pRb (5). In addition, other proteins are overexpressed in
women worldwide. Every year, nearly 529,000 new cases
this neoplasm such as PI3K (phosphatidylinositol 3-kinase),
of cervical cancer are diagnosed. Most cases are reported
EGF-R, b-catenin, and Erk, and anti-apoptotic proteins such
in developing countries where cervical cancer is one of
as the Bcl-2 sub-family (6e9). All of these alterations are part
the major causes of cancer mortality in women (1). Epide-
of the cervical cancer network and present an opportunity for
miological and molecular data indicate that persistent infec-
developing therapies against any of the participating ele-
tion with ‘‘high-risk’’ human papillomavirus (HPV) is a
ments. In this review, we highlight the role of these pathways
prerequisite for cervical cancer development and that it is
in cervical carcinogenesis, their alterations and the conse-
associated with other pathologies, such as head and neck
quences of these abnormalities. In addition, we review the
cancers and anal cancer (2). More than 100 types of HPV
current preclinical and clinical data for designed agents that
have been identified to date; 35 are classified as high-risk
target elements of these pathways.
and another 40 as low-risk based on oncogenic potential (3).
The conventional treatment of cervical cancer consists
HPV overexpression of E6 and E7 oncoproteins is the
of surgery, radiation and chemotherapy alone or in combi-
main oncogenic stimulus for cell transformation in cervical
nation (10). Such treatments remain suboptimal, with resid-
cancer. High-risk E6 proteins induce the degradation of
ual tumors observed in 40e50% of patients with advanced
cancer (11,12). Therefore, there is a need for implementing
Address reprint requests to: Marcela Lizano, Instituto Nacional de
Cancerologıa, Av. San Fernando 22, Col. Seccion XVI, Tlalpan, Mexico novel therapeutic approaches that, in addition to the stan-
City, D.F., 14080 Mexico; Tel: (þ52) (55) 5628-0400 ext.133; FAX: dard treatments, could improve the overall survival and
(þ52) (55) 5628-0426; E-mail: lizanosoberon@gmail.com quality of life of cervical cancer patients.

0188-4409/$ - see front matter. Copyright Ó 2014 IMSS. Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.arcmed.2014.10.008
526 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

Signal Transduction Pathways Implicated in Cervical the ATP-binding cleft of the enzyme and stabilizes the
Cancer protein in a conformation that prevents substrate binding
and phosphorylation (26). Sorafenib is being assessed in a
ERK/MAPK Signaling Pathway
phase I/II clinical trial in combination with radiotherapy
The ERK/MAPK pathway, also known as the RAF/MEK and cisplatin to determine its biological activity in cervical
pathway, is a ubiquitous signal transduction pathway that cancer (www.clinicaltrails.gov; protocol: NCT00510250)
regulates crucial cellular processes, including proliferation, (Table 1 and Figure 1).
differentiation, angiogenesis and survival (13). Impor- Small molecule inhibitors of MEK1/2 are highly specific
tantly, overexpression or activation of components of this protein kinase inhibitors. Since the development of the first
pathway is believed to contribute to tumorigenesis, tumor MEK inhibitors PD98059 and U0126 (27), a considerable
progression and metastatic disease in a variety of solid tu- number of preclinical studies have investigated various
mors (14). The ERK/MAPK pathway lies downstream of MEK1/2 inhibitors (28).
various growth factors such as EGF (epidermal growth fac- PD98059 is a flavonoid and a potent inhibitor of MEK
tor). Growth factor binding results in receptor phosphory- (29). The suppression of MEK by PD98059 results in resis-
lation, which activates an adapter molecule complex tance to cisplatin, but not to doxorubicin (another common
known as GRB2/SHC/SOS. This in turn activates the anti-cancer drug based on DNA intercalation). Both drugs
RAF/MEK/ERK pathway, which triggers a cascade of spe- induce NF-kB activation in SiHa (30). U0126 is a chemi-
cific phosphorylation events. Within the RAF/MEK/ERK cally synthesized organic compound that is a very selective
pathway, the small GTPase RAS and the serine/threonine and potent inhibitor of MEK1 and MEK2. In contrast to
kinase RAF are the key molecular signal regulators (15). PD098059, which only inhibits inactive MEK, U0126
Intermediate signaling is regulated by MEK1 and MEK2, inhibits both active and inactive MEK-1/2 (31).
which are responsible for phosphorylating and activating CI-1040 is another potent and selective MEK inhibitor
the final downstream signaling molecules known as that has displayed preclinical activity in a broad spectrum
ERK1 and ERK2 (16). ERK1/2 regulates cellular activity of tumor xenografts including those derived from colon
by acting on more than 100 substrates in the cytoplasm cancer and melanoma (32). CI-1040 treatment produces a
and nucleus, including indirect inducers of gene expres- reduction of the pMAPK levels in the cervical tumor cell
sion, transcription factors and cell cycle-related kinases line A431.
(17). RAS, another member of the RAF family, also has From the MAPK cascade, the MEK enzymes are the
a regulatory role in the Erk/MAPK pathway and other most attractive molecules for cancer therapy. Fourteen
signaling pathways, such as the PI3K/Akt/mTOR, PKC MEK inhibitors are currently undergoing testing (33), with
and the RALGDS pathways, which have been implicated two being tested for cervical malignancy, U0126 and trem-
in carcinogenesis (18,19). Ras is activated in |20% of hu- atinib (GSK1120212) or in the treatment of recurrent or
man cancers, including cervical cancer, where it has been persistent cervical cancer in combination with the Akt
linked to the metastatic conversion of tumor cells (20). inhibitor GSK214179 (www.clinicaltrials.gov, protocol:
Mutations of Ras and Myc amplification are frequently NCT01958112).
associated with the development of recurrent cervical can- The MAPK network has been the subject of intense
cer (21,22). research and pharmaceutical scrutiny to identify novel
target-based approaches for cancer treatment. Among these
Therapeutic targets for ERK/MAPK pathway in cervical kinases, RAF and MEK have received substantial attention,
cancer. Epidermal growth factor receptor (EGFR) overex- but many more preclinical studies in cervical cancer are
pression is frequently observed in cervical cancer and is an required.
independent predictor of poor prognosis in advanced-stage
tumors (23,24). EGFR is a potential target for the treatment
The PI3K/Akt Signaling Pathway
of cancer, and a number of agents targeting EGFR have
been designed (25). There is growing evidence that anti- The PI3K signaling pathway is often activated in various
EGFR strategies exert their anti-tumor activities in part human cancers (34,35). PI3K is a downregulator of the
by interfering with the MAPK and Akt pathways. Table 1 Ras signaling pathway, and it is a ubiquitous lipid kinase
summarizes the agents used to suppress elements of the that is involved in receptor signal transduction (36). This
signaling pathways implicated in cervical carcinogenesis protein is an intracellular transducer with a catalytic and
and apoptotic inducers, either in clinical trials and in vitro a regulatory subunit (37). Activated PI3K converts the
models of cervical cancer. membrane lipid phosphatidylinositol-4,5-bisphosphate
Once the MAPK pathway is activated, the RAF kinase (PtdIns[4,5]P2) to phosphatidylinositol-3,4,5-trisphosphate
takes action. Sorafenib is the first oral RAF kinase modu- (PtdIns[3,4,5]P3). The protein serine-threonine kinases
lator to receive marketing approval for the treatment of Akt and phosphoinositide-dependent kinase 1 (PDK1) are
advanced renal cell carcinoma. This inhibitor interacts with recruited at the PI3K activation sites by directly binding
Table 1. Targeted signal transduction elements in cervical cancer and their mechanism of action

Therapy Signaling pathway Target Mechanism of action Adjuvant treatment tested Clinical protocol References

Sorafenib MAPK Raf kinase Competitive inhibition of Radiotherapy/Cisplatin NCT00510250 Garcıa, 2009
the ATP site
PD98059 MAPK Inactive MEK ½ Protein kinase inhibitors Cisplatin and Doxorubicin - Yeh et al. 2002
U0126 MAPK Active and inactive Protein kinase inhibitors - - Favata et al. 1998
MEK ½
Trematinib MAPK MEK ½ Binds and inhibits MEK ½ GSK2141795 NCT01958112 Miller et al. 2014
action
CI-1040 MAPK MEK Induces decrease in pMAPK - - Montagut, 2009
levels
Wortmannin PI3K p110 Irreversible competitor. Radiotherapy/ - Zhang et al. 2009
Competes with the ATP site Chemotherapy/Roscovitine
LY294002 PI3K p110 Reversible competitor. Radiotherapy - Fuhrman et al. 2007
Competes with the ATP site
inducing apoptosis
Quercetin PI3K p110 Competes with ATP - - Huangh et al. 2009
binding De et al. 2000

