You are on page 1of 30

Rev. Med. Virol. 2015; 25: 24–53.

Published online in Wiley Online Library


(wileyonlinelibrary.com)
Reviews in Medical Virology DOI: 10.1002/rmv.1823

REVIEW
Signaling pathways in HPV-associated cancers
and therapeutic implications
Jiezhong Chen*
School of Biomedical Sciences and Australian Institute for Bioengineering and Nanotechnology, The
University of Queensland, Brisbane, Queensland, Australia

S U M M A RY
Human papillomaviruses (HPVs) are small double-stranded circular DNA viruses with 8 kb genomes. So far, more than
150 HPVs have been identified, and 12 types of HPVs have been conclusively linked to cancer by the International
Agency for Research on Cancer/World Health Organization. Expression of HPV E5, E6 and E7 oncoproteins can alter
multiple signaling pathways to cause cancer. In this review, the signaling pathways activated by these oncoproteins are
summarized, and targeted therapy against key signaling molecules is described. E6 can inactivate tumor protein 53 and
PDZ (post synaptic density protein–drosophila disk large tumor suppressor–zonula occludens-1 proteins) while stim-
ulating phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), Wnt and Notch pathways. E7 can inhibit retinoblas-
toma protein and stimulate the PI3K/Akt pathway. Both E6 and E7 can deregulate cellular microRNA expression,
which can alter cellular signaling pathways. E5 can sensitize epidermal growth factor receptor to epidermal growth fac-
tor to increase activation of PI3K/Akt and mitogen-activated protein kinase pathways. E5 can also inhibit the extrinsic
apoptotic pathway. These altered signaling pathways could be critical for the initiation and maintenance of HPV-
associated cancers. Therefore, targeted therapy against the key signaling molecules has therapeutic implications.
Among these, the possibilities of targeting PI3K/Akt, mammalian target of rapamycin, epidermal growth factor recep-
tor and vascular endothelial growth factor have been extensively studied in many cancers. Some inhibitors have been
studied in cervical cancer in both animal models and clinical trials. Although the results are promising, further inves-
tigation is warranted. Copyright © 2015 John Wiley & Sons, Ltd.
Received: 17 May 2014; Revised: 15 October 2014; Accepted: 27 December 2014

INTRODUCTION
*Correspondence to: J. Chen, School of Biomedical Sciences and Australian Human papillomaviruses (HPVs) are a family of
Institute for Bioengineering and Nanotechnology, The University of small double-circular-stranded DNA viruses with
Queensland, St Lucia, Brisbane, Queensland 4072, Australia. genomes containing 8 kb DNA sequences [1]. The
E-mail: j.chen4@uq.edu.au
sequences of some HPVs, such as HPV16 and
Abbreviations used HPV18, which are the most common cancer-related
Akt, protein kinase B; BMI, B cell-specific Moloney murine leukemia HPVs, encode two late genes L1 and L2 and six
virus integration site 1; CBF-1, C-promoter binding factor 1; CDK2,
cyclin-dependent kinase 2; COX-2, cyclooxygenase-2; CSCs, cancer early genes E1, E2, E4, E5, E6 and E7. Although
stem cells; CSL, CBF-1-Su (H) and LAG-1; CTGF, connective tissue E1, E2 and L1 and L2 proteins are essential for all
growth factor; CYR61, cysteine rich 61; DKK-1, dickkopf-related HPVs, expression of other HPV proteins varies in
protein family; EGF, epidermal growth factor; EGFR, epidermal
growth factor receptor; E6AP, E6-associated protein; FZD7, frizzled different HPVs. For example, HPVs 101, 103 and
family receptor 7; GIPC, GAIP-interacting protein, C terminus; 108 do not encode E6 [2], while HPV31 expresses
GSK3b, glycogen synthase kinase 3b; Hes, hairy and enhancer of split; E8 [3]. These HPV proteins maintain replication,
Hey, Hes-related repressor protein; HPV, human papillomavirus;
Lag-1, lobster agglutinin 1; LIM, Lin11, Isl-1 and Mec-3; MAGI-1, amplification and release of the viruses. Among
Membrane-associated guanylate kinase, WW and PDZ domain- them, E5, E6 and E7 are major oncogenes that pro-
containing protein 1; miRNAs, mircroRNAs; mTOR, mammalian mote host cell proliferation to facilitate viral ampli-
target of rapamycin; NF-κB, nuclear factor-κB; NFX1, nuclear tran-
scription factor, X-box binding 1; NICD, Notch intracellular domain; fication. E6 and E7 can be integrated into host
p53, tumour protein 53; PDK1, putative 3-phosphoinositide-dependent genomes in some cases and expressed in high
kinase 1; PI3K, phosphoinositide 3-kinase; pRb, retinoblastoma protein; levels, while E5 can only be expressed from viral
PTEN, phosphatase and tensin homologue; STAT-1, signal transducer
and activator of transcription 1; TIP-2, Tax-interacting protein-2; genomes and maintained as episomes in host cells
VEGF, vascular endothelial growth factor. [4,5]. Infections with some HPVs cause cancers

Copyright © 2015 John Wiley & Sons, Ltd.


Signaling pathways in HPV-associated cancers 25

including cervical cancer, head and neck cancer,


vulvar cancer, vaginal cancer, penile cancer and
anal cancer. So far, more than 150 types of HPVs
have been identified. Among them, 12 types of
HPVs can cause cancer (http://monographs.iarc.
fr/ENG/Monographs/vol90/index.php), and the
list could be expanded with more cancer-related
HPVs identified. HPV16 and HPV18 are responsi-
ble for 50% and 20% of cervical cancer respectively
[5]. Although HPV infections are very common,
only a very small percentage of HPV-infected peo-
ple develop cancer. HPV infections elicit immune
responses, which clear most HPV viruses. Persis- Figure 1. Signaling molecules altered by E6 protein. Human papil-
lomavirus E6 inhibit tumor protein (p)53, PDZ proteins and some
tent infections greatly increase the risk for carcino- microRNAs (miRNAs). E6 activates protein kinase B (Akt), Notch
genesis. Understanding the mechanisms of HPV- and Wnt pathways. E6 also increases telomerase activity. PDZ, post
caused cancer could be helpful for the prevention synaptic density protein–drosophila disk large tumor suppressor–
and treatment of the disease. zonula occludens-1 protein

Differences in the sequences caused by amino


acids as well as variation in codon usage of E5, E6 proliferation, apoptosis, genomic instability, migra-
and E7 genes have been associated with their tion and drug resistance, the outcomes of targeted
ability to initiate cancer [6–9]. A study has shown therapy against several altered signaling molecules
that E7 is sufficient to immortalize cells, while are described.
E6 can increase the effect of E7, that is E6 and E7
can cooperate to immortalize primary human HUMAN PAPILLOMAVIRUS E6 PROTEIN
epithelial cells. The complementary effects of E6 Human papillomavirus E6 is a small protein
and E7 could be explained by their different down- without enzymatic activity. For example, HPV16
stream targets. Inhibition of E6 led to increased E6 consists of 151 amino acids. E6 protein acti-
tumor protein 53 (p53) but not retinoblastoma vates several carcinogenic pathways and inhibits
protein (pRb), while inhibition of E7 resulted in tumor suppressor protein p53 and post synaptic
increased pRb but not p53 [10]. E5 is also an density protein–drosophila disk large tumor
oncoprotein, which promotes carcinogenic signal- suppressor–zonula occludens-1 protein (PDZ)
ing pathways to increase the effects of E6 and E7. (Figure 1). The survival pathways activated by
However, E6 and E7 together are not sufficient, re- E6 include phosphoinositide 3-kinase (PI3K)/pro-
quiring introduction of another cancer risk factor tein kinase B (Akt), Wnt and Notch. E6 activates
for tumor formation. For example, oncogene Ras telomerase expression and activity, thereby
has been shown to cause cancer together with E6 inhibiting telomere shortening and increasing cell
and E7 [11]. Deletion of tumor suppressor gene immortalization. Several miRNAs have been al-
RXRα and expression of E6 and E7 together have tered by E6, leading to signaling pathway
also been shown to cause malignant lesions of cer- changes.
vical tissue [12].
Many studies have been performed to elucidate Inactivation of p53 protein
the mechanisms for E6 and E7 to cause cancer, Protein p53, encoded by TP53 gene, is a well-
showing their interactions with many signaling known tumor suppressor, which prevents cancer
molecules. In this review, the effects of these initiation by maintaining genomic stability
oncoproteins on intracellular signaling pathways [13,14]. It is activated by DNA damage and envi-
are summarized in association with the roles of ronmental stimuli such as oxidative stress and
these signaling pathways in carcinogenesis and osmotic shock as well as viral infections [13,14].
cancer progression. Several microRNAs (miRNAs) Activated p53 can slow down the cell cycle to
altered by E6 and E7 and associated cell biological allow damaged DNA to be repaired. Activated
activities are also described. After a discussion of p53 binds to DNA to increase expression of protein
the changed pathways on cell behaviors including p21, which inhibits cyclin-dependent kinase 2

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
26 J. Chen

(CDK2) to decrease G1/S transition. Activated p53 HPV amplified very poorly [21]. Silencing of p53
can also initiate apoptosis if DNA is severely dam- or expression of ectopic wild type E6 partially re-
aged. Through these processes, p53 prevents accu- stored amplification, whereas three missense E6
mulation of cells with DNA damage so that it mutations that did not effectively destabilize p53
decreases tumor initiation. In contrast, inactivation complemented the null mutant poorly. E6 can de-
of p53 results in the accumulation of abnormal grade p53 in E6AP-null mice indicating that other
cells that have gene mutations, facilitating cancer ubiquitin ligases may also be involved in E6-
initiation. mediated p53 degradation [22].
E6 protein inactivates p53 and thus avoids apo- The ability of E6 to degrade p53 has been corre-
ptosis of host cells that have DNA damage. High- lated with HPV’s ability to cause cancer. Mesplede
risk type HPV E6 binds to an LXXLL motif (where et al. examined the p53 degradation ability of E6
L = leucine and X= any amino acid) on cellular E3 proteins from 29 types of HPVs [23]. It was found
ubiquitin ligase E6-associated protein (E6AP) [15– that variation of the p53 degradation ability was
17]. The formed E6-E6AP complex then recruits more than 100 times with high-risk HPVs having
and ubiquitinates p53, which facilitates a higher ability to degrade p53. It was also found
proteasome-mediated degradation of p53 (Figure 2) that the variants of a type of HPV such as HPV16
[18,19]. This E6-induced p53 decrease has been rec- and HPV33 had different p53 degradation ability
ognized as a major mechanism for E6 to cause can- but the difference was much less than in different
cer. Inactivation of p53 introduced by dominant types of HPVs.
negative p53 is sufficient to maintain HPV DNA While it is certain that degraded p53 by HPV E6
with E6 inactivating mutations in human via E6AP plays a critical role in HPV-associated
keratinocytes [20]. In organotypic cultures, an E6 cancer, other alternatives have also been revealed.
null mutant accumulated high levels of p53, and Studies reported that HPV E6 also degraded other
two members of p53 family p63β and p73 [24,25].
These two proteins have similar structures to p53
and also can inhibit cell proliferation and promote
apoptosis [26]. Thus, inactivation of p63β and p73
is important for cancer development [27]. Park
et al. has shown that E6 directly binds to and inac-
tivates p73, but it is not via E3 ubiqitination [25].

Inhibition of post synaptic density protein–


drosophila disk large tumor suppressor–
zonula occludens-1 proteins
The PDZ is a structural domain of 80–90 amino
acids, which is shared by many proteins and
named from the initial letters of three proteins—
post synaptic density protein (PSD95), drosophila
disk large tumor suppressor (Dlg1), and zonula
occludens-1 protein (zo-1) [28]. There are now more
than 250 proteins that have a PDZ domain. PDZ
proteins interact with c-tail amino acids of targeted
proteins to regulate multiple biological processes,
including cell adhesion, tight junction, cell polarity,
cytoskeleton, ion channels and signaling pathways.
Figure 2. E6/p53 pathway. Tumor protein (p)53 family members can Some PDZ proteins are tumor suppressor proteins,
maintain genomic stability through decreasing cell cycle and and thus, loss of these proteins facilitates cancer
increasing apoptosis under stimulation such as ultraviolet (UV), formation [29]. High-risk types of HPV E6 proteins
hypoxia and viral infections. Human papillomavirus E6 can bind
to ubiquitin ligase E6-associated protein (E6AP) or other ligases to
have a PDZ-binding motif at their extreme carboxy
degrade p53. E6 can also act on p63 and p73 directly to destabilize termini [30–32]. Binding of E6 to PDZ proteins re-
these proteins sults in dysfunction of these proteins.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 27

Both HPV16 and HPV18 E6 bind to Dlg C- Several studies have shown that E6 can activate
terminus for transformation and mutation of E6 this pathway through various mechanisms. E6
loses the ability to bind and transform rodent cells inactivates PTEN through PDZ proteins, leading
[30,31]. HPV18 E6 protein interacts with TIP-2/ to increased pAkt as well as increased cell prolif-
GIPC, leading to polyubiquitination and eration [37,38,47]. The mammalian target of
proteasome-mediated degradation of the protein rapamycin (mTOR) kinase is a downstream target
[33]. In HeLa cells, RNAi silencing of E6 decreases of Akt and also activated by blood levels of amino
TIP-2/GIPC levels. TIP-2/GIPC can increase ex- acids and mitogen-activated protein kinase
pression of the TGF-beta type-III receptor at the (MAPK) pathway. It has been demonstrated that
cell membrane and thus increase the antiprolifera- mTOR is activated by E6 as indicated by increased
tive effect of TGF-beta. Depletion of TIP-2/GIPC ribosomal protein S6 kinase, which is regulated by
in HeLa cells reduced the antiproliferative effect mTOR [48,49]. E6-E6AP complex binds and de-
of TGF-beta, while silencing of E6 blocked the pro- grades the mTOR inhibitor tuberous sclerosis
liferation of HeLa cells. complex 2 (TSC2). Another study also showed
HPV E6 acts on PDZ-partitioning defective 3 that HPV16 E6 expression caused an increase in
protein, which regulates tight junctions and cell mTOR complex 1 activity indicated by enhanced
polarity [34,35]. E6 caused translocation of PDZ- phosphorylation of mTOR as well as its down-
partitioning defective 3, leading to the loss of cell stream targets ribosomal protein S6 kinase and eu-
polarity to facilitate tumor formation. Massimi karyotic initiation factor binding protein 1 under
et al. showed that E6 targeted hDlg, hScrib conditions of nutrient deprivation [50]. However,
(Scribble), MUPP1, membrane-associated a decrease in TSC2 levels in HPV16 E6-expressing
guanylate kinase, WW and PDZ domain- cells was not detected. Instead, upstream kinases
containing protein 1 (MAGI-1), MAGI-2 and putative 3-phosphoinositide-dependent kinase 1
MAGI-3 through E6AP to increase transformation (PDK1) and mTOR complex 2 were activated,
[35,36]. HDlg and MAGI-2 interact with phospha- leading to increased phosphorylation of Akt [50].
tase and tensin homologue (PTEN), which regu- In human foreskin keratinocyte (HFK) cells, ex-
lates the PI3K/Akt pathway [37,38]. E6 has high pression of HPV16 E6 resulted in sustained activa-
affinity for the PDZ domain of MAGI-1 protein, tion of receptor protein tyrosine kinases including
and mutation of the residue lysine 499 abolished epidermal growth factor receptor (EGFR), insulin re-
the effect of E6 on MAGI-1 [39]. ceptor beta and insulin-like growth factor receptor-
beta, which are upstream of the PI3K/Akt pathway
Activation of phosphoinositide [51]. E6 can also increase signaling adaptor protein
3-kinase/protein kinase B growth factor receptor-bound protein 2 to activate
The PI3K/Akt pathway is a major cancer survival the PI3K pathway [51].
pathway (Figure 3) [40,41]. PI3K regulates Akt and Activation of Akt can produce a cascade of
Rac-1. Akt has a broad range of downstream targets, changes in downstream targets. Akt can phosphor-
which control cell proliferation, cell growth, cell ylate E6 to promote its ability to interact with pro-
mobilization, angiogenesis and cell survival [40,41]. tein 14-3-3σ, which is important in carcinogenesis
The pathway has been associated with increased [52]. HPV has also been associated with increased
cancer initiation, progression, metastasis and drug expression of c-myc, a downstream protein of Akt
resistance. Thus, inhibition of the pathway has been [53–56]. E6 has been reported to act directly on c-
proposed for the treatment of cancer [42]. Targeted myc, leading to activation of telomerase activity
therapy of PI3K/Akt has been studied extensively [10,57]. Controversially, two studies showed that
in many cancers such as breast cancer, melanoma, E6 accelerated c-myc degradation [58,59].
colon cancer and prostate cancer [40,43,44]. Both While it is certain that E6 can cause activation
genetic defects and environmental factors are in- of the PI3K/Akt pathway and its downstream
volved in activation of the pathway. Gene defects targets nuclear factor-κB (NF-κB), mTOR, 14-3-3
have been detected in almost all elements of this and c-myc, the effects of E6 on other downstream
pathway [42]. Obesity has been shown to increase targets of Akt are largely not studied. It will
this pathway, leading to increased cancer inci- also be interesting to investigate the biological
dence [45,46]. effects mediated by the E6/PI3K/Akt pathway

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
28 J. Chen

Figure 3. E6/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. PI3K catalyzes phosphatidylinositol-4,5-bisphosphate (PIP2) into
phosphatidylinositol-3,4,5-trisphosphate (PIP3), which promotes (PDK1) to phosphorylate Akt. Activated Akt can influence cell survival through
mitochondrial Bcl-2 family members, cell proliferation through cyclin D1 and nuclear factor-κB (NF-κB), cell growth through mammalian target
of rapamycin and angiogenesis through vascular endothelial growth factor (VEGF). Akt blocks apoptosis through decreasing tumor protein (p)
53, p21 and p27. Akt also increases genomic instability. E6 can activate PI3K through receptor protein tyrosine kinase or direct interaction with
PI3K. E6 can also block tuberous sclerosis complex 1/2 (TSC1/2) to increase mammalian target of rapamycin complex 1 (mTORC1) activity to in-
crease cell growth and can block pro-apoptotic proteins Bad and Bax to decrease apoptosis. PTEN, phosphatase and tensin homologue; RPTK,
receptor protein tyrosine kinase; HIF, hypoxia-induced factor; S6K, ribosomal protein S6 kinase; GSK, glycogen synthase kinase; Rheb, Ras
homologue enriched in brain; 4E-BP, eukaryotic initiation factor 4E binding protein; MDM2, murine double minute; Foxo1, forkhead box O1;
MAPK, mitogen-activated protein kinase; mTORC2, mammalian target of rapamycin complex 2

in terms of cell proliferation, survival, migration phosphorylated at serine and threonine residues.
and drug resistance. Phosphorylation of β-catenin induces its
ubiquitination by β-TcRP ubiquitin ligase, leading
Activation of the Wnt pathway to degradation. In activation status of canonical
Wnt ligands and the associated pathway regulate Wnt signaling pathway, intracellular Dishevelled
cellular proliferation and differentiation processes protein is phosphorylated and interacts with
and thus play critical roles in normal tissue homeo- Axin2, leading to dysfunction of the degradation
stasis [60] and in pathologic conditions such as complex and accumulation of β-catenin. The accu-
cancers [61–63]. Activation of the Wnt pathway mulated β-catenin is translocated into the nucleus
results in accumulation of β-catenin, which in turn and binds members of the T-cell factor/lymphoid
increases transcription of a broad range of genes to enhancer factor family of transcription factors to
promote cell proliferation. In inactivation status of regulate target genes including c-jun, c-myc [64],
the Wnt pathway, β-catenin forms a “degradation cyclin D1 [65], multidrug resistance 1[66],
complex” with other proteins including glycogen matrilysine [67], Axin2 [68], survivin, vascular en-
synthase kinase-3β (GSK3β), casein kinases, ade- dothelial growth factor (VEGF), COX-2 and matrix
nomatous polyposis coli and Axin2 and metalloproteinases [69]. It also targets positive and

