Subido por Wen Cartagena

5 cancer cervicouterino

Anuncio
Molecular Biology Reports
https://doi.org/10.1007/s11033-020-05503-6
REVIEW
Cervical cancer and potential pharmacological treatment with snake
venoms
Alejandro Montoya‑Gómez1 · Leonel Montealegre‑Sánchez1
Eliécer Jiménez‑Charris1
· Herney Andrés García‑Perdomo2
·
Received: 27 November 2019 / Accepted: 6 May 2020
© Springer Nature B.V. 2020
Abstract
Cervical cancer is the fourth most common cancer worldwide in women. Apoptosis reactivation has become the main strat‑
egy for decreasing cancer proliferation. There is a need to extend the search for new drugs to implement more effective and
less toxic strategies for cervical cancer treatment. Research has been carried out to find new drugs that have minimal side
effects and that focus on the tumor microenvironment, particularly in the induction of cellular apoptosis and cell migration
and the inhibition of angiogenesis. Potent toxins from snake venoms have shown potential as sources for the synthesis of
new drugs with such characteristics. The present work aimed to describe cervical cancer characteristics, associated risk
factors, current treatments and to highlight the effects of toxins isolated from the venom of snakes of the Viperidae family
on cervical cancer cell lines.
Keywords Anticancer agent · Toxins · Cytotoxicity · Apoptosis · Tumor cell lines · Translational medicine
Introduction
Cervical cancer (CC) is considered a global public health
problem. In 2018, 569,847 new cases of CC were reported
worldwide, and approximately 311,365 deaths were directly
attributable to this type of tumorigenic disease [1]. In
Colombia, according to the Cali Cancer Registry, 8963 cases
of CC were registered from 1962 to 2007. Between 2007 and
2011, there were a total of 4462 new CC reports and 1861
deaths [2]. The mortality rate standardized by age between
2009 and 2011 was 7 per 100,000 women [3]. Therefore,
the high morbidity associated with this cancer type gener‑
ates a significant socioeconomic impact on the country. The
previous observations pose a challenge for the health system
that highlights a need to design and implement management
* Alejandro Montoya‑Gómez
[email protected]
* Eliécer Jiménez‑Charris
[email protected]
1
Grupo de Nutrición, Facultad de Salud, Universidad del
Valle, Calle 4B # 36–00, Edificio 116, Oficina 5002, Cali,
Colombia
2
Departamento de Cirugía/Urología, Escuela de Medicina,
Universidad del Valle, Cali, Colombia
policies to reduce the incidence of and mortality from this
cancer.
Pelvic radiotherapy (PR), chemotherapy (CWCC), and
brachytherapy are the most common interventions to treat
CC patients [4]. In general, 50% to 60% of patients achieve a
cure with this approach, but local recurrence and tumor pro‑
gression to metastasis after treatment are frequent outcomes,
especially in patients with late diagnosis and advanced stage
tumors. In that sense, there are few effective treatments for
patients with recurrent CC. Deaths often occur after patient
withdrawal from this kind of treatment to avoid a significant
decrease in their quality of life due to severe pain, bleeding,
and other debilitating symptoms. Currently, efforts are being
made to improve the healing potential of PR with more pre‑
cise tumor targeting and intensification [5]. However, it is
unlikely that this approach alone will solve the problem [6].
Chemotherapy is not specific and can affect both tumor
and healthy cells [7]. There are medications and alternative
treatments used to counteract the adverse effects induced
by these drugs. However, this strategy is not sufficient or
may even induce other side effects that add discomfort to
patients [8]. In that sense, until now, no drugs have been
developed that attack cervical carcinoma cells selectively
without affecting healthy tissue viability. Some reviews
have highlighted studies on molecules isolated from snake
13
Vol.:(0123456789)
Molecular Biology Reports
venoms and their effects on different types of cancer cells
[9–11]. Nonetheless, there are no reviews that describe the
effects of these molecules on CC cells. Thus, the purpose
of this manuscript was to describe bioprospecting studies
of snake venom toxins with potential use in CC treatment.
Cervical cancer epidemiology
Cancer is one of the leading causes of nonviolent death,
both in developing countries and in developed countries.
Lung, prostate, colorectal, stomach, and liver cancer are the
most common cancer types in men, while breast, colorectal,
lung, cervix and thyroid cancer are the most common among
women [12]. Globally, there were 530,000 cervical cancer
cases in 2012, with 85% of these occurring in less developed
regions [13]. Approximately 11.5 million people will die of
cancer in 2030 [14].