Molecular Targets in Cervical Cancer


Indole-3-carbinol PI3K Akt Inactivates Akt by - NCT00910884 Bell et al. 2000
demethylation of PTEN. Jin et al. 1999
Induces apoptosis Yuan et al. 1999
MK-2206 PI3K Akt Allosteric inhibitor of Akt - - Cherrin et al. 2010
1/2 Tolcher et al. 2009
Genistein PI3K Akt Avoids Akt phosphorylation Radiotherapy NCT00001696 (Completed Yashar et al. 2005
at Ser473 for toxicity) Su-Hyeong et al.
2009
Oridonin PI3K Akt Reduction of Akt - - Hu et al. 2007
phosphorylation
Tripterygium PI3K Akt Reduction of Akt - - Yang et al. 2006
phosphorylation
Temsirolimus PI3K mTOR Forms a complex with the Topotecan NCT01217177 Temkin et al. 2010
FK506 binding protein-12 NCT01026792 (Completed)
Everolimus PI3K mTOR Forms a complex with the Paclitaxel Cisplatin/ NCT01217177 Campone et al. 2009
FK506 binding protein-12 Chemotherapy NCT01026792 (Completed)
Cetuximab EGF EGFR extracellular Avoids the ligand binding Cisplatin/Topotecan (Toxic) NCT00957411 Hertlein et al. 2010
fraction Radiation/Cisplatin NCT00997009 Kurtz et al.Moore
Chemotherapy et al. 2012
Santin et al. 2011
Nimotuzumab EGF EGFR extracellular Avoids the ligand binding Cisplatin/Gemcitabine, NCT02095119, NCT01938105,
fraction chemoradiotherapy, NCT02083211, NCT02039791,
Carboplatin/Paclitaxel, NCT01301612
Brachytherapy
Matuzumab EGF EGFR extracellular fraction Avoids the ligand binding Platinum - Blohmer et al. 2005
Lapatinib EGF EGFR tyrosine kinase Tyrosine kinase inhibitor - NCT00430781 Monk et al. 2010
fraction (Completed)
(continued on next page)

527
528
Table 1 (continued )
Therapy Signaling pathway Target Mechanism of action Adjuvant treatment tested Clinical protocol References

Bevacizumab VEGF VEGFR-A Avoids the ligand binding Fluorouracil/Capecitabine NCT00025233 (Completed) Wright et al. 2006
Topotecan/Cisplatin NCT00626561 (Terminated) Monk et al. 2009
Radiotherapy/Cisplatin NCT00803062 Zighelboim et al. 2013
Schefner et al. 2011
Pazopanib VEGF VEGF tyrosine kinase Tyrosine kinase inhibitor Lapatinib/Chemotherapy NCT00430781 Monk et al. 2010
fraction
Lapatinib EGF EGFR tyrosine kinase Tyrosine kinase inhibitor - - Monk et al. 2010
fraction
Imatinib PDGF PDGFR kinase fraction Tyrosine kinase inhibitor - - Taja et al. 2006
Gefitinib EGF EGFR tyrosine kinase Tyrosine kinase inhibitor - NCT00049556 Goncalves et al. 2008
fraction (Completed)

Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539


Erlotinib EGF EGFR tyrosine kinase Tyrosine kinase inhibitor Cisplatin/Radiotherapy NCT00031993 Nogueira et al. 2008
fraction (Completed) Schilder et al. 2009
3,3-diindolylmethane MAKP Several Downregulates elements of - - Zhu et al. 2012
PI3K the PI3K and MAPK
pathways
Retinoids Apoptosis Fas/FasL EGF, Induces FasL. Deregulates - - Sun et al. 2000
EGF pathway Hembree et al. 1996
N101-2 PI3K/Akt Fas/FasL Arrests cell cycle at sub-G1 - - Kim et al. 2012
Extrinsic apoptosis PI3K/Akt phase. Induces apoptotic
cell death
Silibilin Not yet elucidated Cell cycle-dependent Induces G arrest and - - Zhang et al. 2012
kinases apoptotic cell death
Ganoderic acid derivates Intrinsic apoptosis Not known yet Decrease mitochondrial - - Liu et al. 2012
membrane potential
Recombinant human Apoptosis Death receptor 4/5 Binds the DR directly and Radiation - Maduro et al. 2008
TRAIL acts as an agonist
Luteolin Apotptosis DR5 Induces Bid cleavage and rhTRAIL - Horinaka et al. 2005
caspase-8 activation
Aspirine Intrinsic apoptosis Not specified Suppresses the activation of TRAIL - Im and Jang, 2012
Erk 1/2
Arsenic trioxide Multiple signaling E6 Induces a reduction in E6 NCT00005999 Wen et al. 2012
pathways protein levels with cell (Completed)
cycle arrest in G2-M in
HPV expressing cells
Avenanthramide 2p Wnt/b-catenin b-catenin Induces b-catenin - - Wang et al. 2012
degradation and avoids its
nuclear accumulation
Triptolide Wnt/b-catenin b-catenin Induces b-catenin - - Wang et al. 2012
degradation and avoids its
nuclear accumulation

Different elements designed to block signal transduction through inhibiting certain elements are presented. Current preclinical and clinical evaluations of such compounds are cited.
Molecular Targets in Cervical Cancer 529

Figure 1. MAPK, PI3K/Akt and Wnt pathways and therapeutic interventions. After ligand binding, the receptors start the signaling cascade downstream the
molecular effectors. In cancer cells signaling pathways are overfunctional because several elements are either overexpressed or overactivated. The figure
shows the main elements in those pathways and the directed therapeutic agents. Most of those agents block tyrosine kinase activity or downstream kinases.
MAbs are intended to block the ligand binding to its receptor and dimerization.

to PtdIns(3,4,5)P3 (38). The proteins recruited to profiles in cervical cancer specimens, supported by the
PtdIns(3,4,5)P3 facilitate the phosphorylation of Akt by observation that the PI3K catalytic subunit is amplified in
PDK1 (39). This phosphorylation stimulates the catalytic cervical cancers (46).
activity of Akt, resulting in the phosphorylation of other HCCR oncogenes, which are related to the tumorigen-
proteins that affect cell cycle entry, cell proliferation, and esis process through their interaction with p53, regulate
anti-apoptosis. PI3K/Akt signaling in cervical cancer (47).
The termination of PI3K signaling by the degradation of
PtdIns(3,4,5)P3 can be mediated by at least two different Therapeutic targets for PI3K/Akt pathway in cervical
types of phosphatases. One is Src-homology 2-containing cancer. The first relatively specific PI3K inhibitors
phosphatase (SHIP), which dephosphorylates the 5 position described were Wortmannin (Wm), a naturally occurring
of the inositol ring to produce PtdIns(3,4)P2. The other is metabolite of Penicillium funiculosum, and LY294002
PTEN (phosphatase and tensin homologue deleted on chro- (2-(4-morpholinyl)-8-phenyl-chromone), derived from the
mosome (10), which dephosphorylates the 3 position of flavonoid quercetin (48). Both pharmacologic agents target
PtdIns(3,4,5)P3 to produce PtdIns(4,5)P2. In many human the p110 catalytic subunit of PI3K (49) by competing with
cancer types, deleted PTEN and upregulated PI3K lead to ATP binding (50).
the enhanced cellular synthesis of PtdIns(3,4,5)P3. Thus, LY294002 is a reversible PI3K inhibitor that has a very
PI3K and PTEN are deeply involved in cell survival short half-life, is insoluble in aqueous solutions, and has
pathways, which include the regulation of gene expression, off-target activity and high toxicity. These characteristics
vesicular trafficking, cellular metabolism and cytoskeletal have limited its clinical application (49,51). However, this
rearrangements (40e42). In fact, growing evidence sug- metabolite has been shown to cause apoptosis in various hu-
gests PTEN as a promising biological marker because man cervical cancer derived cells in vitro, in addition to cell
PTEN expression correlates with tumor incidence, size growth inhibition (52), through the up-regulation of the
and progression, making this protein an excellent candidate FOXO1 nuclear activity by reducing PI3K mediated phos-
for early diagnosis and an opportunity for therapeutic inter- phorylation (53).
vention (43). LY294002 has also been reported to cause a potent
The constitutive activation of the PI3K/Akt signaling radiosensitization via double-strand DNA break repair
pathway has been firmly established as a major determinant inhibition in HeLa cells (54). Conversely, Wm is a potent
of tumor cell growth and survival in a number of solid tu- irreversible inhibitor of PI3K (55). It enhances apoptosis
mors (44). AKT1 gene alterations have been reported in a in several cell lines associated with the inhibition of
genomic analysis identifying an amplification of the region Akt activation. Wm radiosensitizes cancer cells (49) by
14q32.33 (45). These results were validated by expression decreasing the activities of both the ATM protein and
530 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