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 29

negative regulators of the Wnt pathway including glioma and head and neck cancers. The pathway
Axin2, Wg, FZD7, DKK-1 and sFRP-2 to form auto- comprises four Notch receptors (Notch 1–4 in
regulation [69]. humans), a group of transmembrane proteins, and
Accumulated nuclear β-catenin has been com- their ligands (Jagged1, 2 and delta-like ligand 1, 3
monly found in human HPV16-positive invasive and 4) [82,83]. Binding to their ligands on the sur-
cancer samples but also in early dysplastic lesions, face of neighboring cells leads to the cleavage of
indicating activation of the Wnt pathway by HPV Notch receptor by γ-secretase and subsequent re-
oncogenes [70,71]. The nuclear accumulation of β- lease of the Notch intracellular domain (NICD)
catenin correlates with tumor progression in cervi- (Figure 4). As the constitutively active domain of
cal cancer patients [72]. The accumulated β-catenin the Notch receptor, NICD translocates to the nu-
also correlates with HPV infection in cell lines in- cleus where it binds to and forms a complex with
cluding HPV16-positive oropharyngeal cancer cell the transcriptional regulator termed CBF-1-Su (H)
lines 147 T and 090, HPV-negative cell line 040 T and LAG-1 (CSL) and mastermind-like protein,
and cervical cell lines SiHa (bearing integrated leading to the displacement of co-repressors previ-
HPV16) and HeLa (bearing integrated HPV18) [73]. ously bound to CSL and recruitment of
E6 gene silencing in HPV-positive cells reduced nu- co-activators. The co-activators then induce expres-
clear β-catenin substantially, suggesting a critical role sion of the target genes, such as the hairy and en-
of E6 in the activation of the Wnt pathway. The hancer of split (Hes) and Hes-related repressor
mechanism has been associated with a protein called protein (Hey) families. Accumulating evidence
seven in absentia homologue, which increases indicates that dysregulated NCID can also activate
proteasomal degradation of β-catenin. E6 can de- the PI3K/Akt pathway, which in turn causes a
crease seven in absentia homologue mRNA and pro- cascade of target proteins [84–88].
tein levels substantially [73]. Lichtig et al. also The expression of Notch 1 increases with progres-
showed that HPV16 E6 activated the Wnt/β-catenin sion of cervical cancer; Notch 1 has been detected in
pathway; the mechanism is independent of the abil- invasive cervical cancer but not low-grade cancer
ity of E6 to target p53 for degradation or bind to the [89]. Daniel et al. showed that Notch-1 receptor ex-
PDZ-containing E6 targets but requires E6AP [74]. pression was increased during the progression from
In vivo mouse experiments showed that E6 expres- cervical intraepithelial lesions to invasive cervical
sion led to accumulation of β-catenin [75]. Full- carcinoma [90]. Moreover, main cellular localization
length E6 oncoprotein expression in K14E6 mice en- of Notch-1 protein changed from cytoplasmic to nu-
hanced the nuclear accumulation of β-catenin and clear with the transition from intraepithelial lesions
the accumulation of cellular β-catenin-responsive III to microinvasive carcinoma [90]. The regulation
genes. These effects were not observed when a trun- of the Notch signaling pathway by HPV E6 has been
cated E6 oncoprotein that lacks the PDZ-binding do- demonstrated in cervical cancer cell lines. At late
main was expressed alone (K14E6ΔPDZ mice), passage but not early W12 cell line, which is an
indicating that the effect of E6 was mediated by the HPV type-16-positive human cervical low-grade
PDZ domain [75]. Although the Wnt pathway may lesion-derived cell line, jagged1 is upregulated,
be a possible mediator for increased β-catenin, while manic fringe is decreased [91]. It was con-
PI3K/Akt is also well known to cause accumulation firmed by an increase of Notch-driven report activity
of β-catenin through inactivation of GSK3β [76]. and a decrease of manic fringe promoter-driven re-
Therefore, further studies are needed for a unified port activity in late passage W12. Inhibition of the
explanation of E6-induced β-catenin accumulation. Notch pathway by expression of manic fringe,
dominant-negative Jagged1 or RNAi silencing of
Activation of the Notch pathway Jagged1 inhibits the tumorigenicity of CaSki, an in-
Notch signaling plays important roles in both nor- vasive cervical carcinomas-derived cell line. HPV in-
mal physiological activities such as cell growth, dif- creases the Notch pathway through distinct
ferentiation, immune responses and organ mechanisms [92]. HPV16 E6 can interact with cellu-
development and various diseases including cancer lar protein NFX1-123 and increase NFX1-123 protein
[77–81]. Increased expression of the Notch pathway expression [92]. Overexpression of NFX1-123 in
has been associated with progression of several ma- HPV16 E6-expressing keratinocytes increased
lignancies such as prostate cancer, breast cancer, Notch-1 mRNA levels. Weijzen et al. demonstrated

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
30 J. Chen

Figure 4. E6/Notch pathway. Notch receptor is activated by its ligand Jagged1 from another cell and cleaved by gamma secretase to pro-
duce Notch intracellular domain (NICD), which translocates into the nucleus to form a complex with CBF-1-Su (H) and LAG-1 (CSL) and
mastermind-like protein (maml) to promote transcription of hairy and enhancer of split (Hes) and Hes-related repressor protein (Hey).
NECD, Notch extracellular domain; URR, upstream regulatory region

that presenilin-1 mediated the effect of E6 on the However, a recent study showed that the Notch
Notch pathway in mouse and human primary cell pathway was activated in 32% of HNSCC patient
lines transfected HPV16 E6 ([93]). samples and only 9% (4 out of 37) had inactivation
A controversial opinion about the Notch pathway of the pathway, casting doubt whether inactivation
in HPV-associated cancer has been proposed. It was of Notch is required for these cancers [101].
shown that Notch 1 was decreased in HPV-positive
cervical cancer cell lines HeLa, C40I, C4-II, SiHa and Telomerase activation
Caski compared with HPV-negative C33a and pri- Telomerase is a ribonucleoprotein enzyme for main-
mary keratinocytes [94]. However, expression of taining telomeric structures at the end of chromo-
Notch 2 in all these cell lines was similar to primary somes, and its increased activity is important for
keratinocytes. Nevertheless, overexpression of carcinogenesis [102–105]. In normal cells, telome-
Notch 1 in HPV-positive cells decreased their rase activity is very low so that telomeres shorten
growth. This indicates that Notch could be a tumor with every round of DNA replication and become
suppressor in cervical cancer as in some cancers shorter over time, which leads to replicative senes-
such as chronic myelomonocytic leukemia, cutane- cence. Expression of the telomerase catalytic sub-
ous and lung cancers [95–97]. It has been demon- unit in normal cells increased telomere length,
strated that Notch 1 is controlled by p53. resulting in indefinite cell proliferation [106]. In can-
Knockdown of p53 by short interfering RNA cer cells, inhibition of telomerase activity decreased
(siRNA) decreased Notch-1 levels, while activation cell proliferation [107]. Telomerase activation is crit-
of p53 by nutlin increased Notch-1 levels in human ical for the immortalization of primary human
keratinocyte cancer cell lines [98]. Suppression of keratinocytes by the high-risk HPV E6 [108].
the Notch signaling pathway together with acti- E6 is able to increase telomerase activity by up-
vated Ras resulted in cancer cell formation in pri- regulation of telomerase reverse transcriptase
mary keratinocytes, providing evidence for (TERT), which is encoded by the human telomerase
carcinogenic effect of loss of Notch 1[98]. There were reverse transcriptase (hTERT) gene [108,109]. E6 in-
also reports that Notch was lost because of Notch 1 duces the hTERT promoter via interactions with the
inactivating mutation in 10–15% of patients and the cellular ubiquitin ligase, E6AP. E6 increases hTERT
loss could increase head and neck squamous cell via NFX1-123. NFX1-123 interacts with hTERT
carcinomas (HNSCC) carcinogenesis [99,100]. mRNA and stabilizes it, leading to greater

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 31

telomerase expression [92]. E6 protein also interacts not HPV6 E6 reduced miR-218 expression, while si-
directly with the hTERT protein [110]. Unlike its ef- lencing of E6 increased miR-218 expression in cervi-
fect on p53, E6 binding to hTERT protein does not cal cancer cells. LAMB3, a laminin protein, is
cause protein degradation both in vitro or in vivo increased in cervical cancer specimens and identi-
but increases telomerase activation by a post- fied as a target of miR-218 [113]. Therefore, LAMB3
transcriptional mechanism [110]. was increased by E6 [112]. LAMB3 regulates cell dif-
ferentiation, migration, adhesion, proliferation and
MicroRNAs involved in E6-mediated survival, so silencing of LAMB3 in cervical cancer
signaling pathways cells reduced cell survival and migration [113]. Con-
MicroRNAs are small non-coding RNAs contain- sistently, overexpression of miR-218 also reduced
ing 19–24 nucleotides, which regulate gene ex- cancer cell migration and invasion in both HPV-
pression by targeting transcripts or translation positive and HPV-negative cervical cancer cell lines.
[111]. In humans, a large number of genes are reg- E6 decreases miR-125b [124], which inhibits
ulated by miRNAs, so miRNAs regulate many cervical cancer growth and promotes apoptosis
biological processes. Studies have shown that via inhibition of the PI3K/Akt pathway [125].
many miRNAs are involved in E6-mediated sig- Another study reported that miR-125b was highly
naling pathways. Martinez et al. used Ambion reduced in HPV-positive cells [126]. However, a
(Ambion Technologies, Austin, TX, USA) arrays recent study showed that miR-125 was increased
to show that three miRNAs were overexpressed in cervical cancer and miR-125b could target the
and 24 underexpressed in cervical cell lines con- Bak promoter to reduce its expression and thus
taining integrated HPV-16 DNA [112]. Another decrease apoptosis [127]. This discrepancy has been
study showed that miRNAs miR-363, miR-33 and explained by multiple targets of miR-125b with its
miR-497 were upregulated, whereas miR-155, roles depending on different expression levels of
miR-181a, miR-181b, miR-29a, miR-218, miR-222, target genes [128].
miR-221 and miR-142-5p were downregulated in MicroR-375 is decreased in HPV-associated can-
HPV-positive head and neck cancer cells [113]. cer, which negatively regulates HPV16 and 18 tran-
E6 decreases miR-34a [114,115], which is an im- scripts [129]. MicroR-375 directly acts on E6AP and
portant cell cycle regulator [116,117]. MicroRNA- decreases its activity [129]. Thus, transfection of
34a is a target of p53, and the effect of E6 on miR-375 resulted in increased p53, p21 and 14-3-3.
miRNA-34a is mediated by decreased p53 In gastric cancer, miR-375 was shown to inhibit
[114,115]. This is consistent with the fact that PDK1 and 14-3-3zeta directly, and overexpression
miR-34a is reduced in both normal cervical tissue of miR-375 increased apoptosis via the caspase
and cervical lesions with high-risk HPV infection pathway [130].
[118]. Targeting of HPV16 E6 has been shown to E6 could regulate more miRNAs, so the list
increase miR-34a [119]. One of the targets of miR- might increase after more studies are performed.
34a is p18Ink4c [120]. Silencing of E6 or overex- A recent study showed that in organotypic raft cul-
pression of miRNA-34a in HeLa cells decreased tures of foreskin and vaginal keratinocytes HPV16
p18Ink4c, while inhibition of miRNA-34a in- and HPV18 changed 13 miRNAs. MicroR-25, miR-
creased p18Ink4c. Protein p18Ink4c is an inhibitor 92a and miR-378 were increased, while miR-22,
of CDK4/6, but its role in cervical cancer is not miR-27a, miR-29a and miR-100 were decreased.
well studied. Other targets of miR-34a have also The increased miR-25, miR-92a and miR-378 were
been revealed. MicroRNA-34a is negatively corre- confirmed in cervical patients’ specimens [131].
lated with CDK4, Sirt1 and myeloblastosis protein The significance of these changes in cellular signal-
[121]. In cell biology, miRNA-34a has been associ- ing pathways is not elucidated yet.
ated with hypoxia increased epithelial-
mesenchymal transition and docetaxel resistance HUMAN PAPILLOMAVIRUS E7 PROTEIN
in breast cancer [122,123]. E7 is a potent cell cycle regulator with 98 amino
Martinez et al. showed that miR-218 was particu- acids containing three domains of E7 called con-
larly reduced in HPV-positive cervical cancer indi- served regions 1–3. It interacts with more than 50
cated by microarray, real-time PCR and northern cellular factors [132,133]. The significance of most
blot analyses [112]. Expression of HPV16 E6 but of these interactions is unknown. However, several

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
32 J. Chen

downstream targets have been identified including [147]. However, knockout of three Rb family mem-
its interactions with pRb, the PI3K/Akt pathway bers in mice is not sufficient to cause cervical carci-
and miRNAs. noma although development of high-grade cervical
intraepithelial neoplasia can develop [148].
Inhibition of retinoblastoma protein
Dysfunction and inactivation of pRb through vari- Activation of phosphoinositide
ous factors have been associated with initiation of 3-kinase/protein kinase B
many cancers [134–136]. E7 is one factor that inacti- Several studies have shown that E7 can activate
vates pRb [137,138] (Figure 5). the PI3K/Akt pathway. Menges et al. expressed
Several mechanisms for E7 to alter pRb signaling HPV16 E7 in HFK cells and showed upregulation
have been elucidated [133,139–141]. Retinoblastoma of Akt activity in organotypic raft cultures [149].
protein is a pocket protein, which binds to transcrip- The mechanism of increased Akt activity has been
tion factors E2F1-3 to prevent E2F1-3 functions. When regarded as inhibition of pRb by E7 as follows.
E7 binds its pocket and inactivates its function to bind First, the ability of E7 to increase Akt activity is
E2F1-3, E2F1-3 is released. E2F1-3 can bind cell cycle correlated with its ability to bind to and inactivate
regulators DP-1 or DP-2, leading to increased cell Rb. Second, silencing of Rb by short hairpin RNAs
cycle from G1 to S, decreased apoptosis and increased (shRNAs) in differentiated keratinocytes increased
genomic instability. HPV16 E7 proteins can destabi- Akt activity. Third, increased Akt activity and loss
lize pRb through proteasomal degradation mediated of Rb were also correlated in HPV-positive cervical
by cullin 2 ubiquitin ligase complex reprogrammed high-grade squamous intraepithelial lesions.
by HPV E7 [142]. A difference in an amino acid in Protein kinase B upregulation by E7 may be
E7 sequence could affect its binding ability and degra- mediated by protein phosphatase 2A (PP2A)
dation ability, accounting for carcinogenic ability of [150,151]. PP2A is known to inhibit Akt phosphory-
high-risk or low-risk HPVs [143,144]. lation. Pim et al. demonstrated that E7 could bind
E7 also acts on E2F proteins directly. It binds to both the Mr 35 000 catalytic subunit and the Mr
E2F1 and increases its activity [145]. E7 can also 65 000 structural subunit of PP2A to inhibit PP2A
block the transcriptional suppressor activity of activity [150]. Liu et al. found that PP2A mRNA
E2F6 [146]. E2F6 is usually upregulated by E2F1 and protein levels were detected in 73% and 53% cer-
to provide a feedback regulation [146]. vical cancer samples respectively but not in normal
In addition, E7 also inactivates other pocket pro- cervical tissues [151]. Knockdown of E7 downregu-
teins including p130 and p107 [137]. Proteins p130 lated both mRNA and protein expression of PP2A.
and p107 regulate E2F4 and E2F5, which also play E7 negatively regulates p27 via Akt. HPV16 E7
key roles in cell cycle regulation. The effects of E7 enhanced both the cytoplasmic retention of p27 in
on three pRb family members may be synergistic HFKs, which was reduced by PI3K/Akt inhibition
[152]. A standard wound assay showed that E7 in-
creased the migration of HFKs, which was
Akt/p27 dependent.
Rho family guanosine triphosphatases (GTPases)
play critical roles in cytoskeleton and cell migra-
tion. Under extracellular environment stimulation,
GTPases can transfer inactive guanosine
diphosphate-bound states to the active guanosine
triphosphate-bound form, which in turn changes
cell morphology and promotes cell migration
through Rho family members [153–156]. HPV E7
regulates Rho family members to increase cell mi-
gration through Rac1 and RhoA [152,157]. RhoA
Figure 5. E7/retinoblastoma protein (pRb) pathway. pRb forms a is crucial for efficient cell migration and cell spread-
complex with E2F1 to keep E2F1 in inactivation status. E7 can
ing. Todorovic et al. used mass spectrometry identi-
bind with pRb, allowing the release of E2F1. E2F1 binds to cell
cycle regulator DP1, causing increasing cell cycle from G1 to S, fying p190RhoGTPase-activating protein (p190) as
decreased apoptosis and increased genomic instability a binding partner of HPV16 E7 [157]. Protein