Cervical cancer originates in the cervix epithelium (squa‑
mocolumnar junction that may include external squamous
Fig. 1 Chronological progression of cervical cancer. Genetic and epi‑
genetic factors modify basal cells in the cervical epithelium. Dyspla‑
sia can progress from mild to severe, in which case it is recognized as
a precursor lesion. The late stage, known as carcinoma in situ (CIS)
or cervical intraepithelial neoplasia grade 3 (CIN III), is characterized
13
cells, internal glandular cells, or both) with cervical intraepi‑
thelial neoplasia grade 2 or 3 (CIN II or CIN III) as a precur‑
sor lesion (Fig. 1). CIN and carcinoma in situ (CIS) can turn
into invasive cancer [15]. Precursor lesions are the initial
manifestation. They usually exhibit slow and progressive
evolution over time, which usually occurs in stages. Lesions
evolve to cancer in situ when they only involve the epithe‑
lial surface and then to invasive cancer when the compro‑
mised tissue crosses the basement membrane and invades
the stroma of the cervix [16].
The classification of precancers and CC is performed
according to the appearance of the tissues under the micro‑
scope. The two most common CC types are squamous cell
carcinoma and adenocarcinoma [17]. Most CCs are squa‑
mous cell carcinomas (9 out of 10 cases). They originate
from cells in the ectocervix, most frequently in the transfor‑
mation zone (where the ectocervix joins the endocervix).
The rest of the CCs are adenocarcinomas, which originate
from glandular cells (the endocervix). Commonly, earlystage CC does not manifest signs or apparent symptoms.
by induction of angiogenesis and recruitment of immune cells. In this
stage, there is a transformation to invasive cancer with alterations in
the surrounding cells and an increase in angiogenesis and immune
cell recruitment processes
Molecular Biology Reports
However, in more advanced stages, vaginal bleeding, unu‑
sual vaginal discharge, pelvic pain, dyspareunia, and post‑
coital hemorrhage may occur [18]. When detected early,
treatment is relatively simple and effective [19]. Therefore,
the higher social risk for the acquisition of potentially fatal
disease derives from the lack of periodic gynecological
examinations and Papanicolaou exams [15, 20].
All types of solid tumors, such as CC, require the acti‑
vation of the angiogenesis process, which is necessary to
receive oxygen and nutrients supplying their metabolic
requirements. The progression of cervical intraepithelial
neoplastic lesions to invasive carcinoma is associated with
angiogenesis. The previous study showed an increase in the
density and size of the vessels in the stroma-epithelial cervi‑
cal contact zone (Fig. 1). Angiogenesis promotes the overex‑
pression of vascular endothelial growth factors (VEGF) and
the expression of extracellular proteases, including matrix
metalloproteinase 9 (MMP-9) [21, 22]. Smith-McCune and
Weidner [23] showed that neovascularization occurs in a
series of steps that facilitate the diffusion of proangiogenic
compounds towards dysplastic tissue. For this reason, it is
necessary to find molecules that affect tumor proliferation
not only aimed at reactivating the apoptotic process but also
aimed at altering the proangiogenic potential of cancer cells.
Human Papillomavirus infection and other
risk factors
The main risk factor for CC is infection by human papil‑
lomavirus (HPV), outperforming other known risk factors
[24–27]. Transient HPV infection is common in young
women, and its persistence leads to an increased risk of pre‑
cancerous and cancerous lesions [28–30]. Currently, more
than 200 types of HPV viruses have been recognized on the
basis of their DNA sequences [31]. Certain types of HPV
can cause warts on or around the female and male genital
organs, as well as in the anal area. Based on an association
with precursor lesions, HPVs can be classified as low-risk
and high-risk. Low-risk HPV includes types, but is not lim‑
ited to 6, 11, 42, 43, and 44. High-risk HPV includes types
such as 16, 18, 31, 33, 35, which are strongly linked to cervi‑
cal intraepithelial neoplasia and CC [31].
Epidemiological evidence links more than 50 types of
HPV that infect the genital tract. Types 16 and 18 are the
most closely related to the presentation of high-grade dys‑
plasia and CC. HPV type 18 is the most commonly associ‑
ated with cervical adenocarcinoma, while HPV 16, followed
by HPV 18, is the most frequent types detected in squamous
cell carcinoma [32, 33]. The presence of integrated forms
of HPV 16 increases with the severity of cervical neoplasia,
although in some women with the invasive disease, episomal
forms of this type have been detected. In contrast, integrated
forms of HPV 18 are almost always associated with highgrade cervical intraepithelial neoplasia (HGCIN) and inva‑
sive disease [34]. Aguayo et al. described two conditions
for the conversion to malignant tumors: the interruption of
intracellular and extracellular pathways generated by the
presence of HPV and the expression of oncoproteins for the
maintenance of the malignant phenotype [35].