DNA-PK (DNA-dependent protein kinase catalytic subunit) phosphorylated Akt protein at Ser473 compared to control
(56). In addition, Wm promotes a chemosensitizer effect in cells, resulting in a dose-dependent induction of apoptosis in
HeLa cells when challenged with the experimental drug cells that display constitutively active Akt (70). Genistein is
roscovitine (57). a small molecule found in high abundance in natural soy prod-
Quercetin is a naturally occurring flavonoid that can ucts (71). Genistein significantly suppressed the cell growth of
inhibit a broad range of protein kinases. In fact, it is the pro- HeLa and CaSki cells, perhaps due to Akt inhibition (72). It
totypal compound from which a variety of derivatives, also functions as a radiosensitizer for cervical cancer cells
including LY294002, were designed (50). Huangh et al. such as CaSki and ME180 (73). This drug has been tested
showed that quercetin inhibits HeLa cell proliferation and in numerous clinical trials of prostate, breast and pancreatic
induces cell death via the Ca2þ-dependent mitochondrial cancer. However, genistein has not been tested in patients with
apoptosis pathway (58). In addition, quercetin inhibits the cervical neoplasia until recently (clinicaltrials.gov).
adhesion, migration and invasion of HeLa cells (52). Two more natural products have shown Akt inhibitory
Finally, testing of quercetin in mouse models showed potential: oridonin, which is isolated from Rabdosia rubes-
that this agent is able to arrest or reverse the progression cens (74), and triptolide, the main active component of the
of cervical neoplasia (59). However, quercetin has not been traditional Chinese herbal medicine Tripterygium wilfordii
investigated in clinical trials to date. Hook (75). Both products were tested in HeLa cells where
The phytochemical indole-3-carbinol (I3C) is an they were effective apoptotic agents with concomitant
anti-carcinogenic and anti-estrogenic compound found reduction of Akt phosphorylation.
abundantly in cruciferous vegetables such as broccoli and Downstream in the PI3K/Akt pathway there is a distal
brussels sprouts. It inhibits the growth of benign tumors component known as mTOR. Rapamycin, the prototypic
of laryngeal tissue caused by HPV-16 in a mouse model mTOR inhibitor, was discovered in 1975 on the island of
and was shown to be effective in the treatment and preven- Rapa Nui as a potent anti-fungicide that is produced natu-
tion of laryngeal papillomas caused by HPVs (60). It is rally by Streptomyces hygroscopicus (76). More recently,
clear that I3C inactivates Akt in tumor cells by upregulating Rapamycin analogues such as CCI-779 and RAD-001 have
the tumor suppressor protein PTEN. The mechanism by been explicitly designed for development as anti-cancer
which I3C upregulates PTEN is unknown, but the demethy- drugs (50). These drugs are small molecule inhibitors that
lation of PTEN by I3C (61) and the upregulation of the function intracellularly, forming a complex with the
transcription factor Egr-1, a regulator of PTEN expression, FK506 binding protein-12 (FKBP-12), which is then recog-
are potential mechanisms (62). nized by mTOR (77).
Bell and co-workers carried out a clinical trial using oral CCI-779, known as temsirolimus, is an intravenously
I3C to treat women with cervical intraepithelial neoplasia administered agent approved by the U.S. Food and Drug
(CIN) (63). The authors reported a statistically significant Administration (FDA) and the European Medicines Agency
regression of CIN in patients treated with I3C, compared (EMEA) for the treatment of advanced renal cell carcinoma
with placebo. Moreover, indole-3-carbinol has been used (77,78). CCI-779 has anti-tumor activity in a variety of other
as adjuvant treatment for recurrent respiratory papillomato- human cancer models (79). In fact, O100 clinical trials for
sis (RRP) with promising outcomes (64). this drug are registered at the clinical trial website. A phase
Once the PI3K/Akt pathway is activated the number of I trial undertaken by Temkin et al. tested this drug in combi-
outputs increases as the signal moves down the pathway. In- nation with topotecan in a variety of gynecological malig-
hibiting PI3K should provide the maximum inhibition of all nancies and showed that the regimen may be safe for
outputs, whereas inhibiting downstream targets, such as Akt, women who have not previously received radiation (80).
and even further downstream targets, such as mammalian Another rapamycin analog, RAD-001, known as
target of rapamycin (mTOR), will give progressively more everolimus, is an oral agent that has recently obtained
selective output inhibition (65). Although Akt is the central U.S. FDA and EMEA approval for the treatment of
player in this important pro-survival pathway, few direct advanced renal carcinoma (77). Everolimus displays
Akt inhibitors have been developed that may be directly antiproliferative effects on cancer cells, yields anti-
translated to the treatment of humans (66). A series of naph- angiogenic activity in established tumors, and has
thyridine and naphthyridinone allosteric dual inhibitors of synergistic activity with paclitaxel in preclinical models
Akt1 and 2 have been developed (67). MK-2206 is a com- (81). Some clinical trials are now recruiting patients
pound from this class of Akt inhibitors. Treatment of the cer- with cervical tumors (www.clinical trails.gov; protocol:
vical cell line C33A with MK-2206 resulted in the NCT01217177) (Table 1 and Figure 1).
redistribution of Akt from the membrane to the cytoplasm
(68). Phase I studies in solid tumors suggested that MK-
The EGFR/VEGFR Signaling Pathway
2206 is an effective therapeutic drug (69).
Banerjee et al. found that genistein (40 ,5,7- Epidermal grow factor receptor (EGFR) and its family
trihydroxyisoflavone) treatment reduced the level of the members Her-2/Erb-2, Her-3/Erb-3 and Her4/Erb-4
Molecular Targets in Cervical Cancer 531

belong to a tyrosine kinase receptor family. Together with receptor dimerization, internalization and downregulation,
their ligands, these receptors are involved in O70% of all which in turn inhibits receptor activation and signaling
cancers (82). EGFR is associated not only with the prolif- (98). Cetuximab has been approved for the treatment of
eration of tumor cells but also with enhanced tumor cell squamous cell carcinoma of the head and neck in combina-
survival, angiogenesis and metastatic spread (83). At least tion with radiotherapy (99). In addition, cetuximab
six ligands are known to directly activate EGFR: combined with irinotecan increased response rates and
epidermal growth factor (EGF), transforming growth progression-free survival in patients with colorectal cancer
factor alpha (TGF-a), heparin binding EGF-like growth (100). However, a recent report by Hertlein et al. found no
factor (HB-EGF), amphiregulin, betacellulin, and epiregu- advantage for cetuximab monotherapy in patients with
lin. These proteins are synthesized as transmembrane advanced cervical cancer (101), whereas Kurtz et al.
precursors and are proteolytically cleaved by metallopro- demonstrated that the combination of cisplatin-topotecan-
teases to release the mature growth factor in an autocrine cetuximab induces a high rate of serious adverse and/or
or paracrine manner (84). Extracellular ligand binding fatal events at the standard dose and schedule (102). In
causes dimerization of the EGFR, which becomes auto- another study, the feasibility of cetuximab combined with
phosphorylated at distinct tyrosine residues by the recep- whole pelvic radiation and cisplatin compared with cetuxi-
tor’s intrinsic kinase activity and, depending on the mab combined with extensive field radiation and cisplatin
ligand, intracellular pathways can be activated (85). The was investigated. The latter combination was highly toxic
best-known pathways are the ERK1/2 and PI3 kinases as and not feasible for the treatment of patients with cervical
previously described. cancer (103). Santin et al. demonstrated that cetuximab is
Many studies have shown that the overexpression of well tolerated as a mono-drug with limited activity in the
EGFR is linked to the development of cervical cancer study population, with specific activity and limited effect
(8,24,86,87) and that its expression is correlated with poor on patients with squamous cell histology (104).
prognosis and metastasis (23,88e90). Because EGFR pro- Nimotuzumab, another EGFR inhibitor, inhibits the
teins are cell surface receptors, they can be directly targeted. EGF-related signaling and ligand-independent signaling,
The relationship between EGFR expression and cervical can- but at higher concentrations than cetuximab (105,106),
cer has led to the development of many therapeutic agents suggesting its potential use as a therapy in tumors of epithe-
targeting this receptor (91), with the monoclonal antibodies lial origin (107). Because EGFR is overexpressed in
(mAb) the most promising in a series of clinical trials. a considerable number of tumors of the cervix, it is
Another method of targeting EGF is the use of small- currently undergoing testing in a variety of clinical
molecule, adenosine triphosphate-competitive inhibitors of trials alone or in combination (www.clinicaltrails.gov;
the tyrosine kinase receptors (92), as reviewed here. protocols: NCT02095119, NCT01938105, NCT02083211,
Angiogenesis is a fundamental step in the formation of NCT02039791, NCT01301612). Two studies are testing
metastasis. EGF-activated signals may induce the synthesis the efficacy and tolerability of cetuximab in cervical cancer
of vascular endothelial growth factor (VEGF), promoting patients (www.clinicaltrials.gov; protocols: NCT00957411
angiogenesis, tumor growth cell survival, and metastasis and NCT00997009).
(93). In cervical cancer, the angiogenic process is related Matuzumab, a humanized immunoglobulin G1 mono-
to VEGF expression (94), which correlates with severity clonal anti-EGFR antibody, was effective in patients with
in precursor lesions and invasive disease (95). The VEGF cervical cancer that progressed after treatment with
family includes VEGF-A to F, with VEGF-A as the most platinum-based chemotherapy. In the 38 patients enrolled
important member. Cooper et al. evaluated intra-tumoral in the study, the antibody was well tolerated, with two cases
microvessel density (IMD) in patients treated with pelvic of partial response and nine cases with stable disease (108).
irradiation and reported that a high vascularity was associ- Bevacizumab is a mAb directed against VEGF-A. It was
ated with poor survival rates in which IMD acted as a prog- the first anti-angiogenic agent that was successfully used in
nostic marker (96). colorectal cancer (109). Bevacizumab delayed the time to
progression by 4 months and increased the median survival
Platelet-derived growth factor. Platelet-derived growth fac- in patients. Bevacizumab has also been tested in cervical
tor (PDGF) is a very strong stimulator of blood vessel forma- cancer patients in whom prior therapy failed. Studies of
tion and has also been associated with the pathogenesis of bevacizumab plus fluorouracil or capecitabine found clin-
cervical cancer. This ligand and its receptors are frequently ical benefit in 67% of patients. Therapy was well tolerated;
expressed in cervical cancer-derived cell lines (97). thus, bevacizumab showed anti-tumor activity in pretreated
cervical cancer (110). Subsequent studies demonstrated a
Therapeutic targets for EGFR and VEGFR in cervical can- favorable toxicity profile and similar results for bevacizu-
cer. Cetuximab is a human/murine chimeric immunoglob- mab (111). Another study showed that the combination of
ulin G2 mAb that targets EGFR. Preclinical studies showed bevacizumab with topotecan and cisplatin induces an
that cetuximab binds to EGFR with high affinity, inducing improvement in both the overall survival and progression-
532 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