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 33

p190, one of the GTPase-activating protein families HUMAN PAPILLOMAVIRUS E5 PROTEIN


stimulated the intrinsic GTPase activity of the Rho E5, a small hydrophobic protein, is also an onco-
proteins, leading to Rho inactivation and increased gene of HPVs but much less studied compared
cell migration. with E6 and E7. E5 is not integrated into host ge-
nomes but is expressed by episomes. In an animal
MicroRNAs altered by E7 model, E5 alone can also cause cancer. In addition,
E7 decreases miR-203 during keratinocyte differen- E5 can increase the carcinogenic effects of E6 and
tiation, which is a tumor suppressor and thus in- E7 [175]. Some signaling pathways activated by
creases carcinogenesis [158]. Recently, several E5 have been revealed (Figure 6). The activation
other targets of miR-203 have been identified. of signaling pathways by E5 is complementary or
MicroR-203 has been shown to target anti- overlapping with E6-mediated and E7-mediated
apoptotic protein bcl-w to increase cell survival pathways.
[159], target survivin to cause G1 cell cycle arrest
[160] and target LIM and SH3 protein 1 to inhibit Epidermal growth factor receptor
migration and invasion [161]. MicroR-203 can also Epidermal growth factor receptor is a tyrosine
target p63, which maintains the balance of epithe- kinase, which consists of an extracellular ligand-
lial cell proliferation and differentiation. binding domain, a transmembrane region and a
E7 decreases miRNA-205 via pRb [162]. transduction module consisting of a tyrosine
MicroRNA-205 was considered to be a tumor sup- motif and several autophosphorylation sites
pressor [163] but has recently been demonstrated to [176,177]. EGFR is activated by multiple ligands
increase cell proliferation via E2F1 [164]. Lower such as epidermal growth factor (EGF), epiregulin
miRNA205 has been associated with poor prognosis and TGFα. EGF/EGFR is a well-known survival
of HPV-associated head and neck cancer [165]. Xie pathway in many cancers. It can increase cell pro-
et al. showed that miR-205 expression was frequently liferation and decrease cell apoptosis via its
higher in human cervical cancer [166]. In vitro exper- downstream target proteins including Akt, MAPK
iments demonstrated that miR-205 promoted cell and COX-2 [178]. EGFR and Src have feed-
proliferation and migration in human cervical cancer forwarded regulation to activate the activity of
cells. Two miR-205 targets, cysteine rich 61 (CYR61) each other through phosphorylation [178]. E5
and connective tissue growth factor (CTGF), were has been shown to increase sensitivity of EGFR
identified. Consistently, both CYR61 and CTGF were to the stimulation of EGF [179–181]. Changes of
downregulated in cervical cancer tissues [166]. Both downstream effectors have also been investigated.
CYR61 and CTGF belong to CYR61/CTGF/ E5 has been shown to increase VEGF expression
nephroblastoma family of growth regulators and via activation of Akt and MAPK [182]. Another
have both carcinogenic or tumor suppressor effect- study has shown that it also activates MAPK in-
dependent tissue types. Overexpression of CYR61 dependent of EGFR [183]. Activation of Akt can
in non-small cell lung cancer cells resulted in also block Bax-caused apoptosis [182].
decreased cell proliferation and colony formation
[167]. CTGF reduced cell migration in oral cancer Fas/FasL
[168]. However, the effect of CYR61 and CTGF in E5 has been demonstrated to inhibit the extrinsic
cervical cancer still needs to be established. pathway [184]. The pathway is also called cell
MicroR-15a, miR-15b and miR-15b have been death pathway in which specific death receptors
shown to be upregulated by E7 through E2F1 (Drs) are activated by their corresponding ligands,
and E2F3 [169,170]. The expression of these for examples tumor necrosis factor (TNF) receptor
miRNAs is increased in cervical cancer samples 1 or 2 (TNF R1 and TNF R2) by TNF-alpha, Fas re-
[170,171]. These miRs have tumor suppressor ac- ceptor by Fas ligand, DR1, DR2 by TNF-related
tivity [172,173], decreasing cyclin E1 and leading apoptosis-inducing ligand and TNF-like weak in-
to cell cycle arrest [174]. Their upregulation may ducer of apoptosis by Fas-associated death
inhibit E2F1-promoted cell cycle. MicroR-15 targets domain-like interleukin-1 beta-converting enzyme
E2F1. Therefore, the regulation of miRs by E7 [185]. The extrinsic pathway is initiated by FasL,
could play complicated effects in HPV-caused which binds to Fas receptor, leading to activation
carcinogenesis. of DRs, causing caspase 8 activation and apoptosis.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
34 J. Chen

Figure 6. E5-induced pathways. E5 can sensitize epidermal growth factor receptor (EGFR) to epidermal growth factor stimulation, leading
to increased activation of EGFR downstream pathways phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated
protein kinase (MAPK)/extracellular signal-regulated kinases (ERK). Akt can upregulate vascular endothelial growth factor (VEGF) to in-
crease angiogenesis and block Bax to decrease apoptosis. E5 can also block the Bap31-induced Fas/FasL apoptotic pathway

E5 can inhibit this pathway via various mechanisms. facilitate accumulation of mutated genes necessary
In HaCaT cells, E5 expression reduced FasL- for carcinogenesis. Cell proliferation is caused by
stimulated Fas by twofold and subsequently inhibited cell growth and division controlled by cell cycle.
caspase 8 activation [186,187]. TNF-related apoptosis- Cell cycle proceeds in four phases as indicated in
inducing ligand-induced cell death was also inhibited Figure 7 including G1, S, G2 and M phases [192].
by E5. The mechanism could be the binding of E5 to Each phase of a cell cycle is regulated by a family
Bap31 protein, which is supported as follows: (1) E5 of serine/threonine protein kinases, which are het-
and Bap31 can be coimmunoprecipitated; (2) they erodimers consisting of a CDK (catalytic) and a cy-
are colocalized in the endoplasmic reticulum; and (3) clin (regulatory) [192]. E6 and E7 can affect these
deletion of C terminus of E5 results in the loss of the cyclins via various pathways to promote cell prolif-
interaction [188]. When Bap31 is cleaved into eration. Among them, cyclin D is most studied. E6
p20Bap31, it promotes cell apoptosis through the DR can activate the Akt/myc pathway and decrease
pathway, and binding of E5 could prevent such p16 to increase cyclin D [53]. Increased cyclin B1
cleavage [189]. A recent report demonstrated that E5 by E6 has also been reported, which promotes G2
could activate A4, a small transmembrane lipopro- phase [193–195]. HPV 16 E7 can increase cyclin A
tein, which in turn increased cell proliferation. A4 is by direct interaction [196,197]. Silencing of E6/E7
known to interact with Bap31 [190]. by siRNA reduced cervical cancer cell proliferation
in both in vitro and in vivo [198,199].
THE CONSEQUENCES OF SIGNALING Debnath et al. studied co-operative effects of acti-
ALTERATIONS IN HUMAN vation of Akt and expression of E7 or cyclin D1 in
PAPILLOMAVIRUS-INFECTED CELLS cultured cancer cells [200]. It was found that Akt
activation alone or E7 expression alone was insuffi-
Cell proliferation cient to maintain cell proliferation without growth
Cancer is defined as abnormal cell growth charac- factors. However, the combination of activation of
terized by increased cell proliferation and de- Akt and expression of E7 markedly increased cell
creased apoptosis. Abnormally increased cell proliferation as indicated by cell proliferation
proliferation is one of the main characteristics of marker Ki-67Akt downstream target mTOR that
cancer [191]. Although not all hyperproliferations has been shown to play a key role as rapamycin-
cause cancer, increased cell proliferation could inhibited cell growth in E6/E7 transduced

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 35

Figure 7. Human papillomaviruses, cell cycle and cell proliferation. A cell undergoes a cycle for proliferation with four phases, G1, S, G2
and M. Cell cycle is sophisticatedly regulated by cyclins and cyclin-dependent kinases (CDKs). E7 can block Rb/E2F to increase cyclin-E
activity and thus accelerate cell cycle. E7 can also promote cyclin-A activity. E6 increases the activities of cyclins D and B

epithelial cells [201]. PDZ protein MAGI-1 has been and pRb was sufficient to cause oral keratocyte im-
shown to mediate the E6-induced cancer cell prolif- mortalization [206]. Activation of PI3K/Akt by
eration [39], and expression of MAGI-1 through E5/E6/E7 can increase cell survival by reducing
disrupting its interaction with E6 resulted in de- the intrinsic apoptotic pathway [207]. PDZ protein
creased proliferation. MAGI-1 has been shown to mediate the E6-
In patients with HPV-associated cancers, cell pro- decreased cancer cell apoptosis [39]. Overexpres-
liferation in tumor tissues is increased. It has been sion of MAGI-1 through disruption of its interac-
shown that cell cycle proteins p16, Ki-67, cyclin tion with E6 resulted in increased apoptosis. E5
D1, p53 and ProEx C are altered in 144 cervical tis- can decrease both extrinsic and intrinsic pathways
sue samples, showing increased proliferation [202]. (Figure 6) [182,184]. Therefore, HPVs use multiple
Molecules related to cell proliferation have a signif- oncoproteins, which alter multiple signaling path-
icant increase from low-grade to high-grade lesions ways to decrease host cell apoptosis.
and cancer in HPV-associated cancers [203].
Genomic instability
Apoptosis Genomic instability plays a key role in carcinogen-
Another characteristic of cancer cells is decreased esis. It promotes gene mutations, which in turn af-
cell apoptosis [191]. There are two pathways in- fect cell proliferation and apoptosis. HPVE5, E6
volved in cell apoptosis: extrinsic, also called the and E7 could increase genomic instability via sev-
death receptor pathway, and intrinsic, also called eral mechanisms. Akt activation can cause genomic
the mitochondrial pathway. E5/E6/E7 can de- instability [208]. Alteration of the pRb/E2F1 path-
crease apoptosis through both apoptotic pathways way by E7 also leads to genomic instability [209]. In-
via altering multiple signaling pathways. Targeting activation of p53 decreases DNA damage-induced
HPV E6 with peptide aptamers caused cancer cell cell death and increases tolerance to genomic insta-
apoptosis [204]. Qi et al. showed that simultaneous bility. HPV-caused genomic instability is initiated
silencing of HPV18 E6 and E7 induced apoptosis by abnormal amplification of centrosomes [5]. Cen-
in HeLa cells, indicating an important role for trosomes are microtubule-organizing centers,
these oncoproteins in maintaining cancer cell sur- consisting of a pair of centrioles and pericentriolar
vival [205]. proteins [210]. Centrosomes are important regula-
E6 inhibition of p53 can greatly reduce intrinsic tors of cell division to ensure a cell is divided into
apoptosis to reduce DNA damage-initiated apopto- two [210]. In normal conditions, centrosome dupli-
sis. Smeets et al. showed that inactivation of p53 cates once prior to mitosis to form two spindle

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
36 J. Chen

poles. The duplication begins in late M phase/early resistance to chemotherapeutic drug 5-fluorouracil
G1 phase. HPV E7 causes rapid induction of centro- (5-FU) and oxaliplatin [229,230]. In cervical cancer,
somes with more than two copies, leading to abnor- activation of PI3K/Akt causes resistance to radio-
mal cell division and genomic instability [211,212]. therapy, and inhibition of the pathway increased
The pathway is mediated by CDK2, which is acti- efficacy of radiation [233]. A recent study
vated by Akt [213,214]. Increased CDK2 activity in demonstrated that activation of the PI3K/Akt
turn promotes polo-like kinase 4, a rate-limiting en- pathway by E6 caused drug resistance to cisplatin
zyme in centriole replication [215]. It is known that in HPV-associated lung cancer mediated by the
deletion of polo-like kinase 4 causes defects in cen- downstream target of the PI3K/Akt pathway
triole replication, while overexpression results in inhibitors of anti-apoptotic protein [234]. Overex-
multiple centrioles [215]. pression of hTERT increased 5-FU effect on HeLa
Genomic instability can cause accumulation of cells to cause apoptosis [235].
gene mutations. In cervical cancer, many gene mu- Targeting these signaling pathways has been an
tations have been reported with several mutations effective strategy to overcome drug resistance in
involved in the PI3K/Akt pathway including cancer treatment [236,237]. Many approaches, par-
PIK3CA, PTEN and EGFR and Ras [216–220]. These ticularly, small molecule inhibitors, have been de-
mutations can further activate the PI3K/Akt path- veloped for the treatment. The combination
way in cervical cancer. A report showed that therapy of targeted therapy with other treatment
PIK3CA mutated at 31.3%, Kirsten rat sarcoma viral regime is detailed in the late section—therapeutic
oncogene homologue at 8.8% and EGFR at 3.8% of implications.
cervical cancers [218]. The list of gene mutations in
HPV-associated cervical cancer has been expanded Cell mobility and metastasis
in a recent study including recurrent E322K substi- Cell migration plays a key role in cancer metas-
tutions in the MAPK1 gene (8%), inactivating muta- tasis. To be able to migrate to a new place, a can-
tions in the HLA-B gene (9%) and mutations in cer cell must detach from the original site,
EP300 (16%), FBXW7 (15%), NFE2L2 (4%), TP53 migrate to the new place, attach and grow in a
(5%) and ERBB2 (6%) [221]. This study also con- new environment. Signaling pathways play key
firmed previously reported mutations such as roles in the process of cancer metastasis [238].
PIK3CA (14%), PTEN (6%) and STK11 (5%). There- They can control cell morphological change, cell
fore, HPV oncogenes initiate signaling pathway al- polarity and cell mobility to meet the require-
terations, which cause genomic instability, leading ments of migration. Many signaling changes
to accumulation of gene mutations, which accelerate caused by HPV E6 and E7 are associated with
changes of signaling pathways necessary for cell morphology and migration such as the
carcinogenesis. PI3K/Akt and RhoA pathways [152,157].
MicroRNAs have also been associated with cell
Drug resistance morphology control. Therefore, HPV E6 and E7
Drug resistance to anticancer therapeutic agents is can also facilitate cancer metastasis. Loss of
a major reason responsible for treatment failure PTEN has been associated with increased metas-
and high rate of cancer-related deaths [222–224]. tasis in patients [239]. Activation of MAPK in-
Patients may not respond to initial therapy (inher- creased metastasis of cervical cancer [240].
ent drug resistance) or develop drug resistance sub- Antiviral treatment has been shown to have
sequently after effective initiation (acquired anti-metastatic effect in HPV-positive cells [241].
resistance) [222]. Multiple mechanisms for drug re-
sistance have been elucidated including drug target Cancer stem cells
mutations, increased drug detoxification system to It has been realized that cancer cells in a tumor or in
reduce intracellular drug concentrations, increased a cell line are heterogeneous, which are composed
resistance to apoptosis and abnormal activation of of many different phenotypic cancer cells
signaling pathways [225–232]. [242,243]. One small population of cancer cells is
PI3K/Akt is well known to cause drug resistance called cancer stem cells (CSCs). These cells have
in cancer. Activation of the pathway by insulin, the property of renewal and can differentiate to
IGF-1 or mutations has been associated with drug other types of cancer cells. CSCs are considered to

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 37

be only cells within a tumor that can proliferate ex- THERAPEUTIC IMPLICATIONS
tensively and drive tumorigenic growth [242,243]. HPV-associated cancers that cannot be removed by
The other types of cancer cells are terminal differen- surgery can be treated by multiple approaches such
tiated cells, which are not renewed. CSCs have also as chemotherapeutic agents, antiviral therapy, im-
characteristics of normal stem cells, that is slow cy- munotherapy, radiotherapy, phytochemicals and
cling, altered DNA repair machinery and high ex- targeted therapy (Figure 8).
pression levels of anti-apoptotic genes and ATP-
binding cassette (ABC) transporters [244]. These Chemotherapy
characteristics render CSCs drug resistant, for ex- A number of chemotherapeutic agents have been
ample CSCs are less sensitive to cisplatin than used in cervical cancer. Cisplatin has been shown
non-CSCs in HNSCC [245]. to be most effective among all drugs tested includ-
Cancer stem cells have been identified in acute ing cisplatin, carboplatin, 5-FU, cyclophosphamide,
myeloid leukemia and later found in other cancer chlorambucil, melphalan, methotrexate, vincristine,
types as well, for example breast cancer, brain can- bleomtcin, adriamtcin, mitomycin C, ifosfamimde,
cer, colon cancer, neck and head cancer, pancreas pclitaxel, irinotecan, gemcitabine, vinorelbine, do-
cancer, liver cancer and melanoma. In cervical can- cetaxel, doxorubicin and mitolactol. However, cis-
cer, CSCs have been isolated by various biomarkers platin only has 10–20% of responses [249]. An
including cell surface markers, Hoechst staining increase of dosage of cisplatin resulted in severe re-
and formation of spheres. Qi et al. isolated sphere nal toxicity. Carboplatin is a derivative of cisplatin
cells from cervical cancer cell line HeLa [246]. These but with reduced renal toxicity. Overall, single che-
stem cells account for only about 1% of total popu- motherapeutic agents can only produce low effi-
lation. They are small, round cells with increased cacy, which can only extend patients’ lives to
biomarkers Oct3/4, CD133 and breast cancer resis- 12 months and thus is regarded as a palliative ther-
tance protein. However, aldehyde dehydrogenase apy [250]. As single agents are not efficient, various
is not changed. Stem cells isolated have been shown combination therapies have been applied. Chemo-
to have increased drug resistance to chemothera- therapeutic agents have been used for double or
peutic agent Trichostatin and radiation treatment. triplet combination therapy to increase therapeutic
CSCs have also been isolated from two other cervi- effectiveness. For example, cisplatin has been used
cal cell lines SiHa and CaLo by their increased abil- together with 5-FU, and the combination therapy
ity to efflux Hoechst 33342 [247]. These cells had showed various responses from 22% to 58% [251–
increased ABCG2 and ABCB1. 253]. Cisplatin and capecitabine together produced
Not many studies have been performed for the responses of 30–50% [254,255]. However, the me-
role of HPV in CSCs. It has been compared with dian survival was not improved.
HPV-positive HNSCC cancers with HPV-negative The use of chemotherapeutic agents usually
ones for the amount of CSCs contained. Zhang results in drug resistance caused by activation of
et al. found that HPV16-positive HNSCC had a signaling pathways. Combination of chemothera-
greater intrinsic CSC pool than HPV-negative peutic agents and targeted therapy could be more
HNSCC [248], while Tang et al. showed that the effective, as activation of signaling pathways is
proportion of CSC was not significantly different one of the major reasons for drug resistance of che-
in HPV-positive or HPV-negative HNSCC cell lines motherapeutic agents. For example, 5-FU can stim-
[245]. These experiments may not be able to pro- ulate the Src and PI3K/Akt pathways, resulting in
vide valuable evidence for the role of HPV in CSCs drug resistance and inhibition of Src that increased
because other cancers can be caused by risk fac- the efficacy of 5-FU markedly [256].
tors, which also affect CSC abundance. Transduc-
tion of HPV negative cancer cells with HPV Antiviral and anti-oncogene therapies
E6/E7 can reflect the role of E6/E7 in CSCs. It Because expression of HPV oncogenes through epi-
has been shown to increase colony formation that somes or integrated E6 and E7 genes is important
was observed in both CSCs and non-CSCs. How- for the maintenance of cancer cells, elimination or
ever, its effect on stemness is not well studied. This inhibition of HPVs, HPV oncogenes has been
is an important area, where detailed studies are studied for the treatment of HPV-associated can-
warranted. cers. Several antiviral agents have been tested.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
38 J. Chen

Figure 8. Therapy strategy against human papillomavirus (HPV)-associated cancers. HPV oncogenes E5, E6 and E7 activates cellular signaling
pathways to promote cancer cell survival. Treatment strategy can target the process in different levels including decreasing HPVs and their
oncogene expression by short hairpin RNAs (shRNAs) or small molecule inhibitors, targeting cellular signaling pathways by inhibitors or
phytochemicals and killing cancer cells by chemotherapy or radiotherapy or immunotherapy