The viral DNA of HPV-16 contains eight open reading
frames (ORFs) and a regulatory region called the long con‑
trol region (LCR). It contains an origin of replication and
transcriptional regulatory elements [36]. The first region
after LCR contains 6 ORFs corresponding to the E6, E7,
E1, E2, E4, and E5 genes, which encode proteins necessary
for virus replication and cell transformation. The last two
regions code for viral capsid proteins: L1, the main struc‑
tural protein, and L2, the binding protein for DNA encapsu‑
lation [37]. In terms of integration, HPV (mainly HPV-16)
probably has first contact with the cell, through the polysac‑
charide heparan sulfate and then enters the cell mediated
by receptors such as alpha-6 integrin [38]. Once the virus
integrates, there is a disruption of the synthesis of the E2
protein, which is a repressor of the p97 promoter; this causes
overexpression of the E6 and E7 proteins, which induces
the loss of P53 and pRb proteins. In cervical carcinogen‑
esis, HPV integration in basal cells generates deregulation
of the expression of these two viral oncogenes. This kind of
deregulation is considered a critical event for the progression
towards the disease [35].
Another factor strongly implicated in the development
of cervical cancer is the sensitivity of the tissue to estrogen
[39–42]. The transformation zone is a highly estrogen-sen‑
sitive area of the genital tract, where most HPV injuries and
their sequelae occur. This contrasts with the fact that there
are other areas of the genital tract with low estrogen sensitiv‑
ity that, when infected with HPV, show equal or higher loads
of HPV without the development of a neoplasm [43, 44].
These observations suggest that this hormone plays a vital
role in CC development, and there is a dynamic amplifica‑
tion of HPV and estrogen effects. In the transformation zone,
16 α-hydroxyestrone metabolites prolong estrogen effects.
This compound is genotoxic, and it comes from the conver‑
sion of estradiol. This conversion increases in the presence
of HPV-16 [42]. Estrogen stimulates the expression of onco‑
genes in HPV-infected cervical cell lines [45]. The previ‑
ous affirmation strengthens the hypothesis of the synergistic
effect of both factors in the development of cervical cancer.
This information is supported by studies showing a role
for the estrogen receptor Erα in the development of CC in
murine models and by other studies showing the relationship
between increased expression of the E6 and E7 oncogenes
and the presence of estrogen [41]. The role of the receptor
has been analyzed in other studies, and it has been found
that its expression is relatively constant in tumor-associated
13
Molecular Biology Reports
stromal fibroblastic cells, with which it is possible that the
progression of CC is stimulated [40]. Regarding the type of
HPV most likely to cause CC in the presence of estrogen,
it was found in a recent study that HPV-16 is more strongly
associated with the development of CC than other types of
HPV; in addition, it has an additive effect with high endog‑
enous estradiol levels that leads to an increase the risk of
developing CC [39].
In addition, other risk factors, such as smoking [46], pro‑
longed use of oral contraceptives [47, 48], a high number of
full-term pregnancies [49], early age at the first intercourse, a
high number of sexual partners [50], nutritional status, immu‑
nosuppression [51, 52], and exposure to diethylstilbestrol in
the uterus [53], have been shown to be related to CC.
Cervical cancer interventions
and treatments
The pathophysiology of CC involves very long periods, and
when detected early, there is ample opportunity for action,
making its prevention or cure possible. Preinvasive lesions,
which in most cases present mild dysplasia, should be moni‑
tored rather than treated because spontaneous regressions
are very common (Fig. 1). Treatment schemes are defined
according to age and can include control tests such as colpos‑
copy [19, 20, 54, 55]. A specialist colposcopist or gynecolo‑
gist should assess HPV-related disturbances in pap smears
to make a definitive diagnosis and define the intervention
[19, 20, 54, 55].
Currently, there are two types of procedures: ablation
and excision. Cryotherapy is the most widely used method
of ablation and involves the application of low tempera‑
tures to reach a freezing point that will lead to cell death.