free survival of patients with recurrent or persistent cervical The intrinsic apoptotic pathway involves procaspase-9,
cancer (112). Another study of bevacizumab in combina- which is activated downstream of mitochondrial pro-
tion with radiotherapy and cisplatin showed a very safe pro- apoptotic events at the so-called apoptosome, a cytosolic
file with an improvement of overall survival and without death signaling protein complex formed upon release of
adverse events (113). An independent study is currently cytochrome c from the mitochondria. In this case, it is
testing the efficacy of paclitaxel and cisplatin or topotecan the dimerization of procaspase-9 molecules at the Apaf-1
in combination with or without bevacizumab (www. scaffold that is responsible for caspase-9 activation (120).
clinicaltrails.gov; protocol: NCT00803062). Once the initiator caspases have been activated, they can
The tyrosine kinase inhibitors (TKIs) pazopanib and la- proteolytically activate the effector procaspases 3, 6, and
patinib, which target VEGF and EGF, respectively, have 7, which subsequently cleave a specific set of protein sub-
demonstrated antiangiogenic activity in a phase II clinical strates, including the procaspases themselves (120).
trial performed by Monk and co-workers (2010) in women Various proteins regulate the apoptotic process at
with recurrent or advanced cervical cancer (114). This trial different levels. FLIPs (FADD-like interleukin-1b
compared antiangiogenesis therapy (pazopanib) with converting enzyme-like protease) and FLICE/caspase 8
EGF-based therapy (lapatinib). The authors suggest the su- (FADD-like ICE) inhibitory proteins interfere with the initi-
periority of pazopanib due to its favorable toxicity profile, ation of apoptosis directly at the level of death receptors.
easier administration (oral), high antiangiogenic activity The IAPs (inhibitor of apoptosis proteins) constitute
and higher overall survival (11.6 weeks longer than the another class of regulatory proteins. IAPs bind to and
lapatinib arm). Erlotinib, another TKI, has been evaluated inhibit caspases. They might also function as ubiquitin li-
as monotherapy (115), as has gefitinib (116). These were gases, promoting the degradation of the caspases that they
well tolerated but without an objective response. The bind (121).
authors declared that monotherapy with those agents had A very important feature in cancer establishment is the
no effect in patients with advanced or recurrent cervical modulation of apoptosis mediated by either viruses or onco-
cancer. However, another phase I trial showed that the genes supporting proliferation, immortalization, transfor-
use of erlotinib is feasible, presenting an acceptable toxicity mation and finally cancer (122). Cervical cancer is not an
profile in patients undergoing erlotinib in combination with exception, with a disruption in the apoptotic pathway pri-
cisplatin and radiotherapy (117). Finally, the potential use marily due to the continued expression of E6 protein. The
of another TKI, imatinib, has been tested in an in vitro pilot E6 protein binds to p53 with the aid of E6AP and prevents
study using cervical cancer-derived cells. The authors p53 from inducing apoptosis by targeting it for degradation
showed that imatinib inhibited cell growth by hampering via the ubiquitin-proteasome pathway (123). In malignant
the phosphorylation of PDGFR, suggesting that it may be cells, these physiological apoptotic pathways are often
a good target for the development of a therapy for cervical disturbed, which allows for uncontrolled cell proliferation
cancer (97) (Table 1 and Figure 1). (124).
Downregulation of Fas expression is a common abnor-
mality in gynecological cancers (125). One study focused
The Apoptotic Signaling Pathways
on the pattern of expression of apoptotic-related proteins
The life and death of cells must be balanced to maintain showed that Fas is lost as long as the lesion progresses.
tissue homeostasis. Disruption of this balance can lead to The FasL, TRAIL and death receptors (DR) 4 and 5 are
a severe disturbance that may result in cancer (118). Cells frequently expressed in the cytoplasm of tumor cells in cer-
have an intrinsic mechanism to control tissue homeostasis vical cancer patients. Because DR4, DR5, and TRAIL are
linked to apoptosis. Defects in the apoptosis-inducing path- frequently observed, the absence of TRAIL expression
ways can eventually lead to the expansion of a population was associated with a higher pathological complete
of neoplastic cells. response rate to radiotherapy (126,127). Conversely, HPV
In principle, there are two alternative pathways that E5 protein impairs the TRAIL- and FasL-mediated
initiate apoptosis. One pathway, occasionally referred to apoptosis by downregulating the Fas receptor and inter-
as the extrinsic pathway, is mediated by death receptors fering with the DISC formation upon the TRAIL signal
on the cell surface, which are activated when the death- in raft cultures of E5 expressing keratinocytes, which were
inducing ligand binds to the death receptors (DR). Upon completely protected from FasL- and TRAIL-induced cell
binding, caspases (cysteine aspartyl-specific proteases) death (128). Similarly, when UV radiation is used to induce
8 and 10 become activated which, in turn, activates cas- stress, E5 expressing human keratinocytes are protected
pases 3, 6 and 7. The other apoptotic pathway, the intrinsic from apoptosis (129).
pathway, is mediated by the mitochondria. Fas and TRAIL
receptors are the main regulatory elements in the extrinsic Therapeutic targets for apoptotic signaling pathways in
pathway and are expressed by a broad panel of normal cervical cancer. The induction of apoptosis is most likely
epithelial cells (119). the underlying mechanism of therapeutic strategies that
Molecular Targets in Cervical Cancer 533

have been or are being tested. Several approaches have suc- and IGF signaling pathways (137), induction of Fas and in-
cessfully induced apoptosis in cervical cancer-derived cell crease in Fas/Fas ligand-mediated apoptosis (138). I3C has
lines by using chemical compounds such as luteolin (130) exhibited chemopreventive effects in preclinical studies.
and aspirin (131), either alone or in combination with radio- This agent has anti-estrogenic activities and induces
therapy or by using soluble apoptotic inducers such as apoptosis (139). Transgenic mice harboring tumors were
TRAIL (132). fed a diet supplemented with I3C, and the incidence of can-
Recently, a number of pro-apoptotic agents have been cer in these animals was dramatically reduced (60).
tested in cervical cancer cells. The N101-2 compound, a Conversely, 3,3-diindolylmethane (DIM), an important
flavonoid derivate, showed clear apoptotic effects in CaSki polymer derived from I3C, exerts antiproliferative and
and SiHa cells by arresting the cell cycle at the G1 phase, pro-apoptotic effects in a cellular model by affecting the
activating intrinsic pathways, and inhibiting the PI3K/Akt MAPK and PI3K pathways when used as a mono-drug
signaling pathway (133). Similarly, silibilin induced a (140).
dose-dependent apoptotic death in HeLa cells by activating A promising preclinical report from Wen et al. found
both the mitochondrial and death receptor-mediated that arsenic trioxide induces apoptosis by specifically
pathway (134). Meanwhile, ganoderic acid derivatives targeting HPV-expressing cells, decreasing cell viability
demonstrated therapeutic potential in HeLa cells, inducing 48e60% compared with control cells, which decreased
apoptosis by decreasing mitochondrial membrane potential 16% (141). This effect is due to downregulation of E6
in a very specific manner (135). expression and the subsequent rescue of p53 and cas-
Retinoids, a class of compounds related to vitamin pase 3 levels that are conducive to cell death. Certainly,
A, may play a role as chemopreventive agents in the end point for cervical cancer therapy is to induce cell
HPV-associated CIN and cervical cancer. In vitro treatment death within the tumor, and most of the registered
of HPV-immortalized keratinocytes and cervical carcinoma protocols at www.clinicaltrails.gov aim to improve
cell lines with retinoids downregulated E6/E7 transcription overall survival and progression-free survival with a
and upregulated wild type p53 expression (136), with high apoptotic rate and low toxic profile (Table 1 and
several consequences including the deregulation of EGF Figure 2).

Figure 2. Apoptotic signaling pathway and current agents inducing apoptosis. The apoptotic pathway starts when the death signal is received by death re-
ceptors (DR) allowing the intracellular activation of caspase 8. The signal proceeds through the effector caspase cleavage with cell death as the final end.
Caspase 8 induces Bid activation which, in turn, allows cytochrome C release from mitochondria, with subsequent caspase 9 activation enhancing the
apoptotic signal. The yellow arrows show the targeted elements that have been tested either in clinical and preclinical cervical cancer models studies by
different agents (bold) with the final aim of activating the apoptotic pathway via extrinsic or intrinsic mechanisms.
534 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