Cidofovir has been shown to restore p53 and in- these pathways such as reactivation of p53, inhibi-
crease radiosensitivity in HPV-associated cancers tion of the PI3K/Akt pathway, inhibition of the
[257]. RNA interference has been used to inhibit ex- Notch pathway and supplementation of decreased
pression of E6 and E7 [258]. Transduction of miRNAs. Among them, targeted therapies against
lentiviral vectors delivered shRNA against E6/E7 the EGFR, VEGF, Src, PI3K/Akt and Erk path-
into HeLa cells caused apoptosis [259–262]. Combi- ways have been the best studied to date (Figure 9).
nation of shRNA against both E6 and VEGF was
more effective for the treatment of cervical cancer Anti-phosphoinositide 3-kinase/protein
than single shRNA or combination therapy with kinase B
chemotherapeutic drugs [262,263]. It has been Activation of the PI3K/Akt pathway has been as-
shown that siRNA against E6 increased the sensi- sociated with increased cancer cell proliferation,
tivity of SiHa cells to cisplatin [264]. In a xenograft decreased apoptosis, increased cell migration and
model of cervical cancer, injection of shRNA decreased drug sensitivity in many cancers in-
against E6/E7 reduced tumor growth [265]. A cluding cervical cancer [41,45,229,267,268].
range of small molecule inhibitors have been devel- Targeted therapy against PI3K/Akt has been used
oped to inhibit E6 and E7 or their binding to part- in many cancers [236,269]. Noh et al. showed that
ner proteins [266]. activation of Akt by HPV 16 E7 was responsible
for cancer cells to escape immune responses
Targeted therapy [270]. Therefore, targeting PI3K/Akt may be an
As E6/E7 affects cancer via signaling pathways, effective approach for the treatment of HPV-
modulation of these signaling pathways has thera- associated cancer.
peutic implications. As multiple signaling path- Several studies have tested the effect of
ways have been altered by E6 and E7, it is PI3K/Akt pathway inhibitors on cervical cell lines.
possible to use multiple small chemicals to reverse Rashmi et al. used two allosteric Akt inhibitors

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 39

(SC-66 and MK-2206) in combination with the glu- temsirolimus has been used in a phase-II clinical
cose analog 2-deoxyglucose in C33A cells, which trial. It increased cervical cancer patient survival
have activating PIK3CA (E545K, E542K) and time, that is 3% partial response, and 57.6% had
inactivating PTEN (R233*) mutations [271]. Re- stable disease [274]. Coppock et al. established a
sults showed that inhibition of the PI3K/Akt by mouse cancer cell line by transducing both E6/E7
these inhibitors decreased cell viability through a oncogenes and mutated H-Rasv12 gene into mouse
non-apoptotic mechanism. 2-Deoxyglucose further oropharyngeal epithelial cells for the test of the ef-
increased cell viability. SC-66 also inhibited cell fect of rapamycin [201]. The cells can grow into tu-
migration. Xia et al. tested the effect of mor in a xenograft model. Rapamycin has been
PI3K/Akt/COX-2 pathway inhibitors Ly294002 used to treat the mice in combination with cis-
and celecoxib on the sensitivity of radiotherapy platin, decreasing cancer cell proliferation and
in HeLa cells and found that the combination of prolonging long-term survival rate of mice with
Ly294002, celecoxib and radiation produced treat- the xenograft tumors.
ment effects [233].
The combination of inhibition of PI3K/Akt and Anti-vascular endothelial growth factor
radiotherapy has also been tested in an animal Human papillomavirus oncogenes cause upregula-
model of cervical cancer [272,273]. In BALB/C tion of VEGF, which can increase cancer cell sur-
nude mice with xenografted HeLa cells, LY294002 vival and angiogenesis. In cervical cancer, VEGF
decreased cell survival and xenograft tumor is overexpressed. Therefore, anti-VEGF has been
growth. However, the combination of LY294002 used in clinical trial for the treatment of cervical
and radiation resulted in synergistic reduction of cancer. Bevacizumab, a monoantibody against
tumor growth [273]. VEGF, is a most-studied agent for anti-VEGF ther-
apy. A recent study showed that addition of
Anti-mammalian target of rapamycin bevazumab to chemotherapeutic cisplatin–
Inhibitors of PI3K/Akt downstream signaling mol- paclitaxel or topotecan–paclitaxel regime increased
ecule mTOR have also been developed. These in- treatment efficacy, increasing response rate from
hibitors have been tested in cervical cancer cell 36% to 48% and overall survival time from 13.3
lines and animal models. Among them, months to 17.0 months [275]. It can also increase

Figure 9. Targeted therapy against key signaling pathways in human papillomavirus-associated cancers. Many small molecule inhibitors
have been developed to target key signaling molecules in human papillomavirus-associated cancers including epidermal growth factor
receptor (EGFR), extracellular signal-regulated kinases (ERK), vascular endothelial growth factor (VEGF), Src and mammalian target of
rapamycin (mTOR). Dual inhibitors against both phosphoinositide 3-kinase (PI3K) and mTOR have also been developed. Akt, protein ki-
nase B; GSK, glycogen synthase kinase

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
40 J. Chen

treatment efficacy of radiation and cisplatin alone D1. EGCG has been demonstrated to increase the
[276]. Many small molecule inhibitors of VEGF sensitivity to cisplatin [285].
have also been developed and tested in clinical tri- Curcumin has also been shown to suppress HPV
als with benefit but limited improvement of sur- oncogene expression and thus increase p53 and
vival time. pRb [290]. Singh et al. showed that curcumin de-
creased estrodiol-induced cervical cancer cell pro-
Anti-epidermal growth factor receptor liferation and caused apoptosis [291] Curcumin
Epidermal growth factor receptor gene amplifica- can decrease the levels of E7, proliferating cell nu-
tion has been associated with poorer outcome of clear antigen and cyclin D1 elevated by estrogen
cervical cancer, and thus, anti-EGFR has been em- but not E6, telomerase and p16. Curcumin over-
phasized in the treatment of cervical cancer [277]. came drug resistance to cisplatin in SiHa cells by
Both antibody against EGFR (cetuximab) and small inhibiting metalloprotease 1, P-glycoprotein, cyclin
molecule inhibitors of EGFR (gefitinib and erloti- D1 and upregulation of p53, peripheral benzodiaz-
nib) have been tested. Cetuximab produced syner- epine receptor, p21 and p27 [292]. Nanoparticle-
gistic effects with antiviral agent cidofovir in packed curcumin increased the sensitivity of cervi-
HeLa and Me180 cells in both in vitro cell cultures cal cancer to chemotherapeutic agent paclitaxel
and in vivo xenograft model [278]. Cetuximab has through inhibition of the Akt pathway [293]. In a
also been tested in a clinical trial with platinum xenograft model using CaSki cells, curcumin re-
and 5-FU in 121 head and neck cancer patients duced tumor size by decreasing VEGF, COX-2
and extended overall survival time to 11 months and EGFR [294]. A cream containing curcumin
[279]. Another phase-II clinical trial with combina- eliminated HPV+ cells in mice [295].
tion of cetuximab and cisplatin showed that the re- Quercetin has also caused HeLa apoptosis and
gime was well tolerated but cetuximab had no cell cycle arrest in G2/M phase [296,297]. It was
further benefit than cisplatin alone [280]. Gefitinib shown to increase expression of p53 and p21 and
also resulted in stable disease in 20% of patients decrease cyclin D1. The mitochondrial apoptotic
of recurrent cervical cancer in a clinical trial [281]. pathway was activated with upregulation of pro-
Clinical trials of erlotinib showed safety [282] but apoptotic bcl-2 family members and cytochrome
limited benefit even in combination with topotecan c and Apaf-1 and caspases and downregulation
[283] and cisplatin [284]. of bcl-2 and survivin. Quercetin also inhibited
NF-κB [296].
Applications of phytochemicals
Phytochemicals could be used for the treatment of Immunotherapy
cancer because of their ability to inhibit multiple An effective immune system is necessary for the
signaling pathways. Some phytochemicals have eradication of cancer cells, which have been weak-
been well studied in cervical cancer such as epi- ened by chemotherapy or targeted therapy. Al-
gallocatechin gallate (EGCG), resveratrol and though infection of HPV can elicit immune
curcumin and quercetin. EGCG is a major green responses against virus and can clear virus in most
tea component, which decreases cell proliferation cases, HPV oncogenes E5, E6 and E7 can inhibit im-
and increases drug sensitivity in cervical cancer mune responses to escape the clearance. E5 reduces
[285,286]. The mechanisms are inhibition of several HLA-1 expression by interacting with HLA-1
key molecules of signaling pathways. In Caski, heavy chain [298]. E7 can reduce STAT-1 and de-
EGCG induced G1 arrest and apoptosis [287,288]. crease transporter associated with antigen
Treatment of HeLa cells with EGCG induced processing/interferon regulatory factor and thus
dose-dependent and time-dependent inhibitions reduce HLA class 1 [299]. Therefore, HPV antigen
of proliferation and increased apoptosis [289]. presentation is reduced, and immune responses
EGCG activated the mitochondrial apoptotic path- against HPV-positive cells are decreased.
way indicated by increases of reactive oxygen spe- Various immunotherapies have been tried in cer-
cies, Bax/Bcl-2 ratio, cytochrome-c release, cleavage vical cancer and vulval intraepithelial neoplasia
of procaspase-3 and -9 and poly(ADP-ribose)-poly- (VIN) to strengthen the immune responses to elim-
merase. EGCG also inhibited the activity of Akt inate virus or against cancer cells. Imiquimod, a
and NF-κB and reduced expression levels of cyclin toll-like receptor agonist, has been tested to activate

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 41

innate immune cells such as dendritic cells, mono- increased activator protein 1, NF-κB and MAPK
cytes and macrophages in HPV-associated cancers. [316]. More detailed studies are needed to elucidate
Imiquimod increased HPV clearance and reduced the role of estrogen in the carcinogenesis of cervical
tumor size in a clinical trial with 59 cervical cancer cancer. Nevertheless, ERα has been demonstrated
patients [300]. The mechanism was shown to be the to play an important role in cervical cancer in ani-
normalization of immune cell counts [301] and in- mal experiment, and selective estrogen receptor
creased CD8+ cell recognition to E7 immunization modulators raloxifene can decrease tumor growth
[302]. Topical application of 5% imiquimod cream [317,318].
in 62 VIN patients resulted in 47 complete re- In addition to inhibiting ERα, activation of ERβ
sponses and 19 partial responses [303]. The effect has also been tested for the treatment of cervical
of imiquimod was time-dependent, and patients cancer. Genistein, a phytoestrogen, which
had complete regression rate 32% (6 out of 19) at stimulates ERβ, induced apoptosis of HeLa cells
week 10, 58% (11 out of 19) at week 20 and 63% [319] and sensitized chemotherapeutic agents cis-
(12 out of 19) at week 52 [304]. E6 and E7 proteins platin and camptothecins [320,321]. Geistein was
have been used to elicit specific immune response shown to inhibit the PI3K/Akt, NF-κB, MAPK
to HPV-positive cells. The test in 20 patients with and MMP-9 pathways and activate apoptotic path-
VIN showed partial efficacy with increased CD4 ways [321–324].
and CD8 T-cell responses [305]. E7 has been carried Antiestrogen phytochemical indole-3-carbinol
by a naked DNA vector for its expression in cells to was shown to prevent incidence of cervical cancer
increase MHC processing, resulting in increased in a transgenic model [325]; 19 out of 25 mice devel-
immunogenicity of E7 and overcoming immune oped cervical cancer in control group, while only 2
tolerance [306]. Imiquimod increased the effect of out of 24 had cancer in indole-3-carbinol supple-
this DNA vaccine [307]. Cytokines have also been mented group. In cervical cancer cell lines, indole-
used to activate immune system in treating cervical 3-carbinol was shown to cause apoptosis through
cancer. The most commonly used cytokines are IL-2, the intrinsic pathway indicated by the reduction
IL-12, granulocyte-macrophage-CSF and IFN-alpha. of Bcl-2 protein [326]. Indole-3-carbinol also
Systemic application of cytokines is highly toxic, but prevented PTEN loss [327] and decreased cyclin-E
local application is well tolerated [308]. levels [328].

Antiestrogen therapy Targeting cancer stem cells


Estrogen plays a key role in several cancers such as It has been recognized that CSCs play key roles
endometrial, prostate, colon and breast cancers. Ep- in cancer treatments. Conventional therapy can
idemiological evidence has shown that estrogen is result in decreased differentiated cancer cells
also important in cervical cancer [309]. In an animal while CSCs are resistant to the therapy. The ther-
model, estrogen has been shown to have apy could eliminate differentiated cancer cells,
cooperating effect with HPV 16 oncogenes E6 and leading to initial shrinkage of tumor. However,
E7 [310–312]. There are two types of estrogen re- remained CSCs will continue to produce differen-
ceptors—estrogen receptor-alpha (ERα) and estro- tiated cancer cells, causing relapse of cancer.
gen receptor-beta (ERβ). The activation of these Therefore, elimination of CSCs could lead to cur-
receptors has opposite effects in some cancers. In able cancer.
colon cancer, ERα increases carcinogenesis via acti- Signaling pathways are important in maintaining
vation of signaling pathways Akt and MAPK, CSCs, such as the PI3K/Akt and MAPK pathways.
while ER-beta inhibits these pathways. Both ERα Akt activation has been associated with stemness
and ERβ are expressed in cervical cancer [313]. in various cancers such as pancreatic cancer, ovarian
ERα is decreased, while ERβ is maintained in cervi- cancer, breast cancer and glioma [329–332]. As stem
cal cancer [313,314]. Interestingly, ERα expression cells are drug resistant, increased stemness may
in stromal cells is increased [314], and ERα in these account for increased drug resistance. Inhibition of
cells is necessary for the development of cervical PI3K/Akt pathway caused a decreased sphere
cancer [315]. The signaling pathways altered by es- formation in cancer cell populations in breast cancer,
trogen in cervical cancer are not well studied. A re- glioma and prostate cancer [333–335]. Studies have
cent study using microassay showed that it also shown that IGF-1 is associated with colon

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
42 J. Chen

CSCs [336]. Activation of the PI3K/Akt pathway Simultaneous inhibition of two or more key sig-
decreases the sensitivity of CSCs to chemothera- naling molecules may produce much better ef-
peutic drugs [335,337]. The downstream of the Akt fects. At present, the inhibition of multiple
pathway such as GSK/β-catenin has been demon- molecules in HPV-associated cancers has not been
strated to be of importance in CSCs [330]. Thus, optimized.
targeting signaling pathways may eradicate CSCs.
This has been demonstrated by a recent break- CONCLUSIONS
through by inhibiting signaling molecule BMI-1 to HPV oncogenes E5, E6 and E7 play key roles in
reduce colon CSCs’ renewal ability [338]. The ap- HPV-associated cancer by activating multiple can-
proach may be applicable in HPV-associated cancer. cer survival signaling pathways and inhibiting can-
A recent study showed that CD200, a membrane cer suppressor proteins. Altered signaling
protein, was overexpressed in HPV-positive pathways in turn promote cell proliferation, de-
HNSCC cell lines and the overexpression of CD200 crease cell apoptosis and increase cell migration
was associated with increased BMI-1 [339]. In vivo, and drug resistance. The effects of E5, E6 and E7
CD200 increased resistance to chemoradiation, indi- could be synergistic. Targeting E5-mediated, E6-
cating the importance of BMI-1 in HPV-associated mediated and E7-mediated signaling pathways is
cancers, and inhibition of BMI-1 may have similar of therapeutic value. Among them, the PI3K/Akt
effect as in colon cancer. pathway activated by E5, E6 and E7 as well as mu-
In summary, various approaches have been tations of genes that encode elements of the path-
used for the treatment of HPV-associated cancers, way could be inhibited, which may provide an
especially cervical cancer. However, the single effective approach for the treatment of HPV-
treatment approach has very limited effect. Com- associated cancer. Although anti-EGFR and anti-
bination therapy may be promising, particularly VEGF have been used in clinical trials in cervical
the combination of targeted therapy with other cancer, outcomes are not satisfactory. As multiple
approaches. Single inhibition of a signaling mole- signaling pathways are activated by HPV onco-
cule in cervical cancer could be not effective. This genes, it may be necessary to inhibit multiple sig-
may be due to the activation of multiple signaling naling molecules for the treatment of HPV-
pathways by HPV oncogenes E5, E6 and E7. associated cancers.