It is a 90% effective method, but the main limitation is the
lack of a biological sample for histopathological studies
[19, 20, 54, 55]. For the excision procedure, the electrosur‑
gical technique uses a fine electric wire loop to completely
remove the affected neoplastic area of the cervix. It is 95%
effective for high-grade dysplasia (large and endocervical
lesions). Additionally, it allows for obtaining histologi‑
cal samples for staging cancer [19, 20, 54, 55]. Clinicians
treat patients according to the tumor classification, general
health conditions of the patient, stage of the disease, and
the decision of the patient [20]. Radiotherapy, chemother‑
apy, palliative care, pain therapy, and rehabilitation are
among the nonsurgical therapies currently used to treat
CC, all necessary for the comprehensive care of women
affected by CC [56].
13
Side effects of nonsurgical treatments used
in CC
Radiation therapy is a treatment that uses high doses of
radiation to kill cancer cells and shrink tumors. External
radiation can be used alone to treat areas of spread cancer
or as a primary treatment for CC in patients who cannot
tolerate chemoradiation. Common side effects of external
radiation therapy include fatigue, stomach problems, diar‑
rhea or soft stools, nausea and vomiting, and skin changes
[15, 17]. Pelvis radiation can also irritate the bladder,
causing discomfort and frequent urges to urinate (radiation
cystitis). Radiation can affect the vulva and vagina, caus‑
ing them to be tender and painful and sometimes causing
bloody discharge. It can also affect the ovaries, causing
changes in menstrual periods and even early menopause.
In addition, radiation can cause low blood counts, which
can lead to anemia and leukopenia, increasing the risks
of serious infections. Blood counts are usually decreased
when patients are given chemotherapy with radiation.
Additionally, fatigue and nausea tend to worsen [15, 17].
Another type of radiation therapy is called brachytherapy
or internal radiation therapy. This intervention is offered as
adjuvant therapy for the treatment of CC. In brachytherapy,
radiation only travels a short distance, so the main effects of
radiation are on the cervix and the walls of the vagina. The
most common side effect is irritation of the vagina, which
becomes flushed and sensitive to pain. Brachytherapy can
also cause many of the same side effects caused by external
radiation [17]. Vaginal dryness and pain during intercourse
can be long-term side effects of radiation (both brachyther‑
apy and external radiation). In addition, pelvic radiation can
also weaken the bones and cause increased susceptibility to
fractures. Hip fractures are the most common and may occur
2 to 4 years after radiation [17].
Drugs currently used in chemotherapy destroy cancer cells
but also cause damage to normal cells, which can result in the
appearance of undesirable effects. These side effects depend
on the period of exposure, type, and dosage of medications
administered. Some common side effects of chemotherapy may
include nausea and vomiting, loss of appetite, hair loss, mouth
ulcers, and tiredness. In addition, blood cell counts could
decrease; therefore, the probability of infections increases.
There is a shortage of platelets; then, blood loss or bruising
can occur after minor cuts or injuries. Additionally, dyspnea
can appear due to the decrease in red blood cell levels [15, 17].
When given chemo- and radiation therapies, side effects
such as nausea, fatigue, and diarrheas are often more
severe than with either treatment alone [15, 17]. Prema‑
ture menopause and infertility can also occur permanently.
Molecular Biology Reports
In addition, medications such as paclitaxel and cisplatin
can produce nerve damage outside the brain and spinal
cord [57]. This condition, called peripheral neuropathy,
sometimes causes symptoms mainly in the feet and hands,
such as numbness, pain, burning, tingling, sensitivity to
cold or heat, and weakness. In most cases, these symptoms
are relieved or can disappear once the treatment finishes,
but in some women, they may persist for a long time [15].
Until now, there have been no strategies involving chemoor radiotherapies that do not involve any side effects in
patients receiving these types of treatments. Therefore,
the search for alternative antitumor molecules with greater
specificity against CC is required. In that sense, natural
sources of pharmacologically active molecules such as
snake venoms are promising areas for drug discovery.
Finding new treatments will help to fight against this type
of pathophysiological problem that affects the female
population.
Snake venoms
Snakes are reptiles without limbs and, thus, have developed
diverse strategies to obtain their food and defend themselves
against possible predators. Some of them developed huge
muscles to constrict their prey. Others developed venom
glands and hollow fangs to inoculate biomolecules that
can paralyze and digest their prey. Snake venoms contain
hundreds of pharmacologically active molecules, including
organic and mineral components (histamine and other aller‑
gens, alkaloids, polyamines), small peptides, and proteins
[58, 59]. Although venoms of the Elapidae and Viperidae
family may contain more than 100 protein components,
these proteins belong to a few large families of proteins
with enzymatic activity (­ Zn2+-dependent metalloproteases,
serine proteases, type II phospholipases ­A2, l-amino acid
oxidase) and proteins without enzymatic activity, such as
natriuretic peptides, disintegrins, inhibitors of Kunitz type
proteases, cystatin, type C lectins, vascular growth factors
and cysteine-rich secretory proteins (CRISPs) [60].