Wnt Signaling Pathway activators such as Wnt ligands, frizzled receptors and
disheveled (Dvl). In cervical cancer cell lines, the overex-
The Wnt signaling pathway is critical in embryonic devel-
pression of Wnt10B-14 (158,159), Fzd-10 (160), and Dvl-
opment and is a large family of secreted growth factors that
1 (161) has been reported.
control cell fate, proliferation, migration, tissue architec-
In a genome expression analysis conducted in HPV-16-
ture, and organogenesis (142,143). Wnt also plays an
associated cervical cancer cases, a significant increase in
important role in adult organisms because it regulates the
Wnt-4, -8a, Fzd2, GDK3b and b-catenin compared with
homeostasis of hematopoiesis (144), angiogenesis (145)
normal cervical epithelia was observed (162).
and adipogenesis (146). Deregulation of the Wnt pathway
Continuous turnover of epithelia is ensured by the self-
has been implicated in cancer (147).
renewal capacity of tissue-specific stem cells (163). In the
It is believed that three different pathways can be
same way, cancer stem cells maintain epithelial tumors
activated upon Wnt receptor activation: the canonical
and b-catenin signaling is essential in sustaining the pheno-
Wnt/b-catenin cascade, the noncanonical planar cell polar-
type of cancer stem cells. In chemically derived mouse skin
ity (PCP) pathway, and the Wnt/Ca2þ pathway. The
tumors, ablation of the b-catenin gene results in the loss of
canonical pathway is the best understood.
cancer stem cells and complete tumor regression (164).
Wnt receptors include the Frizzled family (Fzd) and
Therefore, the b-catenin gene or other elements involved
co-receptors such as the low density lipoprotein
in the Wnt/b-catenin pathway may be targeted to eliminate
receptor-related proteins-5/6 (LRP). Once bound by their
cancer stem cells with the goal of eradicating squamous cell
cognate ligands, the Fzd/LRP co-receptor complex acti-
carcinoma.
vates the canonical pathway, which is characterized by
Multiple therapeutic interventions have been proposed
the accumulation of b-catenin in the cytoplasm and nu-
for the Wnt pathway. Related to cervical cancer, phyto-
cleus. Cytoplasmic b-catenin protein levels are controlled
chemicals with known antioxidant, anti-inflammatory and
by ubiquitination and degradation, which are induced by
chemopreventive properties have been tested in the cervical
phosphorylation by active GSK3b (148). Activation of
cancer-derived cell line, HeLa (165). Naturally occurring
the Wnt signaling pathway inhibits GSK3b and causes
polyphenols have antiproliferative effects through the atten-
the accumulation of b-catenin in the cytoplasm. The
uation of the Wnt pathway by inducing cellular b-catenin
excess of b-catenin promotes its entrance to the nucleus
degradation and a reduction in the nuclear abundance of
where it forms an active transcriptional complex with T-
b-catenin.
cell factor (TCF), which then activates the transcription
Some compounds have been tested in clinical assays for
of target genes including c-myc (149) and cyclin D
solid tumors, trying to reach distinct Wnt regulatory levels
(150,151).
with the use of small molecules, antibodies or peptides
In order for GSK3b to phosphorylate b-catenin, the pres-
(166) directed to different targets such as Wnt ligands, Friz-
ence of adenomatous polyposis coli (APC gene product)
zled receptors or b-catenin. It is clear that due to the
and axin are required (152). PP2A is also necessary because
complexity of Wnt signaling, this pathway cannot be tar-
it inhibits Wnt signaling through its direct interactions with
geted using a single strategy. Additional studies are
APC and axin (153,154). Several mutations in several com-
required to identify possible signaling nodes as targets for
ponents of the Wnt/b-catenin pathway have been studied
Wnt for successful therapeutic interventions in cervical
and identified in a variety of cancers. Wnt signaling may
cancer cells without affecting normal cells (Table 1 and
contribute to the development of cervical carcinoma
Figure 1).
because the activation of the canonical Wnt pathway is
necessary to induce the transformation of HPV immortal-
ized cells (155). Some authors have found that b-catenin
Conclusions and Future Directions
expression is increased in a high proportion of cervical
cancer specimens as assessed by cytoplasmic and nuclear Cervical cancer remains a public health issue among
positive staining, whereas mutations were found in no more women, primarily in developing countries, despite the use
than 20% of the samples (7,156). of the Papanicolaou smear and colposcopy programs.
It has been proposed that b-catenin may be activated by Although the use of screening tests could prevent cervical
the inactivation of negative regulators such as APC and ax- cancer development, 80% of cases are diagnosed in the ter-
in. It is possible that negative regulators of the Wnt/b-cat- minal stages. Surgery, radiation and chemotherapy alone or
enin pathway could be inactivated by methylation in combination are the conventional treatments for such
because the axin and APC genes have enriched CpG islands neoplasms, but some of the cases will persist or recur.
in their promoters, which are hypermethylated in cervical Effective treatment for advanced and/or recurrent cervical
cancer (157). cancer is needed as well as reliable markers for prognosis
Another mechanism involved in the upstream activation related to treatment. Thus, signal transduction elements
of the Wnt/b-catenin pathway is the over-expression of are promising targets for new drugs. Taken together,
Molecular Targets in Cervical Cancer 535

preclinical and clinical data suggest that combinatorial ther- 15. Kolch W. Meaningful relationships: the regulation of the Ras/Raf/
apies will improve outcomes in those patients, and that MEK/ERK pathway by protein interactions. Biochem J 2000;351:
289e305.
progress on this issue will help in managing those patients 16. Roberts LR, Gores GJ. Hepatocellular carcinoma: molecular pathways
in the future. Although some new molecular targeting drugs and new therapeutic targets. Semin Liver Dis 2005;25:212e225.
have shown to be successful, prevention will always be the 17. Halaschek-Wiener J, Wacheck V, Kloog Y, et al. Ras inhibition leads
best therapy. to transcriptional activation of p53 and downregulation of Mdm2:
two mechanisms that cooperatively increase p53 function in colon
cancer cells. Cell Signal 2004;16:1319e1327.
Acknowledgments 18. To MD, Perez-Losada J, Mao JH, et al. Crosstalk between PTEN and
Ras signaling pathways in tumor development. Cell Cycle 2005;4:
This work was partially supported by Consejo Nacional de Cien-
1185e1188.
cia y Tecnologıa CB-166808 and CB-151493.
19. Harden TK, Sondek J. Regulation of phospholipase C isozymes by
ras superfamily GTPases. Annu Rev Pharmacol Toxicol 2006;46:
355e379.
20. Yoshida S, Kajitani N, Satsuka A, et al. Ras modifies proliferation
References and invasiveness of cells expressing human papillomavirus oncopro-
1. Ferlay J, Soerjomataram I, Ervik M, et al. Cancer Incidence and Mor- teins. J Virol 2008;82:8820e8827.
tality Worldwide: IARC Cancer Base. No. 11 [Internet]. Lyon, 21. Alonio LV, Picconi MA, Dalbert D, et al. Ha-ras oncogene mutation
France: International Agency for Research on Cancer; 2013. Avail- associated to progression of papillomavirus induced lesions of uter-
able from: http://globocan.iarc.fr. Accessed October 7, 2014. ine cervix. J Clin Virol 2003;27:263e269.
2. Bouvard V, Baan R, Straif K, et al. WHO International Agency for 22. Narisawa-Saito M, Yoshimatsu Y, Ohno S, et al. An in vitro multistep
Research on Cancer Monograph Working Group. Lancet Oncol carcinogenesis model for human cervical cancer. Cancer Res 2008;
2009;10:321e322. 68:5699e5705.
3. Mu~ noz N, Bosch FX, de Sanjose S, et al. Epidemiologic classifica- 23. Kersemeakers AM, Fleuren GJ, Kenter GG, et al. Oncogene alter-
tion of human papillomavirus types associated with cervical cancer. ations in carcinomas of the uterine cervix: overexpression of the
International Agency for Research on Cancer Multicenter Cervical epidermal growth factor receptor is associated with poor prognosis.
Cancer Study Group. N Engl J Med 2003;348:518e527. Clin Cancer Res 1999;5:577e586.
4. Vande Pol SB, Klingelhutz AJ. Papillomavirus E6 oncoproteins. Vi- 24. Li Q, Tang Y, Cheng X, et al. EGFR protein expression and gene
rol 2013;445:115e137. amplification in squamous intraepithelial lesions and squamous
5. Roman A, Munger K. The papillomavirus E7 proteins. Virology cell carcinomas of the cervix. Int J Clin Exp Pathol 2014;7:
2013;445:138e168. 733e741.
6. Schwarz JK, Payton JE, Rashmi R, et al. Pathway-specific analysis of 25. Davies SP, Reddy H, Caivano M, et al. Specificity and mechanism of
gene expression data identifies the PI3K/Akt pathway as a novel ther- action of some commonly used protein kinase inhibitors. Biochem J
apeutic target in cervical cancer. Clin Cancer Res 2012;18: 2000;351:95e105.
1464e1471. 26. Garcia-Echeverria C. Protein and lipid kinase inhibitors as targeted
7. Shimohara A, Yokoyama Y, Wan X, et al. Cytoplasmic/nuclear anticancer agents of the Ras/Raf/MEK and PI3K/PKB pathways. Pu-
expression without mutation of exon 3 of the beta-catenin gene is rinergic Signal 2009;5:117e125.
frequent in the development of the neoplasm of the uterine cervix. 27. Alessi D, Cuenda A, Cohen P, et al. PD 098059 is a specific inhibitor
Gynecol Oncol 2001;82:450e455. of the activation of mitogen-activated protein kinase kinase in vitro
8. Fukazawa EM, Baiocchi G, Soares FA, et al. Cox-2, EGFR, and and in vivo. J Biol Chem 1995;270:27489e27494.
ERBB-2 expression in cervical intraepithelial neoplasia and cervical 28. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated
cancer using an automated imaging system. Int J Gynecol Pathol protein kinase cascade for the treatment of cancer. Oncogene 2007;
2014;33:225e234. 26:3291e3310.
9. Moody CA, Laimins LA. Human papillomavirus oncoproteins: path- 29. Reiners JJ, Lee J, Clift RE, et al. PD98059 is an equipotent antago-
ways to transformation. Nat Rev Cancer 2010;10:550e560. nist of the aryl hydrocarbon receptor and inhibitor of mitogen-
10. Duenas-Gonzalez A, Cetina L, Mariscal I, et al. Modern management activated protein kinase kinase. Mol Pharmacol 1998;53:438e445.
of locally advanced cervical carcinoma. Cancer Treat Rev 2003;29: 30. Yeh PY, Chuang SE, Yeh KH, et al. Increase of the resistance of hu-
389e399. man cervical carcinoma cells to cisplatin by inhibition of the MEK to
11. Classe JM, Rauch P, Rodier JM, et al, Groupe des Chirurgiens de ERK signaling pathway partly via enhancement of anticancer drug-
Centre de Lutte Contre le Cancer. Surgery after concurrent chemo- induced NF kappa B activation. Biochem Pharmacol 2002;63:
therapy and brachytherapy for the treatment of advanced cervical 1423e1430.
cancer: morbidity and outcome: results of a multicenter study of 31. Favata MF, Horiuchi KY, Manos EJ, et al. Identification of a novel
the GCCLCC (Groupe des Chirurgiens de Centre de Lutte Contre inhibitor of mitogen-activated protein kinase kinase. J Biol Chem
le Cancer). Gynecol Oncol 2006;102:523e529. 1998;273:18623e18632.
12. Keys HM, Bundy BN, Stehman FB, et al. Cisplatin, radiation, and 32. Montagut C, Settleman J. Targeting the RAFeMEKeERK pathway
adjuvant hysterectomy compared with radiation and adjuvant hyster- in cancer therapy. Cancer Lett 2009;283:125e134.
ectomy for bulky stage IB cervical carcinoma. N Engl J Med 1999; 33. Miller CR, Oliver KE, Farley JH. MEK1/2 inhibitors in the treatment
340:1154e1161. of gynecologic malignancies. Gynecol Oncol 2014;133:128e137.
13. Gollob JA, Wilhelm S, Carter C, et al. Role of Raf kinase in cancer: 34. Cantley LC. The phosphoinositide 3-kinase pathway. Science 2002;
therapeutic potential of targeting the Raf/MEK/ERK signal transduc- 296:1655e1657.
tion pathway. Semin Oncol 2006;33:392e406. 35. Vivanco I, Sawyers CL. The phosphatidylinositol 3-kinase AKT
14. Leicht DT, Balan V, Kaplun A, et al. Raf kinases: function, regulation pathway in human cancer. Nat Rev Cancer 2002;2:489e501.
and role in human cancer. Biochim Biophys Acta 2007;1773: 36. Kyu JP, Heum CC, Ramachandran S, et al. Augmentation of sodium
1196e1212. butyrate induced apoptosis by phosphatidylinositol 3-kinase
536 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