REFERENCES
1. Bouvard V, Baan R, Straif K, et al. A re- transformation. Nature Reviews Cancer carcinoma than the prototype. Cancer Re-
view of human carcinogens—Part B: bio- 2010; 10: 550–560. search 1998; 58: 829–833.
logical agents. The Lancet Oncology 2009; 6. Zhao KN, Chen J. Codon usage roles in 10. DeFilippis RA, Goodwin EC, Wu L,
10: 321–322. human papillomavirus. Reviews in Medical DiMaio D. Endogenous human papillo-
2. Nobre RJ, Herráez-Hernández E, Fei J-W, Virology 2011; 21: 397–411. mavirus E6 and E7 proteins differentially
et al. E7 oncoprotein of novel human pap- 7. Bible JM, Mant C, Best JM, et al. Cervical regulate proliferation, senescence, and
illomavirus type 108 lacking the E6 gene lesions are associated with human papillo- apoptosis in HeLa cervical carcinoma
induces dysplasia in organotypic mavirus type 16 intratypic variants that cells. Journal of Virology 2003; 77:
keratinocyte cultures. Journal of Virology have high transcriptional activity and in- 1551–1563.
2009; 83: 2907–2916. creased usage of common mammalian co- 11. Narisawa-Saito M, Inagawa Y,
3. Stubenrauch F, Straub E, Fertey J, Iftner T. dons. Journal of General Virology 2000; 81: Yoshimatsu Y, et al. A critical role of
The E8 repression domain can replace the 1517–1527. MYC for transformation of human cells
E2 transactivation domain for growth inhi- 8. Londesborough P, Ho L, Terry G, Cuzick by HPV16 E6E7 and oncogenic HRAS.
bition of HeLa cells by papillomavirus E2 J, Wheeler C, Singer A. Human papillo- Carcinogenesis 2012; 33: 910–917.
proteins. International Journal of Cancer mavirus genotype as a predictor of persis- 12. Ocadiz-Delgado R, Castañeda-Saucedo E,
2007; 121: 2284–2292. tence and development of high-grade Indra AK, et al. RXRα deletion and
4. Münger K, Baldwin A, Edwards KM, et al. lesions in women with minor cervical ab- E6E7 oncogene expression are sufficient
Mechanisms of human papillomavirus- normalities. International Journal of Cancer to induce cervical malignant
induced oncogenesis. Journal of Virology 1996; 69: 364–368. lesions in vivo. Cancer Letters 2012; 317:
2004; 78: 11451–11460. 9. Zehbe I, Wilander E, Delius H, Tommasino 226–236.
5. Moody CA, Laimins LA. Human papillo- M. Human papillomavirus 16 E6 variants 13. Hoe KK, Verma CS, Lane DP. Drugging
mavirus oncoproteins: pathways to are more prevalent in invasive cervical the p53 pathway: understanding the route

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 43

to clinical efficacy. Nature Reviews Drug 24. Ben KY, Teissier S, Tan M, et al. The hu- and triggers its degradation by the pro-
Discovery 2014; 13: 217–236. man papillomavirus E6 oncogene re- teasome. Journal of Virology 2005; 79:
14. Rufini A, Tucci P, Celardo I, Melino G. Se- presses a cell adhesion pathway and 4229–4237.
nescence and aging: the critical roles of disrupts focal adhesion through degrada- 34. Facciuto F, Valdano MB, Marziali F, et al.
p53. Oncogene 2013; 32: 5129–5143. tion of TAp63β upon transformation. Human papillomavirus (HPV)-18 E6
15. Huibregtse J, Scheffner M, Howley P. PLoS Pathogens 2011; 7: e1002256. oncoprotein interferes with the epithelial
Localization of the E6-AP regions that 25. Park JS, Kim EJ, Lee JY, Sin HS, cell polarity Par3 protein. Molecular Oncol-
direct human papillomavirus E6 bind- Namkoong SE, Um SJ. Functional inacti- ogy 2014; 8: 533–543.
ing, association with p53, and vation of p73, a homolog of p53 tumor 35. Thomas M, Narayan N, Pim D, et al. Hu-
ubiquitination of associated proteins. suppressor protein, by human papilloma- man papillomaviruses, cervical cancer
Molecular and Cellular Biology 1993; 13: virus E6 proteins. International Journal of and cell polarity. Oncogene 2008; 27:
4918–4927. Cancer 2001; 91: 822–827. 7018–7030.
16. Brimer N, Lyons C, Vande Pol SB. Associ- 26. Maas A-M, Bretz AC, Mack E, Stiewe T. 36. Massimi P, Gammoh N, Thomas M, Banks
ation of E6AP (UBE3A) with human papil- Targeting p73 in cancer. Cancer Letters L. HPV E6 specifically targets different cel-
lomavirus type 11 E6 protein. Virology 2013; 332: 229–236. lular pools of its PDZ domain-containing
2007; 358: 303–310. 27. Moll UM, Slade N. p63 and p73: roles in tumour suppressor substrates for
17. Ansari T, Brimer N, Pol SBV. Peptide in- development and tumor formation 11 Na- proteasome-mediated degradation. Onco-
teractions stabilize and restructure hu- tional Cancer Institute. Molecular Cancer gene 2004; 23: 8033–8039.
man papillomavirus type 16 E6 to Research 2004; 2: 371–386. 37. Adey NB, Huang L, Ormonde PA, et al.
interact with p53. Journal of Virology 28. Ye F, Zhang M, Aregger M, et al. Struc- Threonine phosphorylation of the
2012; 86: 11386–11391. tures and target recognition modes of MMAC1/PTEN PDZ binding domain
18. Scheffner M, Huibregtse JM, Vierstra RD, PDZ domains: recurring themes and both inhibits and stimulates PDZ binding.
Howley PM. The HPV-16 E6 and E6-AP emerging pictures. Biochemical Journal Cancer Research 2000; 60: 35–37.
complex functions as an ubiquitin-protein 2013; 455: 1–14 38. Tolkacheva T, Boddapati M, Sanfiz A,
ligase in the ubiquitination of p53. Cell 29. Vanitha KS, Christian K, Miranda T, Law- Tsuchida K, Kimmelman AC, Chan AM.
1993; 75: 495–505. rence B. PDZ domains: the building blocks Regulation of PTEN binding to MAGI-2
19. Scheffner M, Werness BA, Huibregtse JM, regulating tumorigenesis. Biochemical Jour- by two putative phosphorylation sites at
Levine AJ, Howley PM. The E6 nal 2011; 439: 195–205. threonine 382 and 383. Cancer Research
oncoprotein encoded by human papillo- 30. Kiyono T, Hiraiwa A, Fujita M, Hayashi 2001; 61: 4985–4989.
mavirus types 16 and 18 promotes the Y, Akiyama T, Ishibashi M. Binding of 39. Kranjec C, Massimi P, Banks L. Restora-
degradation of p53. Cell 1990; 63: high-risk human papillomavirus E6 tion of MAGI-1 expression in HPV posi-
1129–1136. oncoproteins to the human homologue tive tumour cells induces cell growth
20. Lorenz LD, Cardona JR, Lambert PF. Inac- of the Drosophila discs large tumor sup- arrest and apoptosis. Journal of Virology
tivation of p53 rescues the maintenance of pressor protein. Proceedings of the Na- 2014; JVI: 03247–03213.
high risk HPV DNA genomes deficient in tional Academy of Sciences 1997; 94: 40. Rodon J, Dienstmann R, Serra V,
expression of E6. PLoS Pathogens 2013; 9: 11612–11616. Tabernero J. Development of PI3K inhibi-
e1003717. 31. Lee SS, Weiss RS, Javier RT. Binding tors: lessons learned from early clinical tri-
21. Kho E-Y, Wang H-K, Banerjee NS, Broker of human virus oncoproteins to als. Nature Reviews. Clinical Oncology 2013;
TR, Chow LT. HPV-18 E6 mutants reveal hDlg/SAP97, a mammalian homolog of 10: 143–153.
p53 modulation of viral DNA amplifica- the Drosophila discs large tumor sup- 41. Chen J, Zhao K, Li R, Shao R, Chen C. Ac-
tion in organotypic cultures. Proceedings pressor protein. Proceedings of the Na- tivation of PI3K/Akt/MTOR pathway
of the National Academy of Sciences 2013; tional Academy of Sciences 1997; 94: and dual inhibitors of PI3K and MTOR in
110: 7542–7549. 6670–6675. endometrial cancer. Current Medicinal
22. Massimi P, Shai A, Lambert P, Banks L. 32. Lee C, Laimins LA. Role of the PDZ Chemistry 2014; 21: 3070–3080.
HPV E6 degradation of p53 and PDZ con- domain-binding motif of the oncoprotein 42. Polivka Jr J, Janku F. Molecular targets for
taining substrates in an E6AP null back- E6 in the pathogenesis of human papillo- cancer therapy in the PI3K/AKT/mTOR
ground. Oncogene 2008; 27: 1800–1804. mavirus type 31. Journal of Virology 2004; pathway. Pharmacology & Therapeutics
23. Mesplède T, Gagnon D, Bergeron- 78: 12366–12377. 2014; 142: 164–175.
Labrecque F, et al. p53 degradation activ- 33. Favre-Bonvin A, Reynaud C, Kretz-Remy 43. Zhang X, Li X-r, Zhang J. Current status
ity, expression, and subcellular localiza- C, Jalinot P. Human papillomavirus type and future perspectives of PI3K and
tion of E6 proteins from 29 human 18 E6 protein binds the cellular PDZ pro- mTOR inhibitor as anticancer drugs in
papillomavirus genotypes. Journal of Virol- tein TIP-2/GIPC, which is involved in breast cancer. Current Cancer Drug Targets
ogy 2012; 86: 94–107. transforming growth factor β signaling 2013; 13: 175–187.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
44 J. Chen

44. Grunt W T, Mariani L, Novel G. Ap- and Mad during epidermal differentiation complex in early colorectal carcinogenesis.
proaches for molecular targeted therapy and HPV-associated tumorigenesis. Onco- Cancer Research 2000; 60: 4761–4766.
of breast cancer: interfering with gene 1995; 11: 2487–2501. 67. Green DW, Roh H, Pippin JA, Drebin JA.
PI3K/AKT/mTOR signaling. Current Can- 55. Dellas A, Schultheiss E, Leivas M, Moch β-catenin antisense treatment decreases β-
cer Drug Targets 2013; 13: 188–204. H, Torhorst J. Association of p27Kip1, cy- catenin expression and tumor growth rate
45. Chen J. Multiple signal pathways in clin E and c-myc expression with progres- in colon carcinoma xenografts. Journal of
obesity-associated cancer. Obesity Reviews sion and prognosis in HPV-positive Surgical Research 2001; 101: 16–20.
2011; 12: 1063–1070. cervical neoplasms. Anticancer Research 68. Yan D, Wiesmann M, Rohan M, et al. El-
46. Huang XF, Chen JZ. Obesity, the 1997; 18: 3991–3998. evated expression of axin2 and hnkd
PI3K/Akt signal pathway and colon 56. McMurray H, McCance D. Human papil- mRNA provides evidence that Wnt/β-
cancer. Obesity Reviews 2009; 10: lomavirus type 16 E6 activates TERT catenin signaling is activated in human
610–616. gene transcription through induction of colon tumors. Proceedings of the National
47. Contreras-Paredes A, De la Cruz- c-Myc and release of USF-mediated re- Academy of Sciences 2001; 98:
Hernández E, Martínez-Ramírez I, pression. Journal of Virology 2003; 77: 14973–14978.
Dueñas-González A, Lizano M. E6 vari- 9852–9861. 69. Baarsma HA, Königshoff M, Gosens R.
ants of human papillomavirus 18 differen- 57. Veldman T, Liu X, Yuan H, Schlegel R. Hu- The Wnt signaling pathway from ligand
tially modulate the protein kinase man papillomavirus E6 and Myc proteins secretion to gene transcription: molecular
B/phosphatidylinositol 3-kinase associate in vivo and bind to and coopera- mechanisms and pharmacological targets.
(akt/PI3K) signaling pathway. Virology tively activate the telomerase reverse tran- Pharmacology & Therapeutics 2013; 138:
2009; 383: 78–85. scriptase promoter. Proceedings of the 66–83.
48. Lu Z, Hu X, Li Y, et al. Human papilloma- National Academy of Sciences 2003; 100: 70. Pereira-Suárez AL, Meraz MA, Lizano M,
virus 16 E6 oncoprotein interferences with 8211–8216. et al. Frequent alterations of the β-catenin
insulin signaling pathway by binding to 58. Gross-Mesilaty S, Reinstein E, Bercovich B, protein in cancer of the uterine cervix. Tu-
tuberin. Journal of Biological Chemistry et al. Basal and human papillomavirus E6 mor Biology 2002; 23: 45–53.
2004; 279: 35664–35670. oncoprotein-induced degradation of Myc 71. Fadare O, Reddy H, Wang J, Hileeto D,
49. Zheng L, Ding H, Lu Z, et al. E3 ubiquitin proteins by the ubiquitin pathway. Pro- Schwartz PE, Zheng W. E-cadherin and β-
ligase E6AP-mediated TSC2 turnover in ceedings of the National Academy of Sciences catenin expression in early stage cervical
the presence and absence of HPV16 E6. 1998; 95: 8058–8063. carcinoma: a tissue microarray study of
Genes to Cells 2008; 13: 285–294. 59. Mantovani F, Banks L. The human papillo- 147 cases. World Journal of Surgical Oncol-
50. Spangle JM, Münger K. The human papil- mavirus E6 protein and its contribution to ogy 2005; 3: 38.
lomavirus type 16 E6 oncoprotein acti- malignant progression. Oncogene 2001; 20: 72. Shinohara A, Yokoyama Y, Wan X, et al.
vates mTORC1 signaling and increases 7874–7887. Cytoplasmic/nuclear expression without
protein synthesis. Journal of Virology 2010; 60. Reis M, Liebner S. Wnt signaling in the mutation of exon 3 of the β-catenin gene
84: 9398–9407. vasculature. Experimental Cell Research is frequent in the development of the neo-
51. Spangle JM, Munger K. The HPV16 E6 2013; 319: 1317–1323. plasm of the uterine cervix. Gynecologic
oncoprotein causes prolonged receptor 61. Clevers H, Nusse R. Wnt/β-catenin signal- Oncology 2001; 82: 450–455.
protein tyrosine kinase signaling and en- ing and disease. Cell 2012; 149: 1192–1205. 73. Rampias T, Boutati E, Pectasides E, et al.
hances internalization of phosphorylated 62. Moon RT, Kohn AD, De Ferrari GV, Activation of Wnt signaling pathway by
receptor species. PLoS Pathogens 2013; 9: Kaykas A. Wnt and β-catenin signalling: human papillomavirus E6 and E7 onco-
e1003237. diseases and therapies. Nature Reviews Ge- genes in HPV16-positive oropharyngeal
52. Boon SS, Banks L. High-risk human papil- netics 2004; 5: 691–701. squamous carcinoma cells. Molecular Can-
lomavirus E6 oncoproteins interact with 63. Klaus A, Birchmeier W. Wnt signalling cer Research 2010; 8: 433–443.
14-3-3ζ in a PDZ binding motif- and its impact on development and cancer. 74. Lichtig H, Gilboa DA, Jackman A, et al.
dependent manner. Journal of Virology Nature Reviews Cancer 2008; 8: 387–398. HPV16 E6 augments Wnt signaling in an
2013; 87: 1586–1595. 64. He T-C, Sparks AB, Rago C, et al. Identifi- E6AP-dependent manner. Virology 2010;
53. Kim SH, Koo BS, Kang S, et al. HPV inte- cation of c-MYC as a target of the APC 396: 47–58.
gration begins in the tonsillar crypt and pathway. Science 1998; 281: 1509–1512. 75. Bonilla-Delgado J, Bulut G, Liu X, et al. The
leads to the alteration of p16, EGFR and 65. Tetsu O, McCormick F. β-catenin regulates E6 oncoprotein from HPV16 enhances the
c-myc during tumor formation. Interna- expression of cyclin D1 in colon carcinoma canonical Wnt/β-catenin pathway in skin
tional Journal of Cancer 2007; 120: cells. Nature 1999; 398: 422–426. epidermis in vivo. Molecular Cancer Re-
1418–1425. 66. Yamada T, Takaoka AS, Naishiro Y, et al. search 2012; 10: 250–258.
54. Hurlin P, Foley K, Ayer D, Eisenman R, Transactivation of the multidrug resis- 76. Hatsell S, Rowlands T, Hiremath M,
Hanahan D, Arbeit J. Regulation of Myc tance 1 gene by T-cell factor 4/β-catenin Cowin P. β-catenin and Tcfs in mammary

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 45

development and cancer. Journal of Mam- osteolytic bone metastasis of breast cancer 98. Lefort K, Mandinova A, Ostano P, et al.
mary Gland Biology and Neoplasia 2003; 8: by engaging notch signaling in bone cells. Notch1 is a p53 target gene involved in hu-
145–158. Cancer Cell 2011; 19: 192–205. DOI: man keratinocyte tumor suppression
77. Fortini ME. Notch signaling: the core path- 10.1016/j.ccr.2010.12.022. through negative regulation of ROCK1/2
way and its posttranslational regulation. 89. Zagouras P, Stifani S, Blaumueller CM, and MRCKα kinases. Genes & Development
Developmental Cell 2009; 16: 633–647. Carcangiu ML, Artavanis-Tsakonas S. Al- 2007; 21: 562–577.
78. Artavanis-Tsakonas S, Rand MD, Lake RJ. terations in Notch signaling in neoplastic 99. Agrawal N, Frederick MJ, Pickering CR,
Notch signaling: cell fate control and sig- lesions of the human cervix. Proceedings of et al. Exome sequencing of head and neck
nal integration in development. Science the National Academy of Sciences 1995; 92: squamous cell carcinoma reveals
1999; 284: 770–776. 6414–6418. inactivating mutations in NOTCH1. Sci-
79. Aster JC. In brief: Notch signalling in 90. Daniel B, Rangarajan A, Mukherjee G, ence 2011; 333: 1154–1157.
health and disease. The Journal of Pathology Vallikad E, Krishna S. The link between in- 100. Stransky N, Egloff AM, Tward AD, et al.
2014; 232: 1–3. tegration and expression of human papil- The mutational landscape of head and
80. Dai Y, Wilson G, Huang B, et al. Silencing lomavirus type 16 genomes and cellular neck squamous cell carcinoma. Science
of Jagged1 inhibits cell growth and inva- changes in the evolution of cervical 2011; 333: 1157–1160.
sion in colorectal cancer. Cell Death & Dis- intraepithelial neoplastic lesions. Journal 101. Sun W, Gaykalova DA, Ochs MF, et al. Ac-
ease 2014; 5: e1170. of General Virology 1997; 78: 1095–1101. tivation of the NOTCH pathway in head
81. Radtke F, MacDonald HR, Tacchini-Cottier 91. Veeraraghavalu K, Pett M, Kumar RV, et al. and neck cancer. Cancer Research 2014; 74:
F. Regulation of innate and adaptive im- Papillomavirus-mediated neoplastic pro- 1091–1104.
munity by Notch. Nature Reviews Immunol- gression is associated with reciprocal 102. Bernardes de Jesus B, Blasco MA. Telome-
ogy 2013; 13: 427–437. changes in JAGGED1 and manic fringe ex- rase at the intersection of cancer and ag-
82. Hori K, Sen A, Artavanis-Tsakonas S. pression linked to notch activation. Journal ing. Trends in Genetics 2013; 29: 513–520.
Notch signaling at a glance. Journal of Cell of Virology 2004; 78: 8687–8700. 103. Günes C, Rudolph KL. The role of telo-
Science 2013; 126: 2135–2140. 92. Vliet-Gregg PA, Hamilton JR, meres in stem cells and cancer. Cell 2013;
83. Capaccione KM, Pine SR. The Notch sig- Katzenellenbogen RA. NFX1-123 and hu- 152: 390–393.
naling pathway as a mediator of tumor man papillomavirus 16E6 increase notch 104. Mocellin S, Pooley KA, Nitti D. Telome-
survival. Carcinogenesis 2013; 34: expression in keratinocytes. Journal of Vi- rase and the search for the end of cancer.
1420–1430. rology 2013; 87: 13741–13750. Trends in Molecular Medicine 2013; 19:
84. Santagata S, Demichelis F, Riva A, et al. 93. Weijzen S, Zlobin A, Braid M, Miele L, 125–133.
JAGGED1 expression is associated with Kast WM. HPV16 E6 and E7 oncoproteins 105. Xu L, Li S, Stohr BA. The role of telomere
prostate cancer metastasis and recurrence. regulate Notch-1 expression and cooperate biology in cancer. Annual Review of Pathol-
Cancer Research 2004; 64: 6854–6857. DOI: to induce transformation. Journal of Cellu- ogy: Mechanisms of Disease 2013; 8: 49–78.
10.1158/0008-5472.CAN-04-2500. lar Physiology 2003; 194: 356–362. 106. Bodnar AG, Ouellette M, Frolkis M, et al.
85. Purow BW, Haque RM, Noel MW, et al. 94. Talora C, Sgroi DC, Crum CP, Dotto GP. Extension of life-span by introduction of
Expression of Notch-1 and its ligands, Specific down-modulation of Notch1 sig- telomerase into normal human cells. Sci-
Delta-like-1 and Jagged-1, is critical for gli- naling in cervical cancer cells is required ence 1998; 279: 349–352.
oma cell survival and proliferation. Cancer for sustained HPV-E6/E7 expression 107. Hahn WC, Stewart SA, Brooks MW, et al.
Research 2005; 65: 2353–2363. DOI: and late steps of malignant transforma- Inhibition of telomerase limits the growth
10.1158/0008-5472.CAN-04-1890. tion. Genes & Development 2002; 16: of human cancer cells. Nature Medicine
86. Reedijk M, Odorcic S, Chang L, et al. High- 2252–2263. 1999; 5: 1164–1170.
level coexpression of JAG1 and NOTCH1 95. Wang NJ, Sanborn Z, Arnett KL, et al. 108. Klingelhutz AJ, Foster SA, McDougall JK.
is observed in human breast cancer and Loss-of-function mutations in Notch re- Telomerase activation by the E6 gene
is associated with poor overall survival. ceptors in cutaneous and lung squamous product of human papillomavirus type
Cancer Research 2005; 65: 8530–8537. DOI: cell carcinoma. Proceedings of the National 16. 1996.
10.1158/0008-5472.CAN-05-1069. Academy of Sciences 2011; 108: 109. Veldman T, Horikawa I, Barrett JC, Schle-
87. Lin JT, Chen MK, Yeh KT, et al. Association 17761–17766. gel R. Transcriptional activation of the tel-
of high levels of Jagged-1 and Notch-1 ex- 96. Klinakis A, Lobry C, Abdel-Wahab O, et al. omerase hTERT gene by human
pression with poor prognosis in head and A novel tumour-suppressor function for papillomavirus type 16 E6 oncoprotein.
neck cancer. Annals of Surgical Oncology the Notch pathway in myeloid leukaemia. Journal of Virology 2001; 75: 4467–4472.
2010; 17: 2976–2983. DOI: 10.1245/ Nature 2011; 473: 230–233. 110. Liu X, Dakic A, Zhang Y, Dai Y, Chen R,
s10434-010-1118-9. 97. Network CGAR. Comprehensive genomic Schlegel R. HPV E6 protein interacts phys-
88. Sethi N, Dai X, Winter CG, Kang Y. Tu- characterization of squamous cell lung ically and functionally with the cellular tel-
mor-derived JAGGED1 promotes cancers. Nature 2012; 489: 519-525. omerase complex. Proceedings of the