The biological effects of snake venoms are complex
since every component has different actions and can act in
concert, generating significant pathophysiological effects.
Venoms can be classified as neurotoxic or hemorrhagic/
myotoxic, according to the main effects on animals. In the
first group are the venoms of the Elapidae family, which
possess a wide variety of enzymes of phospholipase type ­A2
­(PLA2). These isoenzymes show different biological effects,
mainly characterized by presynaptic and postsynaptic neu‑
rotoxic activity, cardiotoxicity, and hypotensive, convulsive,
and edema-inducing activities [58]. Snake venoms of the
Viperidae family contain proteins that cause myotoxicity
and interfere with the hemostatic and tissue repair systems
by inhibiting platelet aggregation and causing hemorrhages.
Consequently, envenomation by these snakes usually pro‑
duces persistent bleeding [61].
To develop studies that explore the effects of snake
venom components on tumor cells, at least three experi‑
mental phases are required (Fig. 2) [62]. The first phase
involves venom extraction and fractionation. The latter is
performed by diverse techniques, such as reverse-phase
high-performance liquid chromatography (HPLC). Then,
proteomic analysis can be applied to assign the molecules
to known protein families by a combination of techniques
such as SDS-PAGE, mass spectrometry, and N-terminal
sequencing. Once the isolated and characterized molecules
are available, it is possible to develop the second experi‑
mental phase. It consists of determining whether the mol‑
ecule of interest has cytotoxic effects on tumor and nontu‑
mor cells. Some studies include an in silico phase, in which
a three-dimensional model of the molecule of interest is
obtained mainly by homology modeling. Subsequently, this
process evaluates the interaction of the molecule of inter‑
est with three-dimensional models of receptors and other
protein types and molecules of biological interest available
in structural databases. The third experimental phase con‑
sists of determining the possible mechanisms of cytotoxicity
induced by the molecule of interest on cancer cells. The use
of immunological techniques, flow cytometry, and confocal
microscopy helps to determine mechanisms of cell death
and morphological alterations in tumor cells. In addition, it
is important to determine the ability of proteins to alter the
metastatic ability of these cells by inhibiting proliferation,
adhesion, migration, invasion, or new vessel formation. Cell
migration and Matrigel invasion assays, as well as wound
healing and proangiogenesis assays, are among the most
common in vitro tests used in this phase [62].
Snake venom and its effect on CC cells
Despite the toxicity of snake venoms, there are also many
reports of their antitumor effects against cervical cancer cell
lines (Table 1). For example, in a study carried out in Bra‑
zil, the effect of the whole venom of two species of Bothrops on cervical cell lines, SiHa and HeLa, was evaluated.
The results revealed that venoms from both snakes induced
apoptosis in both cell lines. The authors observed cell cycle
arrest and depolarization of the mitochondrial membrane
in the G0/G1 phase in experiments with HeLa cells, a cell
line representative of the most common type of CC [7]. In a
study conducted in Korea, the total venom of Vipera lebetina
turanica inhibited the proliferation of CaSki cells, a line of
squamous epithelial carcinoma, and C33A cells (adenocar‑
cinoma cell line) in a dose-dependent manner. The venom
induced apoptosis by increasing the expression of DR3 and
13
Molecular Biology Reports
Fig. 2 The experimental design of Jiménez-Charris et al. [62, 79, 80]
was applied to purify and identify Pllans-II, an acidic phospholipase
­A2, and to determine its anticancer effects on cervical adenocarci‑
noma cells. In the first phase of the study, snake venom was obtained
by manual extraction (a), the protein cocktail was separated by RPHPLC (b), and the abundance of each family of proteins was deter‑
mined (c). In the second phase, they evaluated the cytotoxic activity
of Pllans-II on nontumor cells such as C2C12 myoblasts (d), and a
three-dimensional model was constructed based on the amino acid
sequence (e). In the third phase, the ability of Pllans-II to induce
apoptosis in HeLa cells was determined by an annexin-V/propidium
iodide assay (f), and the formation of apoptotic bodies was corrobo‑
rated by scanning electronic microscopy (g). In addition, to investi‑
gate the ability of Pllans-II to inhibit new vessel formation, its effect
on HUVECs was evaluated by a Matrigel tube formation assay (h)
DR5, receptors for tumor necrosis factor, as well as the
expression of the proapoptotic proteins Bax and caspase-3
(cleaved form), -8 and -9. A decrease in the expression of the
antiapoptotic proteins Bcl-2, CIAP-1, and XIAP, evidenced
by the exposure of cell lines with different doses of venom,
accompanied previous findings [63]. The transcription fac‑
tor NF-κB and the p50 nuclear subunit, normally expressed
in greater amounts in tumor tissues and CC cell lines than
in healthy cervical tissues, were also affected by treatment
with V. lebetina turanica venom in both cell lines. It inhib‑
ited the phosphorylated form of IκB in the cytoplasm and
the nuclear expression of subunits p50 and p65; therefore,
NF-κB bound to DNA in both cell lines. This finding could
explain the overexpression of proapoptotic proteins and the
subexpression of anti-apoptotic proteins.