inhibition in the human cervical cancer cell-line. Cancer Res Treat 58. Huangh LQ, Zhang W, Yang Y, et al. Effects and its mechanism of
2006;38:112e117. quercetin on cervical cancer HeLa cells. Zhonghua Fu Chan Ke Za
37. Okkenhaug K, Vanhaesebroeck B. New responsibilities for the PI3K Zhi 2009;44:436e439.
regulatory subunit p85 alpha. Sci STKE 2001;65:1e5. 59. De S, Chakraborty J, Chakraborty RN, et al. Chemopreventive activ-
38. Fresno Vara JA, Casado E, de Castro J, et al. PI3K/Akt signalling ity of quercetin during carcinogenesis in cervix uteri in mice. Phyt-
pathway and cancer. Cancer Treat Rev 2004;30:193e204. other Res 2000;14:347e351.
39. Lawlor MA, Alessi DR. PKB/Akt: a key mediator of cell 60. Jin L, Qi M, Chen D, et al. Indole-3-carbinol prevents cervical cancer
proliferation, survival and insulin responses? J Cell Sci 2001;114: in human papilloma virus type 16 (HPV16) transgenic mice. Cancer
2903e2910. Res 1999;59:3991.
40. Choi Y, Zhang J, Murga C, et al. PTEN, but not SHIP and SHIP2, 61. Qi M, Anderson AE, Chen DZ, et al. Indole-3-carbinol prevents
suppresses the PI3K/Akt pathway and induces growth inhibition PTEN loss in cervical cancer in vivo. Mol Med 2005;11:59e63.
and apoptosis of myeloma cells. Oncogene 2002;21:5289e5300. 62. Virolle T, Adamson ED, Baron V, et al. The Egr-1 transcription factor
41. Liu D, Yang X, Songyang Z. Identification of CISK, a new member directly activates PTEN during irradiation-induced signalling. Nat
of the SGK kinase family that promotes IL-3-dependent survival. Cell Biol 2001;3:1124e1128.
Curr Biol 2000;10:1233e1236. 63. Bell MC, Crowley-Nowick P, Bradlow HL, et al. Placebo-controlled
42. Brunet A, Datta SR, Greenberg ME. Transcription-dependent and in- trial of indole-3-carbinol in the treatment of CIN. Gynecol Oncol
dependent control of neuronal survival by the PI3KeAkt signaling 2000;78:123e129.
pathway. Curr Opin Neurobiol 2001;11:297e305. 64. Higdon JV, Delage B, Williams DE, et al. Cruciferous vegetables and
43. Lu D, Qian J, Yin X, et al. Expression of PTEN and survivin in cer- human cancer risk: epidemiologic evidence and mechanistic basis.
vical cancer: promising biological markers for early diagnosis and Pharmacol Res 2007;55:224e236.
prognostic evaluation. Br J Biomed Sci 2012;69:143e146. 65. Powis G, Ihle N, Kirkpatrick DL. Practicalities of drugging the phos-
44. Chen YL, Law PY, Loh HH. Inhibition of PI3K/Akt signaling: an phatidylinositol-3-kinase/Akt cell survival signaling pathway. Clin
emerging paradigm for targeted cancer therapy. Curr Med Chem Cancer Res 2006;12:2964e2966.
Anticancer Agents 2005;5:575e589. 66. Bowles TL, Parsons C, Muilenburg DJ, et al. Targeted inhibition of
45. Narayan G, Bourdon V, Chaganti S, et al. Gene dosage alterations re- AKT in pancreatic cancer. Curr Cancer Ther Rev 2009;5:288e295.
vealed by cDNA microarray analysis in cervical cancer: identifica- 67. Bilodeau MT, Balitza AE, Hoffman JM, et al. Allosteric inhibitors of
tion of candidate amplified and overexpressed genes. Genes Akt1 and Akt2: a naphthyridinone with efficacy in an A2780 tumor
Chromosom Cancer 2007;46:373e384. xenograft model. Bioorg Med Chem Lett 2008;18:3178e3182.
46. Bertelsen BI, Steine SJ, Sandvei R, et al. Molecular analysis of the 68. Cherrin C, Haskell K, Howell B, et al. An allosteric Akt inhibitor
PI3K-AKT pathway in uterine cervical neoplasia: frequent PIK3CA effectively blocks Akt signaling and tumor growth with only transient
amplification and AKT phosphorylation. Int J Cancer 2006;118: effects on glucose and insulin levels in vivo. Cancer Biol Ther 2010;
1877e1883. 9:493e503.
47. Cho GW, Min SS, Namkoong H, et al. The phosphatidylinositol 3- 69. Tolcher AW, Yap TA, Fearen I, et al. A phase I study of MK-2206, an
kinase/Akt pathway regulates the HCCR-1 oncogene expression. oral potent allosteric Akt inhibitor (Akti), in patients (pts) with
Gene 2006;384:18e26. advanced solid tumor (ST). J Clin Oncol 2009;27:15s.
48. Gharbi SI, Zvelebil MJ, Shuttleworth SJ, et al. Exploring the speci- 70. Banerjee S, Li Y, Wang Z, et al. Multi-targeted therapy of cancer by
ficity of the PI3K family inhibitor LY294002. Biochem J 2007; genistein. Cancer Lett 2008;269:226e242.
404:15e21. 71. Pavese JM, Farmer RL, Bergan RC. Inhibition of cancer cell invasion
49. LoPiccolo J, Blumenthal GM, Bernstein WB, et al. Targeting the and metastasis by genistein. Cancer Metastasis Rev 2010;29:465e482.
PI3K/Akt/mTOR pathway: effective combinations and clinical con- 72. Su-Hyeon K, Sang-Chul L, Yong-Sang S. Involvement of both
siderations. Drug Resist Updat 2008;11:32e50. extrinsic and intrinsic apoptotic pathways in apoptosis induced by
50. Walker EH, Pacold ME, Perisic O, et al. Structural determinants of genistein in human cervical cancer cells. Ann NY Acad Sci 2009;
phosphoinositide 3-kinase inhibition by wortmannin, LY294002, 1171:196e201.
quercetin, myricetin, and staurosporine. Mol Cell 2000;6: 73. Yashar CM, Spanos WJ, Taylor DD, et al. Potentiation of the radia-
909e919. tion effect with genistein in cervical cancer cells. Gynecol Oncol
51. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the PI3k/Akt 2005;99:199e205.
pathway for cancer drug discovery. Nat Rev 2005;4:988e1004. 74. Hu HZ, Yan YB, Xu XD, et al. Oridonin induces apoptosis via PI3K/
52. Zhang W, Zhang F. Effects of quercetin on proliferation, apoptosis, Akt pathway in cervical carcinoma HeLa cell line. Acta Pharmacol
adhesion and migration, and invasion of HeLa cells. Eur J Gynecol Sin 2007;28:1819e1826.
Oncol 2009;30:60e64. 75. Yang JH, Luo SD, Wang YS, et al. Triterpenes from Tripterygium
53. Prasad SB, Yadav SS, Das M, et al. Down regulation of FOXO1 wilfordii Hook. J Asian Nat Prod Res 2006;8:425e429.
promotes cell proliferation in cervical cancer. J Cancer 2014;5: 76. Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22,989), a new anti-
655e662. fungal antibiotic. II. Fermentation, isolation and characterization. J
54. Fuhrman CB, Kilgore J, LaCoursiere YD, et al. Radiosensitization of Antibiot 1975;28:727e732.
cervical cancer cells via double-strand DNA break repair inhibition. 77. Yuan R, Kay A, Berg W, et al. Targeting tumorigenesis: development
Gynecol Oncol 2007;110:93e98. and use of mTOR inhibitors in cancer therapy. J Hematol Oncol
55. Powis G, Bonjouklian R, Berggren MM, et al. Wortmannin, a potent 2009;2:45.
and selective inhibitor of phosphatidylinositol-3-kinase. Cancer Res 78. Faivre S, Decaens T, Raymond E. mTOR and cancer therapy: clinical
1994;54:2419e2423. development and novel prospects. In: Polunovsky VA, Houghton PJ,
56. Arcaro A, Guerreiro AS. The phosphoinositide 3-kinase pathway in eds. Cancer Drug Discovery and Development. 12th ed. New York:
human cancer: genetic alterations and therapeutic implications. Curr Humana Press; 2010. pp. 133e148.
Genomics 2007;8:271e306. 79. Raymond E, Alexandre J, Faivre S, et al. Safety and pharmacoki-
57. Zhang F, Zhang T, Jiang T, et al. Wortmannin potentiates roscovitine- netics of escalated doses of weekly intravenous infusion of CCI-
induced growth inhibition in human solid tumor cells by repressing 779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol
PI3K/Akt pathway. Cancer Lett 2009;286:232e239. 2004;22:2336e2347.
Molecular Targets in Cervical Cancer 537