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
46 J. Chen

National Academy of Sciences 2009; 106: target genes in leukemia. Molecular Medi- Sequence evolution of the intrinsically dis-
18780–18785. cine Reports 2014; 9: 1283–1288. ordered and globular domains of a model
111. Ling H, Fabbri M, Calin GA. MicroRNAs 122. Kastl L, Brown I, Schofield A. miRNA- viral oncoprotein. PloS One 2012; 7:
and other non-coding RNAs as targets for 34a is associated with docetaxel resis- e47661.
anticancer drug development. Nature Re- tance in human breast cancer cells. Breast 133. Roman A, Munger K. The papillomavirus
views Drug Discovery 2013; 12: 847–865. Cancer Research and Treatment 2012; 131: E7 proteins. Virology 2013; 445: 138–168.
112. Martinez I, Gardiner A, Board K, Monzon 445–454. 134. Dick FA, Rubin SM. Molecular mecha-
F, Edwards R, Khan S. Human papilloma- 123. Du R, Sun W, Xia L, et al. Hypoxia-induced nisms underlying RB protein function. Na-
virus type 16 reduces the expression of down-regulation of microRNA-34a pro- ture Reviews Molecular Cell Biology 2013; 14:
microRNA-218 in cervical carcinoma cells. motes EMT by targeting the Notch signal- 297–306.
Oncogene 2008; 27: 2575–2582. ing pathway in tubular epithelial cells. 135. Rubin SM. Deciphering the retinoblastoma
113. Yamamoto N, Kinoshita T, Nohata N, et al. PloS One 2012; 7: e30771. protein phosphorylation code. Trends in
Tumor suppressive microRNA-218 in- 124. Ribeiro J, Sousa H. MicroRNAs as bio- Biochemical Sciences 2013; 38: 12–19.
hibits cancer cell migration and invasion markers of cervical cancer development: a 136. Di Fiore R, D’Anneo A, Tesoriere G, Vento
by targeting focal adhesion pathways in literature review on miR-125b and miR-34a. R. RB1 in cancer: different mechanisms of
cervical squamous cell carcinoma. Interna- Molecular Biology Reports 2014; 41: 1525–1531. RB1 inactivation and alterations of pRb
tional Journal of Oncology 2013; 42: 1523– 125. Cui F, Li X, Zhu X, et al. MiR-125b inhibits pathway in tumorigenesis. Journal of Cellu-
1532. tumor growth and promotes apoptosis of lar Physiology 2013; 228: 1676–1687.
114. Wang X, Wang H-K, McCoy JP, et al. Onco- cervical cancer cells by targeting 137. Park JW, Shin M-K, Pitot HC, Lambert PF.
genic HPV infection interrupts the expres- phosphoinositide 3-kinase catalytic sub- High incidence of HPV-associated head
sion of tumor-suppressive miR-34a unit delta. Cellular Physiology and Biochem- and neck cancers in FA deficient mice is as-
through viral oncoprotein E6. RNA 2009; istry 2012; 30: 1310–1318. sociated with E7’s induction of DNA dam-
15: 637–647. 126. Nuovo GJ, Wu X, Volinia S, et al. Strong in- age through its inactivation of pocket
115. Zheng Z-M, Wang X. Regulation of cellu- verse correlation between microRNA-125b proteins. PloS One 2013; 8: e75056.
lar miRNA expression by human papillo- and human papillomavirus DNA in pro- 138. Münger K, Werness B, Dyson N, Phelps W,
maviruses. Biochimica et Biophysica Acta ductive infection. Diagnostic Molecular Pa- Harlow E, Howley P. Complex formation of
(BBA)-Gene Regulatory Mechanisms 2011; thology 2010; 19: 135–143. human papillomavirus E7 proteins with the
1809: 668–677. 127. Wang Y, Cai N, Wu X, Cao H, Xie L, retinoblastoma tumor suppressor gene prod-
116. Chen F, Hu SJ. Effect of microRNA-34a in Zheng P. OCT4 promotes tumorigenesis uct. The EMBO Journal 1989; 8: 4099–4105.
cell cycle, differentiation, and apoptosis: a and inhibits apoptosis of cervical cancer 139. Ghittoni R, Accardi R, Hasan U, Gheit T,
review. Journal of Biochemical and Molecular cells by miR-125b/BAK1 pathway. Cell Sylla B, Tommasino M. The biological
Toxicology 2012; 26: 79–86. Death & Disease 2013; 4: e760. properties of E6 and E7 oncoproteins from
117. Hermeking H. The miR-34 family in can- 128. Banzhaf-Strathmann J, Edbauer D. Good human papillomaviruses. Virus Genes
cer and apoptosis. Cell Death & Differentia- guy or bad guy: the opposing roles of 2010; 40: 1–13.
tion 2010; 17: 193–199. microRNA 125b in cancer. Cell Communica- 140. Münger K, Basile JR, Duensing S, et al. Bio-
118. Li B, Hu Y, Ye F, Li Y, Lv W, Xie X. Re- tion and Signaling 2014; 12: 30. logical activities and molecular targets of
duced miR-34a expression in normal cervi- 129. Jung HM, Phillips BL, Chan EK. miR-375 the human papillomavirus E7 oncoprotein.
cal tissues and cervical lesions with high- activates p21 and suppresses telomerase Oncogene 2001; 20: 7888–7898.
risk human papillomavirus infection. In- activity by coordinately regulating HPV 141. McLaughlin-Drubin ME, Münger K. The
ternational Journal of Gynecological Cancer E6/E7, E6AP, CIP2A, and 14-3-3zeta. Mo- human papillomavirus E7 oncoprotein. Vi-
2010; 20: 597–604. lecular Cancer 2014; 13: 80. rology 2009; 384: 335–344.
119. Xie X, Piao L, Bullock B, et al. Targeting 130. Tsukamoto Y, Nakada C, Noguchi T, et al. 142. Huh K, Zhou X, Hayakawa H, et al. Hu-
HPV16 E6-p300 interaction reactivates MicroRNA-375 is downregulated in gas- man papillomavirus type 16 E7
p53 and inhibits the tumorigenicity of tric carcinomas and regulates cell survival oncoprotein associates with the cullin 2
HPV-positive head and neck squamous cell by targeting PDK1 and 14-3-3ζ. Cancer Re- ubiquitin ligase complex, which contrib-
carcinoma. Oncogene 2013; 33: 1037–1046. search 2010; 70: 2339–2349. utes to degradation of the retinoblastoma
120. Wang X, Meyers C, Guo M, Zheng ZM. 131. Wang X, Wang H-K, Li Y,et al. tumor suppressor. Journal of Virology 2007;
Upregulation of p18Ink4c expression by microRNAs are biomarkers of oncogenic 81: 9737–9747.
oncogenic HPV E6 via p53-miR-34a path- human papillomavirus infections. Proceed- 143. Heck DV, Yee CL, Howley PM, Münger K.
way. International Journal of Cancer 2011; ings of the National Academy of Sciences Efficiency of binding the retinoblastoma
129: 1362–1372. 2014; 111: 4262–4267. protein correlates with the transforming
121. Tang R, Li J, Yue M, et al. A correlation 132. Chemes LB, Glavina J, Alonso LG, Ma- capacity of the E7 oncoproteins of the hu-
analysis of miRNA 34a and its predicted rino-Buslje C, de Prat-Gay G, Sánchez IE. man papillomaviruses. Proceedings of the

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 47

National Academy of Sciences 1992; 89: 154. Etienne-Manneville S, Hall A. Rho 7d and miR-205 are prognostic markers of
4442–4446. GTPases in cell biology. Nature 2002; 420: head and neck squamous cell carcinoma.
144. Sang B-C, Barbosa MS. Single amino acid 629–635. The American Journal of Pathology 2009;
substitutions in" low-risk" human papillo- 155. Ridley AJ. Rho GTPases and actin dynam- 174: 736–745.
mavirus (HPV) type 6 E7 protein enhance ics in membrane protrusions and vesicle 166. Xie H, Zhao Y, Caramuta S, Larsson C,
features characteristic of the" high-risk" trafficking. Trends in Cell Biology 2006; 16: Lui W-O. miR-205 expression promotes
HPV E7 oncoproteins. Proceedings of the 522–529. cell proliferation and migration of human
National Academy of Sciences 1992; 89: 156. Wojnacki J, Quassollo G, Marzolo MP, cervical cancer cells. PloS One 2012; 7:
8063–8067. Cáceres A. Rho GTPases at the crossroad e46990.
145. Hwang SG, Lee D, Kim J, Seo T, Choe J. of signaling networks in mammals: Im- 167. Tong X, Xie D, O’Kelly J, Miller CW,
Human papillomavirus type 16 E7 binds pact of Rho-GTPases on microtubule or- Muller-Tidow C, Koeffler HP. Cyr61, a
to E2F1 and activates E2F1-driven tran- ganization and dynamics. Small GTPases member of CCN family, is a tumor sup-
scription in a retinoblastoma protein- 2014; 5: 0-1. pressor in non-small cell lung cancer.
independent manner. Journal of Biological 157. Todorovic B, Nichols AC, Chitilian JM, Journal of Biological Chemistry 2001; 276:
Chemistry 2002; 277: 2923–2930. et al. The human papillomavirus E7 pro- 47709–47714.
146. McLaughlin-Drubin ME, Huh K-W, teins associate with and alter 168. Chuang J-Y, Yang W-Y, Lai C-H, Lin C-D,
Münger K. Human papillomavirus type p190RhoGAP function. Journal of Virology Tsai M-H, Tang C-H. CTGF inhibits cell
16 E7 oncoprotein associates with E2F6. 2014; JVI: 03263–03213. motility and COX-2 expression in oral can-
Journal of Virology 2008; 82: 8695–8705. 158. Melar-New M, Laimins LA. Human papil- cer cells. International Immunopharmacology
147. Collins AS, Nakahara T, Do A, Lambert lomaviruses modulate expression of 2011; 11: 948–954.
PF. Interactions with pocket proteins microRNA 203 upon epithelial differentia- 169. Ofir M, Hacohen D, Ginsberg D. MiR-15
contribute to the role of human papillo- tion to control levels of p63 proteins. Jour- and miR-16 are direct transcriptional tar-
mavirus type 16 E7 in the papillomavi- nal of Virology 2010; 84: 5212–5221. gets of E2F1 that limit E2F-induced prolif-
rus life cycle. Journal of Virology 2005; 159. Bo J, Yang G, Huo K,et al. microRNA-203 eration by targeting cyclin E. Molecular
79: 14769–14780. suppresses bladder cancer development Cancer Research 2011; 9: 440-447.
148. Shin M-K, Sage J, Lambert PF. Inactivating by repressing bcl-w expression. FEBS Jour- 170. Myklebust M, Bruland O, Fluge Ø,
all three Rb family pocket proteins is insuf- nal 2011; 278: 786–792. Skarstein A, Balteskard L, Dahl O.
ficient to initiate cervical cancer. Cancer Re- 160. Bian K, Fan J, Zhang X, et al. MicroRNA- MicroRNA-15b is induced with E2F-
search 2012; 72: 5418–5427. 203 leads to G1 phase cell cycle arrest in la- controlled genes in HPV-related cancer.
149. Menges CW, Baglia LA, Lapoint R, ryngeal carcinoma cells by directly British Journal of Cancer 2011; 105:
McCance DJ. Human papillomavirus type targeting survivin. FEBS Letters 2012; 586: 1719–1725.
16 E7 up-regulates AKT activity through 804–809. 171. Wang X, Tang S, Le S-Y, et al. Aberrant ex-
the retinoblastoma protein. Cancer Re- 161. Takeshita N, Mori M, Kano M, et al. miR- pression of oncogenic and tumor-
search 2006; 66: 5555–5559. 203 inhibits the migration and invasion of suppressive microRNAs in cervical cancer
150. Pim D, Massimi P, Dilworth SM, Banks L. esophageal squamous cell carcinoma by is required for cancer cell growth. PloS One
Activation of the protein kinase B pathway regulating LASP1. International Journal of 2008; 3: e2557.
by the HPV-16 E7 oncoprotein occurs Oncology 2012; 41: 1653–1661. 172. Aqeilan R, Calin G, Croce C. miR-15a and
through a mechanism involving interac- 162. McKenna DJ, Patel D, McCance DJ. miR- miR-16-1 in cancer: discovery, function
tion with PP2A. Oncogene 2005; 24: 24 and miR-205 expression is dependent and future perspectives. Cell Death & Dif-
7830–7838. on HPV onco-protein expression in ferentiation 2010; 17: 215–220.
151. Liu J, Wang X, Zhou G, et al. Cancerous in- keratinocytes. Virology 2014; 448: 210–216. 173. Bonci D, Coppola V, Musumeci M, et al.
hibitor of protein phosphatase 2A is 163. Wang Z, Liao H, Deng Z, et al. miRNA-205 The miR-15a–miR-16-1 cluster controls
overexpressed in cervical cancer and up- affects infiltration and metastasis of breast prostate cancer by targeting multiple on-
regulated by human papillomavirus 16 cancer. Biochemical and Biophysical Research cogenic activities. Nature Medicine 2008;
E7 oncoprotein. Gynecologic Oncology Communications 2013; 441: 139–143. 14: 1271–1277.
2011; 122: 430–436. 164. Dar AA, Majid S, de Semir D, Nosrati M, 174. Wang F, Fu X-D, Zhou Y, Zhang Y. Down-
152. Charette S, McCance D. The E7 protein Bezrookove V, Kashani-Sabet M. miRNA- regulation of the cyclin E1 oncogene ex-
from human papillomavirus type 16 en- 205 suppresses melanoma cell prolifera- pression by microRNA-16-1 induces cell
hances keratinocyte migration in an Akt- tion and induces senescence via regulation cycle arrest in human cancer cells. BMB
dependent manner. Oncogene 2007; 26: of E2F1 protein. Journal of Biological Chem- Reports 2009; 42: 725–730.
7386–7390. istry 2011; 286: 16606–16614. 175. Krawczyk E, Suprynowicz FA, Liu X, et al.
153. Hall A. Rho GTPases and the actin cyto- 165. Childs G, Fazzari M, Kung G, et al. Low- Koilocytosis: a cooperative interaction be-
skeleton. Science 1998; 279: 509–514. level expression of MicroRNAs let- tween the human papillomavirus E5 and