80. Temkin SM, Yamada SD, Fleming GF. A phase I study of weekly 100. Bellone S, Frera G, Landolfi G, et al. Overexpresion of epidermal
temsirolimus and topotecan in the treatment of advanced and/or growh factor type-1 receptor (EGF-R1) in cervical cancer: implica-
recurrent gynecologic malignancies. Gynecol Oncol 2010;117: tions for cetuximab-mediated therapy in recurrent/metastatic disease.
473e476. Gynecol Oncol 2007;106:513e520.
81. Campone M, Levy V, Bourbouloux E, et al. Safety and pharmacoki- 101. Hertlein L, Lenhard M, Kirschenhofer A, et al. Cetuximab monother-
netics of paclitaxel and the oral mTOR inhibitor everolimus in apy in advanced cervical cancer: a retrospective study with five pa-
advanced solid tumours. Br J Cancer 2009;100:315e321. tients. Arch Gynecol Obstet 2011;283:109e113.
82. Kim JW, Kim YT, Kim DK, et al. Expression of epidermal growth 102. Kurtz JE, Hardy-Bessard AC, Deslandres M, et al. Cetuximab, topo-
factor receptor in carcinoma of the cervix. Gynecol Oncol 1996; tecan and cisplatin for the treatment of advanced cervical cancer: A
60:283e287. phase II GINECO trail. Gynecol Oncol 2009;113:16e20.
83. Leserer M, Gschwind A, Ullrich A. Epidermal grow factor receptor 103. Moore KN, Sill MW, Miller DS, et al. A phase I trial of tailored ra-
signal transactivation. IUBMB Life 2000;49:405e409. diation therapy with concomitant cetuximab and cisplatin in the
84. Massague J, Pandiella A. Membrane-anchored growth factors. Annu treatment of patients with cervical cancer: a gynecologic oncology
Rev Biochem 1993;62:515e541. group study. Gynecol Oncol 2012;127:456e461.
85. Hackel PO, Zwick E, Prenzel N, et al. Epidermal growth factor re- 104. Santin AD, Sill MW, McMeekin DS, et al. Phase II trial of cetuximab
ceptors: critical mediators of multiple receptor pathways. Curr Opin in the treatment of persistent or recurrent squamous or non-squamous
Cell Biol 1999;11:184e189. cell carcinoma of the cervix: a Gynecologic Oncology Group study.
86. Zhou SY, Feng GF, Chen GR, et al. Analysis of epidermal growth Gynecol Oncol 2011;122:495e500.
factor receptor expression and gene expression status in tissue micro- 105. Berger C, Krengel U, Stang E, et al. Nimotuzumab and cetuximab
array of cervical squamous cell carcinoma. Zhonghua Fu Chan Ke Za block ligand-independent EGF receptor signaling efficiently at
Zhi 2013;48:843e846. different concentrations. J Immunother 2011;7:550e555.
87. Boiko IV, Mitchell MF, Hu W, et al. Epidermal growth factor recep- 106. Vera DR, Eigner S, Henke KE, et al. Preparation and preclinical eval-
tor expression in cervical intraepithelial neoplasia and its modulation uation of 177Lu-nimotuzumab targeting epidermal growth factor re-
during an alpha-difluoromethylornithine chemoprevention trial. Clin ceptor overexpressing tumors. Nucl Med Biol 2012;139:3e13.
Cancer Res 1998;4:1383e1391. 107. Ramakrishnan MS, Eswaraiah A, Crombet T, et al. Nimotuzumab, a
88. Shen L, Shui Y, Wang X, et al. EGFR and HER2 expression in pri- promising therapeutic monoclonal for treatment of tumors of epithe-
mary cervical cancers and corresponding lymph node metastases: lial origin. MAbs 2009;1:41e48.
implications for targeted radiotherapy. BMC Cancer 2008;8: 108. Blohmer J, Gore M, Kuemmel S, et al. Phase II study to determine
232. response rate, pharmacokinetics (PK), pharmacodinamics (PD),
89. Cho NH, Kim YB, Park TK, et al. P63 and EGFR as prognostic pre- safety, and tolerability of treatment with the humanized anti-
dictors in stage IIB radiation-treated cervical squamous cell carci- epidermal growh factor receptor (EGFR) monoclonal antibody
noma. Gynecol Oncol 2003;91:343e353. EMD 72000 (matuzumab) in patients with cervical cancer. Proc
90. Kim GE, Kim YB, Cho NH, et al. Synchronous coexpression of Am Assoc Cancer Res Am Soc Clin Oncol 2005;16:23.
epidermal growth factor receptor and cyclooxygenase-2 in carci- 109. Hurwitz H, Fehrenbacher L, Cartwright T, et al. Bevacizumab (a
nomas of the uterine cervix: a potential predictor of poor survival. monoclonal antibody to vascular endothelial growth factor) prolongs
Clin Cancer Res 2004;10:1366e1374. survival in first-line colorectal cancer (CRC): results of a phase III
91. Mendelsohn J, Baselga J. The EGF receptor family as targets for can- trail of bevacizumab in combination with bolus IFL (irinotecan, 5-
cer therapy. Oncogene 2000;19:6550e6565. fluorouracil, leucovorin) as first-line therapy in subjects with meta-
92. Mendelsohn J, Baselga J. Status of the epidermal growth factor re- static CRC [abstract]. J Clin Oncol 2003;22. Abstract 3646.
ceptor antagonists in the biology and treatment of cancer. Clin Can- 110. Wright JD, Viviano D, Powell MA, et al. Bevacizumab combination
cer Res 2003;21:2787e2799. therapy in heavily pretreated, recurrent cervical cancer. Gynecol On-
93. Lee JS, Kim HS, Park JT, et al. Expression of vascular endothelial col 2006;103:489e493.
growth factor in the progression of cervical neoplasia and its relation 111. Monk BJ, Still MW, Burger RA, et al. Phase II trial of bevacizumab in
to angiogenesis and p53 status. Anal Quant Cytol Histol 2003;25: the treatment of persistent or recurrent squamous cell carcinoma of the
303e311. cervix: a Gynecologic Oncology Group Study. J Clin Oncol 2009;27:
94. Dobbs SP, Hewett PW, Johnson IR, et al. Angiogenesis is associated 1069e1074.
with vascular endothelial growth factor expression in cervical intra- 112. Zighelboim I, Wright JD, Gao F, et al. Multicenter phase II trial of
epithelial neoplasia. Br J Cancer 1997;76:1410e1415. topotecan, cisplatin and bevacizumab for recurrent or persistent cer-
95. Hammes LS, Tekmal RR, Naud P, et al. Up-regulation of VEGF, c- vical cancer. Gynecol Oncol 2013;130:64e68.
fms and COX-2 expression correlates with severity of cervical cancer 113. Schefter TE, Winter K, Kwon JS, et al. A phase II study of bevaci-
precursor (CIN) lesions and invasive disease. Gynecol Oncol 2008; zumab in combination with definitive radiotherapy and cisplatin
110:445e451. chemotherapy in untreated patients with locally advanced cervical
96. Cooper RA, Wilks DP, Logue JP, et al. High tumor angiogenesis is carcinoma: preliminary results of RTOG 0417. Int J Radiat Oncol
associated with poorer survival in carcinoma of the cervix treated Biol Phys 2012;83:1179e1184.
with radiotherapy. Clin Cancer Res 1998;4:2795e2800. 114. Monk BJ, Lopez LM, Zarba JJ, et al. Phase II, open-label study of
97. Taja-Chayeb L, Chavez-Blanco A, Martinez-Tlahuel J, et al. Expres- pazopanib or lapatinib monotherapy compared with pazopanib plus
sion of platelet derived growth factor family members and the poten- lapatinib combination therapy in patients with advanced and recur-
tial role of imatinib mesylate for cervical cancer. Cancer Cell Int rent cervical cancer. J Clin Oncol 2010;28:3562e3569.
2006;6:22. 115. Schilder RJ, Sill MW, Lee YC, et al. A phase II trial of erlotinib in
98. Sato JD, Kawamoto T, Le AD, et al. Biological effects in vitro of recurrent squamous cell carcinoma of the cervix: a Gynecologic
monoclonal antibodies to human epidermal growth factor receptors. Oncology Group Study. Int J Gynecol Cancer 2009;19:929e933.
Mol Biol 1983;1:511e529. 116. Goncalves A, Fabbro M, Lhomme C, et al. A phase II trial to evaluate
99. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab gefitinib as second- or third-line treatment in patients with recurring
for squamous-cell carcinoma of the head and neck. N Engl J Med locoregionally advanced or metastatic cervical cancer. Gynecol On-
2006;354:567e578. col 2008;108:42e46.
538 Manzo-Merino et al./ Archives of Medical Research 45 (2014) 525e539

117. Nogueira-Rodrigues A, do Carmo CC, Viegas C, et al. Phase I trial of 138. Sun C, Cai M, Meadows RP, et al. NMR structure and mutagenesis of
erlotinib combined with cisplatin and radiotherapy for patients with the third BIR domain of the inhibitor of apoptosis protein XIAP. J
locally advanced cervical squamous cell cancer. Clin Cancer Res Biol Chem 2000;275:33777e33781.
2008;14:6324e6329. 139. Yuan ZM, Shioya H, Ishiko T, et al. p73 is regulated by tyrosine ki-
118. Vermeulen K, Van Bockstaele DR, Berneman ZN. Apoptosis: mech- nase c-Abl in the apoptotic response to DNA damage. Nature 1999;
anisms and relevance in cancer. Ann Hematol 2005;84:627e639. 399:814e817.
119. Denault JB, Salvesen GS. Caspases. Curr Protoc Protein Sci 2002; 140. Zhu J, Li Y, Guan C, et al. Anti-proliferative and pro-apoptotic ef-
Chapter 21:Unit 21.8. fects of 3, 30 -diindolylmethane in human cervical cancer cells. Oncol
120. Cho SG, Choi EJ. Apoptotic signaling pathways: caspases and stress- Rep 2012;28:1063e1068.
activated protein kinases. J Biochem Mol Biol 2002;35:24e27. 141. Wen X, Li D, Zhang Y, et al. Arsenic trioxide induces cervical cancer
121. Silke J, Meier P. Inhibitor of apoptosis (IAP) proteins-modulators apoptosis, but specifically targets human papillomavirus-infected cell
of cell death and inflammation. Cold Spring Harb Perspect Biol populations. Anticancer Drugs 2012;23:280e287.
2013;5:2. 142. Wodarz A, Nusse R. Mechanisms of Wnt signaling in development.
122. Fuentes-Gonzalez AM, Contreras-Paredes A, Manzo-Merino J, et al. Annu Rev Cell Dev Biol 1998;14:59e88.
The modulation of apoptosis by oncogenic viruses. Virol J 2013;10: 143. Peifer M, Polakis P. Wnt signaling in oncogenesis and embryogen-
182. esis: a look outside the nucleous. Science 2000;287:1606e1609.
123. Murray-Zmijewski F, Slee EA, Lu X. A complex barcode underlies 144. Austin TW, Solar GP, Ziegler FC, et al. A role for the Wnt gene fam-
the heterogeneous response of p53 to stress. Nat Rev Mol Cell Biol ily in hematopoiesis: expansion of multilineage progenitor cells.
2008;9:702e712. Blood 1997;89:3624e3635.
124. Wiley SR, Schooley K, Smolak PJ, et al. Identification and character- 145. Goodwin AM, D’Amore PA. Wnt signaling in the vasculature.
ization of a new member of the TNF family that induces apoptosis. Angiogenesis 2002;5:1e9.
Immunity 1995;3:673e682. 146. Ross SE, Hemati N, Longo KA, et al. Inhibition of adipogenesis by
125. Das H, Koizumi T, Sugimoto T, et al. Quantitation of Fas and Fas Wnt signaling. Science 2000;289:950e953.
ligand gene expression in human ovarian, cervical and endometrial 147. Polakis P. Wnt signaling and cancer. Genes Dev 2000;14:
carcinomas using real-time quantitative RT-PCR. Br J Cancer 1837e1851.
2000;82:1682e1688. 148. Liu C, Li Y, Semenov M, et al. Control of b-catenin phosphorylation/
126. Reesink-Peters N, Hougardy BM, van den Heuvel FA, et al. Death degradation by a dual-kinase mechanism. Cell 2002;108:837e847.
receptors and ligands in cervical carcinogenesis: an immunohisto- 149. He TC, Sparks AB, Rago C, et al. Identification of c-MYC as a target
chemical study. Gynecol Oncol 2005;96:705e713. of the APC pathway. Science 1998;281:1509e1512.
127. Maduro JH, Noordhuis MG, ten Hoor KA, et al. The prognostic value 150. Shtutman M, Zhurinsky J, Simcha J, et al. The cyclin D1 gene is a
of TRAIL and its death receptors in cervical cancer. Int J Radiat On- target of the b-catenin/LEF-1 pathway. Proc Natl Acad Sci USA
col Biol Phys 2009;75:203e211. 1999;96:5522e5527.
128. Kabsch K, Alonso A. The human papillomavirus type 16 (HPV-16) 151. Tetsu O, McCormick F. b-catenin regulates expression of cyclin D1
E5 protein sensitizes human keratinocytes to apoptosis induced by in colon carcinoma cells. Nature 1999;398:422e426.
osmotic stress. Oncogene 2002;21:947e953. 152. Behrens J, Jerchow BA, Wurtele M, et al. Functional interaction of an
129. Zhang B, Spandau DF, Roman A. E5 protein of human papilloma- axin homolog, conductin, with b-catenin, APC, and GSK3b. Science
virus type 16 protects human foreskin keratinocytes from UV B-irra- 1998;280:596e599.
diation-induced apoptosis. J Virol 2002;76:220e231. 153. Hsu W, Zeng L, Constantini F. Identification of a domain of axin that
130. Horinaka M, Yoshida T, Shiraishi T, et al. The combination of binds to the serine/threonine protein phosphatase 2A and a self-
TRAIL and luteolin enhances apoptosis in human cervical cancer binding domain. J Biol Chem 1999;274:3439e3445.
HeLa cells. Biochem Biophys Res Commun 2005;333:833e838. 154. Kikuchi A. Regulation of b-catenin signaling in the Wnt pathway.
131. Im SR, Jang YJ. Aspirin enhances TRAIL-induced apoptosis via Biochem Biophys Res Commun 2000;268:243e248.
regulation of ERK1/2 activation in human cervical cancer cells. Bio- €
155. Uren A, Fallen S, Yuan H, et al. Activation of the canonical Wnt
chem Biophys Res Commun 2012;424:65e70. pathway during genital keratinocyte transformation: a model for cer-
132. Maduro JH, de Vries EG, Meersma GJ, et al. Targeting pro- vical cancer progression. Cancer Res 2005;65:6199e6206.
apoptotic trail receptors sensitizes HeLa cervical cancer cells to 156. Pereira-Suarez AL, Meraz MA, Lizano M, et al. Frequent alterations
irradiation-induced apoptosis. Int J Radiat Oncol Biol Phys of the b-catenin protein in cancer of the uterine cervix. Tumour Biol
2008;72:543e552. 2001;23:45e53.
133. Kim JH, Kang JW, Kim MS, et al. The apoptotic effects of the flavo- 157. Yang HJ, Liu V, Wang Y, et al. Differential DNA methylation profiles
noid N101-2 in human cervical cancer cells. Toxicol In Vitro 2012; in gynecological cancers and correlation with clinico-pathological
26:67e73. data. BMC Cancer 2006;6:212.
134. Zhang Y, Ge Y, Chen Y, et al. Cellular and molecular mechanisms of 158. Kirikoshi H, Katoh M. Expression and regulation of WNT I0B in hu-
silibinin induces cell-cycle arrest and apoptosis on HeLa cells. Cell man cancer: up-regulation of WNT I0B in MCF-7 cells by beta-
Biochem Funct 2012;30:243e248. estradiol and down-regulation of WNT I0B in NT2 cells. Int J Mol
135. Liu RM, Li YB, Zhong JJ. Cytotoxic and pro-apoptotic effects of Med 2002;10:507e511.
novel ganoderic acid derivatives on human cervical cancer cells 159. Kirikoshi H, Sekihara H, Katoh M. Expression of WNT14 and
in vitro. Eur J Pharmacol 2012;681:23e33. WNT14B mRNAs in human cancer, up-regulation of WNT14 by
136. Narayanan BA, Holladay EB, Nixon DW, et al. The effect of all-trans INF-g and up-regulation of WNT14B by beta-estradiol. Int J Oncol
and 9-cis retinoic acid on the steady state level of HPV16 E6/E7 2001;19:1221e1225.
mRNA and cell cycle in cervical carcinoma cells. Life Sci 1998; 160. Koike J, Takagi A, Miwa T, et al. Molecular cloning of Frizzled-10, a
63:565e573. novel member of the Frizzled gene family. Biochem Biophys Res
137. Hembree JR, Pampusch MS, Yang F, et al. Cultured porcine Commun 1999;262:39e43.
myogenic cells produce insulin-like growth factor binding protein-3 161. Okino K, Nagai H, Hatta M, et al. Up-regulation and overproduction
(IGFBP-3) and transforming growth factor beta-1 stimulates of DVL-I, the human counterpart of the Drosophila disheveled gene,
IGFBP-3 production. J Anim Sci 1996;74:1530e1540. in cervical squamous cell carcinoma. Oncol Rep 2003;10:1223.
Molecular Targets in Cervical Cancer 539

162. Perez-Plascencia C, Vazquez-Ortiz G, Lopez-Romero R, et al. 164. Malanchi L, Peinado H, Kassen D, et al. Cutaneous cancer stem cell
Genomewide expression analysis in HPV16 cervical cancer: iden- maintenance is dependent of beta-catenin signaling. Nature Lett
tification of altered metabolic pathways. Infect Agent Cancer 2008;452:650e654.
2007;2:16. 165. Wang D, Wise ML, Li F, et al. Phytochemicals attenuating aberrant
163. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair follicle bulge activation of b-catenin in cancer cells. PLoS One 2012;7:e50508.
contribute to wound repair but not to homeostasis of the epidermis. 166. Anastas JN, Moon RT. WNT signaling pathways as therapeutic tar-
Nature Med 2005;11:1351e1354. gets in cancer. Nat Rev Cancer 2013;13:11e26.

You might also like