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
48 J. Chen

E6 oncoproteins. The American Journal of 185. Villa-Morales M, Fernández-Piqueras J. 196. Zubillaga-Guerrero M, Illades-Aguiar B,
Pathology 2008; 173: 682–688. Targeting the Fas/FasL signaling pathway Leyva-Vazquez M, et al. The integration
176. Jost M, Kari C, Rodeck U. The EGF recep- in cancer therapy. Expert Opinion on Thera- of HR-HPV increases the expression of
tor - an essential regulator of multiple epi- peutic Targets 2012; 16: 85–101. cyclins A and E in cytologies with and
dermal functions. European Journal of 186. Kabsch K, Alonso A. The human papillo- without low-grade lesions. Journal of
Dermatology 2000; 10: 505–510. mavirus type 16 E5 protein impairs Cytology/Indian Academy of Cytologists
177. Jost M, Huggett TM, Kari C, Boise LH, TRAIL-and FasL-mediated apoptosis in 2013; 30: 1–7.
Rodeck U. Epidermal growth factor HaCaT cells by different mechanisms. Jour- 197. Nguyen CL, Münger K. Direct association
receptor-dependent control of keratinocyte nal of Virology 2002; 76: 12162–12172. of the HPV16 E7 oncoprotein with cyclin
survival and Bcl-xL expression through a 187. Kabsch K, Mossadegh N, Kohl A, et al. The A/CDK2 and cyclin E/CDK2 complexes.
MEK-dependent pathway. The Journal of HPV-16 E5 protein inhibits TRAIL-and Virology 2008; 380: 21–25.
Biological Chemistry 2001; 276: 6320–6326.- FasL-mediated apoptosis in human 198. Yoshinouchi M, Yamada T, Kizaki M, et al.
DOI: 10.1074/jbc.M008210200 DOI: keratinocyte raft cultures. Intervirology In vitro and in vivo growth suppression of
10.1074/jbc.M008210200. 2004; 47: 48–56. human papillomavirus 16-positive cervi-
178. Chen J, Elfiky A, Han M, Chen C, Saif 188. Regan JA, Laimins LA. Bap31 is a novel cal cancer cells by E6 siRNA. Molecular
MW. The role of Src in colon cancer and target of the human papillomavirus E5 Therapy 2003; 8: 762–768.
its therapeutic implications. Clinical Colo- protein. Journal of Virology 2008; 82: 199. Jonson AL, Rogers LM, Ramakrishnan S,
rectal Cancer 2014; 13: 5–13. 10042–10051. Downs Jr LS. Gene silencing with siRNA
179. Pim D, Collins M, Banks L. Human papil- 189. Iwasawa R, Mahul-Mellier AL, Datler C, targeting E6/E7 as a therapeutic interven-
lomavirus type 16 E5 gene stimulates the Pazarentzos E, Grimm S. Fis1 and tion in a mouse model of cervical cancer.
transforming activity of the epidermal Bap31 bridge the mitochondria–ER inter- Gynecologic Oncology 2008; 111: 356–364.
growth factor receptor. Oncogene 1992; 7: face to establish a platform for apoptosis 200. Debnath J, Walker SJ, Brugge JS. Akt acti-
27–32. induction. The EMBO Journal 2011; 30: vation disrupts mammary acinar architec-
180. Crusius K, Auvinen E, Steuer B, Gaissert 556–568. ture and enhances proliferation in an
H, Alonso A. The human papillomavirus 190. Kotnik Halavaty K, Regan J, Mehta K, mTOR-dependent manner. The Journal of
type 16 E5-protein modulates ligand- Laimins L. Human papillomavirus E5 Cell Biology 2003; 163: 315–326.
dependent activation of the EGF receptor oncoproteins bind the A4 endoplasmic re- 201. Coppock JD, Wieking BG, Molinolo AA,
family in the human epithelial cell line ticulum protein to regulate proliferative Gutkind JS, Miskimins WK, Lee JH. Im-
HaCaT. Experimental Cell Research 1998; ability upon differentiation. Virology 2014; proved clearance during treatment of
241: 76–83. 452: 223–230. HPV-positive head and neck cancer
181. Martin P, Vass WC, Schiller JT, Lowy DR, 191. Hanahan D, Weinberg RA. Hallmarks of through mTOR inhibition. Neoplasia (New
Velu TJ. The bovine papillomavirus E5 cancer: the next generation. Cell 2011; 144: York, NY) 2013; 15: 620–630.
transforming protein can stimulate the 646–674. 202. Conesa-Zamora P, Doménech-Peris A,
transforming activity of EGF and CSF-1 re- 192. Bretones G, Delgado MD, León J. Myc and Orantes-Casado FJ, et al. Effect of human
ceptors. Cell 1989; 59: 21–32. cell cycle control. Biiochimica et Biophysica Acta papillomavirus on cell cycle–related pro-
182. Kim S-H, Juhnn Y-S, Kang S, et al. Hu- (BBA) - Gene Regulatory Mechanisms 2014. teins p16, Ki-67, Cyclin D1, p53, and
man papillomavirus 16 E5 up-regulates DOI: 10.1016/j.pharmthera.2013.12.004. ProEx C in precursor lesions of cervical
the expression of vascular endothelial 193. Cho NH, Kang S, Hong S, et al. Elevation carcinoma A tissue microarray study.
growth factor through the activation of of cyclin B1, active cdc2, and HuR in cervi- American Journal of Clinical Pathology
epidermal growth factor receptor, cal neoplasia with human papillomavirus 2009; 132: 378–390.
MEK/ERK1, 2 and PI3K/Akt. Cellular and type 18 infection. Cancer Letters 2006; 232: 203. Portari EA, Russomano FB, de Camargo
Molecular Life Sciences 2006; 63: 930–938. 170–178. MJ, et al. Immunohistochemical expression
183. Crusius K, Rodriguez I, Alonso A. The hu- 194. Hashiguchi Y, Tsuda H, Nishimura S, of cyclin D1, p16Ink4a, p21WAF1, and Ki-
man papillomavirus type 16 E5 protein Inoue T, Kawamura N, Yamamoto K. Re- 67 correlates with the severity of cervical
modulates ERK1/2 and p38 MAP kinase lationship between HPV typing and the neoplasia. International Journal of Gyneco-
activation by an EGFR-independent pro- status of G2 cell cycle regulators in cervi- logic Pathology 2013; 32: 501–508.
cess in stressed human keratinocytes. Vi- cal neoplasia. Oncology Reports 2004; 12: 204. Butz K, Denk C, Ullmann A, Scheffner M,
rus Genes 2000; 20: 65–69. 587–591. Hoppe-Seyler F. Induction of apoptosis in
184. Yuan C-H, Filippova M, Duerksen- 195. Cho NH, Lim SY, Kim YT, Kim D, Kim YS, human papillomavirus positive cancer
Hughes P. Modulation of apoptotic path- Kim JW. G2 checkpoint in uterine cervical cells by peptide aptamers targeting the
ways by human papillomaviruses (HPV): cancer with HPV 16 E6 according to p53 viral E6 oncoprotein. Proceedings of the
mechanisms and implications for therapy. polymorphism and its screening value. National Academy of Sciences 2000; 97:
Viruses 2012; 4: 3831–3850. Gynecologic Oncology 2003; 90: 15–22. 6693–6697.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 49

205. Qi Z, Xu X, Zhang B, et al. Effect of simulta- Polo-like kinase 4 expression. Molecular 226. Gottesman MM. Mechanisms of cancer
neous silencing of HPV-18 E6 and E7 on in- Cancer 2011; 10: 61. drug resistance. Annual Review of Medicine
ducing apoptosis in HeLa cells this paper is 215. Habedanck R, Stierhof Y-D, Wilkinson CJ, 2002; 53: 615–627.
one of a selection of papers published in Nigg EA. The Polo kinase Plk4 functions 227. Longley D, Johnston P. Molecular mecha-
this special issue entitled “Second Interna- in centriole duplication. Nature Cell Biology nisms of drug resistance. The Journal of Pa-
tional Symposium on Recent Advances in 2005; 7: 1140–1146. thology 2005; 205: 275–292.
Basic, Clinical, and Social Medicine” and 216. Yu JJ, Fu P, Pink JJ, et al. HPV infection 228. Luqmani Y. Mechanisms of drug resis-
has undergone the Journal’s usual peer re- and EGFR activation/alteration in tance in cancer chemotherapy. Medical
view process. Biochemistry and Cell Biology HIV-infected East African patients with Principles and Practice 2008; 14: 35–48.
2010; 88: 697–704. conjunctival carcinoma. PloS One 2010; 229. Chen J, Huang X-F, Qiao L, Katsifis A. In-
206. Smeets SJ, van der Plas M, Schaaij-Visser T, 5: e10477. DOI: 10.1371/journal.pone.00 sulin caused drug resistance to oxaliplatin
et al. Immortalization of oral keratinocytes 10477. in colon cancer cell line HT29. Journal of
by functional inactivation of the p53 and 217. Kersemaekers A-MF, Fleuren GJ, Kenter Gastrointestinal Oncology 2011; 2: 27–33.
pRb pathways. International Journal of Can- GG, et al. Oncogene alterations in carcino- 230. Chen J, Katsifis A, Hu C, Huang X-F. Insu-
cer 2011; 128: 1596–1605. mas of the uterine cervix: overexpression lin decreases therapeutic efficacy in colon
207. Thomas M, Banks L. Inhibition of Bak- of the epidermal growth factor receptor is cancer cell line HT29 via the activation of
induced apoptosis by HPV-18 E6. Onco- associated with poor prognosis. Clinical the PI3K/Akt pathway. Current Drug Dis-
gene 1998; 17: 2943–2954. Cancer Research 1999; 5: 577–586. covery Technologies 2011; 8: 119–125.
208. Guirouilh-Barbat J, Wilhelm T, Lopez BS. 218. Wright AA, Howitt BE, Myers AP, et al. 231. Tsuruo T, Naito M, Tomida A, et al. Molec-
AKT1/BRCA1 in the control of homolo- Oncogenic mutations in cervical cancer. ular targeting therapy of cancer: drug re-
gous recombination and genetic stability: Cancer 2013; 119: 3776–3783. sistance, apoptosis and survival signal.
the missing link between hereditary and 219. Wierzbicka M, Jozefiak A, Szydlowski J, Cancer Science 2003; 94: 15–21.
sporadic breast cancers. Oncotarget 2010; et al. Recommendations for the diagnosis 232. Smalley KS, Haass NK, Brafford PA, Lioni
1: 691. of human papilloma virus (HPV) high M, Flaherty KT, Herlyn M. Multiple sig-
209. Hernando E, Nahlé Z, Juan G, et al. Rb in- and low risk in the prevention and treat- naling pathways must be targeted to over-
activation promotes genomic instability by ment of diseases of the oral cavity, phar- come drug resistance in cell lines derived
uncoupling cell cycle progression from mi- ynx and larynx. Guide of experts PTORL from melanoma metastases. Molecular
totic control. Nature 2004; 430: 797–802. and KIDL. Otolaryngologia Polska. The Pol- Cancer Therapeutics 2006; 5: 1136–1144.
210. Tsou M-FB, Wang W-J, George KA, Uryu ish Otolaryngology 2013; 67: 113–134. DOI: 233. Xia S, Zhao Y, Yu S, Zhang M. Activated
K, Stearns T, Jallepalli PV. Polo kinase 10.1016/j.otpol.2013.01.003. PI3K/Akt/COX-2 pathway induces resis-
and separase regulate the mitotic licensing 220. Anderson JA, Irish JC, McLachlin CM, tance to radiation in human cervical can-
of centriole duplication in human cells. De- Ngan BY. H-ras oncogene mutation and cer HeLa cells. Cancer Biotherapy &
velopmental Cell 2009; 17: 344–354. human papillomavirus infection in oral Radiopharmaceuticals 2010; 25: 317–323.
211. Duensing S, Duensing A, Crum CP, carcinomas. Archives of Otolaryngology-- 234. Wu H-H, Wu J-Y, Cheng Y-W, et al.
Münger K. Human papillomavirus type Head & Neck Surgery 1994; 120: 755–760. cIAP2 upregulated by E6 Oncoprotein
16 E7 oncoprotein-induced abnormal cen- 221. Ojesina AI, Lichtenstein L, Freeman SS, et al. via epidermal growth factor
trosome synthesis is an early event in the Landscape of genomic alterations in cervical receptor/phosphatidylinositol 3-kinase/
evolving malignant phenotype. Cancer Re- carcinomas. Nature 2014; 506: 371–375. AKT pathway confers resistance to cis-
search 2001; 61: 2356–2360. 222. Holohan C, Van Schaeybroeck S, Longley platin in human papillomavirus 16/18–
212. Duensing A, Chin A, Wang L, Kuan S-F, DB, Johnston PG. Cancer drug resistance: infected lung cancer. Clinical Cancer Re-
Duensing S. Analysis of centrosome an evolving paradigm. Nature Reviews search 2010; 16: 5200–5210.
overduplication in correlation to cell divi- Cancer 2013; 13: 714–726. 235. Lin G, Chen Q, Yu S, et al. Overexpression
sion errors in high-risk human papilloma- 223. Chen J, Shao R, Zhang XD, Chen C. Appli- of human telomerase reverse transcriptase
virus (HPV)-associated anal neoplasms. cations of nanotechnology for melanoma C-terminal polypeptide sensitizes HeLa
Virology 2008; 372: 157–164. treatment, diagnosis, and theranostics. In- cells to 5-fluorouracil induced growth in-
213. Korzeniewski N, Zheng L, Cuevas R, et al. ternational Journal of Nanomedicine 2013; 8: hibition and apoptosis. Molecular Medicine
Cullin 1 functions as a centrosomal sup- 2677. Reports 2014; 9: 279–284.
pressor of centriole multiplication by regu- 224. Dean M, Fojo T, Bates S. Tumour stem cells 236. Chen J. Targeted therapy of obesity-
lating polo-like kinase 4 protein levels. and drug resistance. Nature Reviews Cancer associated colon cancer. Translational Gas-
Cancer Research 2009; 69: 6668–6675. 2005; 5: 275–284. trointestinal Cancer 2011; 1: 44–57.
214. Korzeniewski N, Treat B, Duensing S. The 225. Chen J, Raymond K. Nuclear receptors, 237. Downward J. Targeting RAS signalling
HPV-16 E7 oncoprotein induces centriole bile-acid detoxification, and cholestasis. pathways in cancer therapy. Nature Re-
multiplication through deregulation of The Lancet 2006; 367: 454–456. views Cancer 2003; 3: 11–22.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
50 J. Chen

238. Chen J. Is Src the key to understanding approaches for cervical cancer. Cancer 2009; 259. Kafri T, van Praag H, Gage FH, Verma IM.
metastasis and developing new treatments 115: 3166–3180. Lentiviral vectors: regulated gene expres-
for colon cancer? Nature Clinical Practice. 250. Duenãs-González A, Cetina L, Coronel J, sion. Molecular Therapy 2000; 1: 516–521.
Gastroenterology & Hepatology 2008; 5: Martínez-Baños D. Pharmacotherapy op- DOI: 10.1006/mthe.2000.0083.
306–307. tions for locally advanced and advanced 260. Miyoshi H, Blomer U, Takahashi M, Gage
239. Eijsink JJ, Noordhuis MG, ten Hoor KA, cervical cancer. Drugs 2010; 70: 403–432. FH, Verma IM. Development of a self-
et al. The epidermal growth factor receptor 251. Weiss GR, Green S, Hannigan EV, et al. A inactivating lentivirus vector. Journal of
pathway in relation to pelvic lymph node phase II trial of cisplatin and 5- Virology 1998; 72: 8150–8157.
metastasis and survival in early-stage cer- fluorouracil with allopurinol for recurrent 261. Logan AC, Haas DL, Kafri T, Kohn DB.
vical cancer. Human Pathology 2010; 41: or metastatic carcinoma of the uterine cer- Integrated self-inactivating lentiviral vec-
1735–1741. vix: a Southwest oncology group trial. Gy- tors produce full-length genomic tran-
240. Su P, Lin Y, Huang R, et al. Epigenetic si- necologic Oncology 1990; 37: 354–358. scripts competent for encapsidation and
lencing of PTPRR activates MAPK signal- 252. Kaern J, Trope C, Abeler V, Iversen T, integration. Journal of Virology 2004; 78:
ing, promotes metastasis and serves as a Kjørstad K. A phase II study of 5- 8421–8436. DOI: 10.1128/JVI.78.16.8421–
biomarker of invasive cervical cancer. On- fluorouracil/cisplatinum in recurrent cer- 8436.2004..
cogene 2012; 32: 15–26. vical cancer. Acta Oncologica 1990; 29: 262. Gu W, Putral L, Hengst K, et al. Inhibi-
241. Amine A, Rivera S, Opolon P, et al. Novel 25–28. tion of cervical cancer cell growth
anti-metastatic action of cidofovir medi- 253. Bonomi P, Blessing J, Ball H, Hanjani P, in vitro and in vivo with lentiviral-vector
ated by inhibition of E6/E7, CXCR4 and DiSaia PJ. A phase II evaluation of cis- delivered short hairpin RNA targeting
Rho/ROCK signaling in HPV+ tumor platin and 5-fluorouracil in patients with human papillomavirus E6 and E7 onco-
cells. PloS One 2009; 4: e5018. advanced squamous cell carcinoma of genes. Cancer Gene Therapy 2006; 13:
242. Reya T, Morrison SJ, Clarke MF, Weissman the cervix: a gynecologic oncology group 1023–1032. DOI: 10.1038/sj.cgt.7700971.
IL. Stem cells, cancer, and cancer stem study. Gynecologic Oncology 1989; 34: 263. Gu W, Payne E, Sun S, Burgess M, McMil-
cells. Nature 2001; 414: 105–111. 357–359. lan NA. Inhibition of cervical cancer cell
243. Frank NY, Schatton T, Frank MH. The 254. Benjapibal M, Thirapakawong C, growth in vitro and in vivo with dual
therapeutic promise of the cancer stem cell Leelaphatanadit C, Therasakvichya S, shRNAs. Cancer Gene Therapy 2011; 18:
concept. The Journal of Clinical Investigation Inthasorn P. A pilot phase II study of cap- 219–227. DOI: 10.1038/cgt.2010.72.
2010; 120: 41. ecitabine plus cisplatin in the treatment of 264. Tan S, Hougardy BM, Meersma GJ, et al.
244. Catalano V, Di Franco S, Iovino F, Dieli F, recurrent carcinoma of the uterine cervix. Human papilloma virus 16 e6 RNA inter-
Stassi G, Todaro M. CD133 as a target for Oncology 2007; 72: 33–38. ference enhances cisplatin and death
colon cancer. Expert Opinion on Therapeutic 255. Errihani H, M’rabti H, Ismaili N, Inrhaoun receptor-mediated apoptosis in human
Targets 2012; 16: 259–267. H, Elghissassi I. Phase II trial of capecita- cervical carcinoma cells. Molecular
245. Tang AL, Owen JH, Hauff SJ, et al. Head bine and cisplatin in advanced, persistent, Pharmacology 2012; 81: 701–709. DOI:
and neck cancer stem cells the effect of or recurrent carcinoma of the cervix. Inter- mol.111.076539.
HPV—an in vitro and mouse study. Otolar- national Journal of Gynecological Cancer 265. Chen J, McMillan N, Gu W. Intra-tumor
yngology–Head and Neck Surgery 2013; 149: 2011; 21: 373–377. injection of lentiviral-vector delivered
252–260. 256. Ischenko I, Camaj P, Seeliger H, et al. In- shRNA targeting human papillomavirus
246. Qi W, Zhao C, Zhao L, et al. Sorting and hibition of Src tyrosine kinase reverts E6 and E7 oncogenes reduces tumor
identification of side population cells in chemoresistance toward 5-fluorouracil in growth in a xenograft cervical cancer
the human cervical cancer cell line HeLa. human pancreatic carcinoma cells: an in- model in mice. Journal of Solid Tumors
Cancer Cell International 2014; 14: 3. volvement of epidermal growth factor re- 2012; 2: 4–10.
247. Villanueva-Toledo J, Ponciano-Gómez A, ceptor signaling. Oncogene 2008; 27: 266. Zhou J, Peng C, Li B, et al. Transcrip-
Ortiz-Sánchez E, Garrido E. Side popula- 7212–7222. tional gene silencing of HPV16 E6/E7
tions from cervical-cancer-derived cell 257. Abdulkarim B, Sabri S, Deutsch E, et al. induces growth inhibition via apoptosis
lines have stem-cell-like properties. Molec- Antiviral agent cidofovir restores p53 in vitro and in vivo. Gynecologic Oncology
ular Biology Reports 2014; 41: 1993–2004. function and enhances the radiosensitivity 2012; 124: 296–302. DOI: 10.1016/j.
248. Zhang M, Kumar B, Piao L, et al. Elevated in HPV-associated cancers. Oncogene 2002; ygyno.2011.10.028.
intrinsic cancer stem cell population in hu- 21: 2334–2346. 267. Faried LS, Faried A, Kanuma T, et al. Pre-
man papillomavirus-associated head and 258. Tan S, de Vries EG, van der Zee AG, de dictive and prognostic role of activated
neck squamous cell carcinoma. Cancer Jong S. Anticancer drugs aimed at E6 and mammalian target of rapamycin in cervi-
2014; 120: 992–1001. E7 activity in HPV-positive cervical cancer. cal cancer treated with cisplatin-based
249. Movva S, Rodriguez L, Arias PH, Current Cancer Drug Targets 2012; 12: neoadjuvant chemotherapy. Oncology
Verschraegen C. Novel chemotherapy 170–184. DOI: BSP/CCDT/E-Pub/00181. Reports 2006; 16: 57–64.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 51

268. Chen J. The Src/PI3K/Akt signal path- squamous cell carcinoma, making the chemosensitivity in cervical cancer cells
way may play a key role in decreased EGFR pathway a novel therapeutic via induction of apoptosis. Life Sciences
drug efficacy in obesity-associated cancer. target. British Journal of Cancer 2011; 2013; 93: 7–16.
Journal of Cellular Biochemistry 2010; 110: 105: 420–427. 286. Yokoyama M, Noguchi M, Nakao Y, Pater
279–280. 278. Deberne M, Levy A, Mondini M, et al. The A, Iwasaka T. The tea polyphenol,( )-epi-
269. Cheng H, Walls M, M Baxi S, Yin M-J. combination of the antiviral agent gallocatechin gallate effects on growth, ap-
Targeting the mTOR pathway in tumor cidofovir and anti-EGFR antibody optosis, and telomerase activity in cervical
malignancy. Current Cancer Drug Targets cetuximab exerts an antiproliferative effect cell lines. Gynecologic Oncology 2004; 92:
2013; 13: 267–277. on HPV-positive cervical cancer cell lines’ 197–204.
270. Noh KH, Kang TH, Kim JH, et al. Activa- in-vitro and in-vivo xenografts. Anti-Can- 287. Ahn WS, Huh SW, Bae S-M, et al. A ma-
tion of Akt as a mechanism for tumor im- cer Drugs 2013; 24: 599–608. jor constituent of green tea, EGCG, in-
mune evasion. Molecular Therapy 2009; 17: 279. de Mello RA, Gerós S, Alves MP, Moreira hibits the growth of a human cervical
439–447. F, Avezedo I, Dinis J. Cetuximab plus cancer cell line, CaSki cells, through apo-
271. Rashmi R, DeSelm C, Helms C, et al. AKT platinum-based chemotherapy in head ptosis, G1 arrest, and regulation of gene
inhibitors promote cell death in cervical and neck squamous cell carcinoma: a ret- expression. DNA and Cell Biology 2003;
cancer through disruption of mTOR sig- rospective study in a single comprehen- 22: 217–224.
naling and glucose uptake. PloS One sive European Cancer Institution. PloS 288. Noguchi M, Yokoyama M, Watanabe S,
2014; 9: e92948. One 2014; 9: e86697. et al. Inhibitory effect of the tea polyphe-
272. Schwarz JK, Payton JE, Rashmi R, et al. 280. Farley J, Sill MW, Birrer M, et al. Phase II nol,( )-epigallocatechin gallate, on
Pathway-specific analysis of gene expres- study of cisplatin plus cetuximab in ad- growth of cervical adenocarcinoma cell
sion data identifies the PI3K/Akt pathway vanced, recurrent, and previously treated lines. Cancer Letters 2006; 234: 135–142.
as a novel therapeutic target in cervical cancers of the cervix and evaluation of epi- 289. Singh M, Singh R, Bhui K, Tyagi S,
cancer. Clinical Cancer Research 2012; 18: dermal growth factor receptor immuno- Mahmood Z, Shukla Y. Tea polyphenols
1464–1471. histochemical expression: a gynecologic induce apoptosis through mitochondrial
273. Liu Y, Cui B, Qiao Y, et al. oncology group study. Gynecologic Oncol- pathway and by inhibiting nuclear factor-
Phosphoinositide-3-kinase inhibition en- ogy 2011; 121: 303–308. κB and Akt activation in human cervical
hances radiosensitization of cervical can- 281. Goncalves A, Fabbro M, Lhomme C, et al. cancer cells. Oncology Research Featuring
cer in vivo. International Journal of A phase II trial to evaluate gefitinib as Preclinical and Clinical Cancer Therapeutics
Gynecological Cancer 2011; 21: 100–105. second-or third-line treatment in patients 2011; 19: 245–257.
274. Tinker A, Ellard S, Welch S, et al. Phase II with recurring locoregionally advanced 290. Maher DM, Bell MC, O’Donnell EA,
study of temsirolimus (CCI-779) in women or metastatic cervical cancer. Gynecologic Gupta BK, Jaggi M, Chauhan SC.
with recurrent, unresectable, locally ad- Oncology 2008; 108: 42–46. Curcumin suppresses human papilloma-
vanced or metastatic carcinoma of the cer- 282. Nogueira-Rodrigues A, Moralez G, virus oncoproteins, restores p53, rb, and
vix. A trial of the NCIC Clinical Trials Grazziotin R, et al. Phase 2 trial of ptpn13 proteins and inhibits benzo [a]
Group (NCIC CTG IND 199). Gynecologic erlotinib combined with cisplatin and pyrene-induced upregulation of HPV
Oncology 2013; 130: 269–274. radiotherapy in patients with locally ad- E7. Molecular Carcinogenesis 2011; 50:
275. Tewari KS, Sill MW, Long III HJ, et al. vanced cervical cancer. Cancer 2014; 120: 47–57.
Improved survival with bevacizumab 1187–1193. 291. Singh M, Singh N. Curcumin counteracts
in advanced cervical cancer. New En- 283. Callegaro-Filho D, Kavanagh JJ, Nick the proliferative effect of estradiol and in-
gland Journal of Medicine 2014; 370: AM, Ramirez PT, Schmeler KM. duces apoptosis in cervical cancer cells.
734–743. Sustained complete response after main- Molecular and Cellular Biochemistry 2011;
276. Schefter TE, Winter K, Kwon JS, et al. A tenance therapy with topotecan and 347: 1–11.
phase II study of bevacizumab in com- erlotinib for recurrent cervical cancer 292. Roy M, Mukherjee S. Reversal of resis-
bination with definitive radiotherapy with distant metastases. Case Reports in tance towards cisplatin by curcumin in
and cisplatin chemotherapy in un- Oncology 2014; 7: 97–101. cervical cancer cells. Asian Pacific Journal
treated patients with locally advanced 284. Nogueira-Rodrigues A, do Carmo CC, of Cancer Prevention: APJCP 2014; 15:
cervical carcinoma: preliminary results Viegas C, et al. Phase I trial of erlotinib 1403–1410.
of RTOG 0417. International Journal of combined with cisplatin and radiotherapy 293. Sreekanth C, Bava S, Sreekumar E, Anto R.
Radiation Oncology Biology Physics 2012; for patients with locally advanced cervical Molecular evidences for the
83: 1179–1184. squamous cell cancer. Clinical Cancer Re- chemosensitizing efficacy of liposomal
277. Iida K, Nakayama K, Rahman M, et al. search 2008; 14: 6324–6329. curcumin in paclitaxel chemotherapy in
EGFR gene amplification is related to 285. Singh M, Bhui K, Singh R, Shukla Y. mouse models of cervical cancer. Oncogene
adverse clinical outcomes in cervical Tea polyphenols enhance cisplatin 2011; 30: 3139–3152.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
52 J. Chen

294. Yoysungnoen-Chintana P, Bhattarakosol through interferon-γ. Clinical Cancer Research progression of cervical cancer in a human
P, Patumraj S. Antitumor and 2014; 20: 5456–5467. papillomavirus-transgenic mouse model.
antiangiogenic activities of curcumin in 303. Westermann C, Fischer A, Clad A. Treat- Proceedings of the National Academy of Sci-
cervical cancer xenografts in nude mice. ment of vulvar intraepithelial neoplasia ences of the United States of America 2005;
BioMed Research International 2014: 817972. with topical 5% imiquimod cream. Interna- 102: 2490–2495.
295. Debata PR, Castellanos MR, Fata JE, et al. tional Journal of Gynecology & Obstetrics 313. López-Romero R, Garrido-Guerrero E,
A novel curcumin-based vaginal cream 2013; 120: 266–270. Rangel-López A, et al. The cervical malig-
Vacurin selectively eliminates apposed hu- 304. Daayana S, Elkord E, Winters U, et al. nant cells display a down regulation of
man cervical cancer cells. Gynecologic On- Phase II trial of imiquimod and HPV ther- ER-α but retain the ER-β expression. Inter-
cology 2013; 129: 145–153. apeutic vaccination in patients with vulval national Journal of Clinical and Experimental
296. Vidya Priyadarsini R, Senthil Murugan R, intraepithelial neoplasia. British Journal of Pathology 2013; 6: 1594–1602.
Maitreyi S, Ramalingam K, Karunagaran Cancer 2010; 102: 1129–1136. 314. Kwasniewska A, Postawski K,
D, Nagini S. The flavonoid quercetin in- 305. Kenter GG, Welters MJ, Valentijn ARP, Gozdzicka-Jozefiak A, et al. Estrogen
duces cell cycle arrest and mitochondria- et al. Vaccination against HPV-16 and progesterone receptor expression in
mediated apoptosis in human cervical oncoproteins for vulvar intraepithelial HPV-positive and HPV-negative cervical
cancer (HeLa) cells through p53 induction neoplasia. New England Journal of Medicine carcinomas. Oncology Reports 2011; 26:
and NF-κB inhibition. European Journal of 2009; 361: 1838–1847. 153–160.
Pharmacology 2010; 649: 84–91. 306. Cheng W-F, Hung C-F, Chai C-Y, 315. Chung S-H, Shin MK, Korach KS, Lambert
297. Bishayee K, Ghosh S, Mukherjee A, et al. Tumor-specific immunity and PF. Requirement for stromal estrogen re-
Sadhukhan R, Mondal J, Khuda-Bukhsh antiangiogenesis generated by a DNA vac- ceptor alpha in cervical neoplasia. Hor-
A. Quercetin induces cytochrome-c release cine encoding calreticulin linked to a tu- mones and Cancer 2013; 4: 50–59.
and ROS accumulation to promote apo- mor antigen. Journal of Clinical 316. Cortés-Malagón EM, Bonilla-Delgado J,
ptosis and arrest the cell cycle in G2/M, Investigation 2001; 108: 669–678. Díaz-Chávez J, et al. Gene expression pro-
in cervical carcinoma: signal cascade and 307. Chuang C-M, Monie A, Hung C-F, Wu T. file regulated by the HPV16 E7
drug-DNA interaction. Cell Proliferation Research treatment with Imiquimod en- oncoprotein and estradiol in cervical tis-
2013; 46: 153–163. hances antitumor immunity induced by sue. Virology 2013; 447: 155–165.
298. Campo M, Graham S, Cortese M, et al. therapeutic HPV DNA vaccination. 2010. 317. Spurgeon ME, Chung S-H, Lambert PF.
HPV-16 E5 down-regulates expression of 308. Peralta-Zaragoza O, Bermúdez-Morales Recurrence of cervical cancer in mice after
surface HLA class I and reduces recogni- VH, Pérez-Plasencia C, Salazar-León J, selective estrogen receptor modulator
tion by CD8 T cells. Virology 2010; 407: Gómez-Cerón C, Madrid-Marina V. therapy. The American Journal of Pathology
137–142. Targeted treatments for cervical cancer: 2014; 184: 530–540.
299. Zhou F, Chen J, Zhao K-N. Human papil- a review. OncoTargets and Therapy 2012; 318. Munger K. Are selective estrogen receptor
lomavirus 16-encoded E7 protein inhibits 5: 315. modulators (SERMs) a therapeutic option
IFN-γ-mediated MHC class I antigen pre- 309. Persson I, Yuen J, Bergkvist L, Schairer C. for HPV-associated cervical lesions and
sentation and CTL-induced lysis by Cancer incidence and mortality in women cancers? The American Journal of Pathology
blocking IRF-1 expression in mouse receiving estrogen and estrogen-progestin 2014; 184: 358–361.
keratinocytes. Journal of General Virology replacement therapy—long-term follow- 319. Dhandayuthapani S, Marimuthu P,
2013; 94: 2504–2514. up of a Swedish cohort. International Jour- Hörmann V, Kumi-Diaka J, Rathinavelu
300. Grimm C, Polterauer S, Natter C, et al. nal of Cancer 1996; 67: 327–332. A. Induction of apoptosis in HeLa cells
Treatment of cervical intraepithelial neo- 310. Arbeit JM, Howley PM, Hanahan D. via caspase activation by resveratrol and
plasia with topical imiquimod: a random- Chronic estrogen-induced cervical and genistein. Journal of Medicinal Food 2013;
ized controlled trial. Obstetrics & vaginal squamous carcinogenesis in hu- 16: 139–146.
Gynecology 2012; 120: 152–159. man papillomavirus type 16 transgenic 320. Papazisis KT, Kalemi TG, Zambouli D,
301. Terlou A, Seters Mv, Kleinjan A, et al. mice. Proceedings of the National Academy et al. Synergistic effects of protein tyro-
Imiquimod-induced clearance of HPV is of Sciences 1996; 93: 2930–2935. sine kinase inhibitor genistein with
associated with normalization of immune 311. Riley RR, Duensing S, Brake T, Münger K, camptothecins against three cell lines
cell counts in usual type vulvar Lambert PF, Arbeit JM. Dissection of hu- in vitro. Cancer Letters 2006; 233:
intraepithelial neoplasia. International Jour- man papillomavirus E6 and E7 function 255–264.
nal of Cancer 2010; 127: 2831–2840. in transgenic mouse models of cervical car- 321. Sahin K, Tuzcu M, Basak N, et al. Sensitiza-
302. Soong RS, Song L, Trieu J, et al. Toll like re- cinogenesis. Cancer Research 2003; 63: tion of cervical cancer cells to cisplatin by
ceptor agonist imiquimod facilitates 4862–4871. genistein: the role of NF B and
antigen-specific CD8+ T cell accumulation 312. Brake T, Lambert PF. Estrogen contributes Akt/mTOR signaling pathways. Journal
in the genital tract leading to tumor control to the onset, persistence, and malignant of Oncology 2012: 461562.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv
Signaling pathways in HPV-associated cancers 53

322. Kim SH, Kim SH, Kim YB, Jeon YT, Lee 328. Nguyen HH, Aronchik I, Brar GA, Nguyen pathway in breast cancer stem-like cells is
SC, Song YS. Genistein inhibits cell growth DH, Bjeldanes LF, Firestone GL. The die- required for viability and maintenance.
by modulating various mitogen-activated tary phytochemical indole-3-carbinol is a Proceedings of the National Academy of Sci-
protein kinases and AKT in cervical cancer natural elastase enzymatic inhibitor that ences 2007; 104: 16158–16163.
cells. Annals of the New York Academy of Sci- disrupts cyclin E protein processing. Pro- 334. Bleau A-M, Hambardzumyan D, Ozawa
ences 2009; 1171: 495–500. ceedings of the National Academy of Sciences T, et al. PTEN/PI3K/Akt pathway regu-
323. Hussain A, Harish G, Prabhu SA, et al. 2008; 105: 19750–19755. lates the side population phenotype and
Inhibitory effect of genistein on the in- 329. Luo X, Dong Z, Chen Y, Yang L, Lai D. En- ABCG2 activity in glioma tumor stem-
vasive potential of human cervical can- richment of ovarian cancer stem-like cells like cells. Cell Stem Cell 2009; 4:
cer cells via modulation of matrix is associated with epithelial to mesenchy- 226–235.
metalloproteinase-9 and tissue inhibitors mal transition through an miRNA- 335. Dubrovska A, Elliott J, Salamone RJ,
of matrix metalloproteinase-1 expression. activated AKT pathway. Cell Proliferation et al. Combination therapy targeting
Cancer Epidemiology 2012; 36: e387–e393. 2013; 46: 436–446. both tumor-initiating and differentiated
324. Kim SH, Kim SH, Lee SC, Song YS. In- 330. He K, Xu T, Xu Y, Ring A, Kahn M, cell populations in prostate carcinoma.
volvement of both extrinsic and intrinsic Goldkorn A. Cancer cells acquire a drug Clinical Cancer Research 2010; 16:
apoptotic pathways in apoptosis induced resistant, highly tumorigenic, cancer 5692–5702.
by genistein in human cervical cancer stem-like phenotype through modulation 336. Hart LS, Dolloff NG, Dicker DT, et al. Hu-
cells. Annals of the New York Academy of Sci- of the PI3K/Akt/β-catenin/CBP pathway. man colon cancer stem cells are enriched
ences 2009; 1171: 196–201. International Journal of Cancer 2014; 134: by insulin-like growth factor-1 and are
325. Jin L, Qi M, Chen D-Z, et al. Indole-3-carbi- 43–54. sensitive to figitumumab. Cell Cycle 2011;
nol prevents cervical cancer in human 331. Wei Y, Jiang Y, Zou F, et al. Activation of 10: 2331–2338.
papilloma virus type 16 (HPV16) trans- PI3K/Akt pathway by CD133-p85 inter- 337. Korkaya H, Paulson A, Charafe-Jauffret E,
genic mice. Cancer Research 1999; 59: action promotes tumorigenic capacity of et al. Regulation of mammary
3991–3997. glioma stem cells. Proceedings of the Na- stem/progenitor cells by PTEN/Akt/β-ca-
326. Chen D-Z, Qi M, Auborn KJ, Carter TH. tional Academy of Sciences 2013; 110: tenin signaling. PLoS Biology 2009; 7:
Indole-3-carbinol and diindolylmethane 6829–6834. e1000121.
induce apoptosis of human cervical can- 332. Singh BN, Kumar D, Shankar S, 338. Kreso A, van Galen P, Pedley NM, et al.
cer cells and in murine HPV16- Srivastava RK. Rottlerin induces autoph- Self-renewal as a therapeutic target in hu-
transgenic preneoplastic cervical epithe- agy which leads to apoptotic cell death man colorectal cancer. Nature Medicine
lium. The Journal of Nutrition 2001; 131: through inhibition of PI3K/Akt/mTOR 2014; 20: 29–36.
3294–3302. pathway in human pancreatic cancer stem 339. Jung YS, Vermeer PD, Vermeer DW, et al.
327. Qi M, Anderson AE, Chen D-Z, Sun S, cells. Biochemical Pharmacology 2012; 84: CD200: Association with cancer stem cell
Auborn KJ. Indole-3-carbinol prevents 1154–1163. features and response to chemoradiation in
PTEN loss in cervical cancer in vivo. Molec- 333. Zhou J, Wulfkuhle J, Zhang H, et al. Acti- head and neck squamous cell carcinoma.
ular Medicine 2005; 11: 59–63. vation of the PTEN/mTOR/STAT3 Head & Neck 2014. DOI: 10.1002/hed.23608.

Copyright © 2015 John Wiley & Sons, Ltd. Rev. Med. Virol. 2015; 25: 24–53.
DOI: 10.1002/rmv

You might also like