Regarding how V. lebetina turanica toxins block NF-κB,
studies have shown that snake venom toxins can bind to
sulfhydryl groups of the p50 subunit or IKKs, inhibiting
the activity of NF-κB in other cancerogenic lines [64, 65].
Finally, the antitumor activity of the venom was evaluated
in vivo in a xenograft mouse model subcutaneously trans‑
planted with CaSki cells. The authors found a 30–40%
decrease in tumor mass and overexpression of DR3, DR5,
and caspase-3, -8, and -9 in venom-treated mice compared
to control mice. Additionally, inhibition of NF-κB, which
was in concordance with the results obtained in cell lines.
These results regarding the in vitro and in vivo down‑
regulation of NF-κB and overexpression of DR3 and DR5
receptors induced by V. lebetina turanica venom could
13
Molecular Biology Reports
Table 1 Effect of snake venoms on cervical cancer cells
Snake
Cervical cancer cell line
Principal effects on cervical cancer cells
Bothrops jararaca and
Bothrops erythromelas
SiHa and HeLa
In SiHa: Cytotoxicity
[7]
In HeLa: arrest in the G0/G1 phase of the cell cycle, and depolarization
of the mitochondrial membrane
Dose-dependent toxic effect on both cell lines
[63]
Increase in the expression of the receptors for tumor necrosis factor
DR3 and DR5
Increase in the expression of the pro-apoptotic proteins Bax, caspases 3
[cleaved form], 8 and 9
Decrease in the expression of the antiapoptotic proteins Bcl-2, CIAP-1,
XIAP
Inhibition of the phosphorylated form of IκB in the cytoplasm, as well
as the nuclear expression of the subunits p50 and p65, and therefore,
blocking the binding of NF-κB to DNA
Decrease between 30 and 40% of the tumor mass
[63]
Overexpression of receptors DR3, DR5, and in Caspase proteins 3, 8, 9
in tumor tissue
Inhibition of NF- κB in tumor tissue
Vipera lebetina turanica CaSki and C33A
Vipera lebetina turanica Xenographic mice with CaSki
cells subcutaneously trans‑
planted
indicate potential targets for the development of new treat‑
ments for CC, especially in resistant tumors.
Antitumor effects of peptides on cervical
cancer cells
Because of their small size, specificity, selectivity, and sta‑
bility, peptides have structural and pharmacological char‑
acteristics that make them very interesting in biomedical
research. Peptides isolated from snake venoms are rich in
disulfide bonds and have a higher affinity for other molecules
than do synthetic peptides. However, the use of peptides iso‑
lated from snake venoms is an emerging area of study [10].
Only four studies have explored the effect of peptides or
molecules derived from snake venom on CC cells (Table 2).
In 1994, Sheu et al. isolated a peptide containing the RGD
domain from the venom of Trimeresurus flavoviridis (later
named Protobothrops flavoviridis) [66]. This peptide, named
References
triflavin, inhibited the binding of HeLa cells to fibronectin,
fibrinogen, vitronectin, and other extracellular matrix com‑
ponents in a dose-dependent manner, with greater affinity
than a synthetic peptide with the RGD motif. Additionally,
it also inhibited the binding to laminin, collagen type I and
type IV, but to a lesser extent. Integrins such as α5β1 and
αVβ3, used by cells to adhere to fibronectin and vitronectin
of the extracellular matrix, respectively, were identified as
potential targets of triflavin. However, the cytotoxicity tests
showed that triflavin did not affect the viability of the cells,
which suggests that the inhibitory effect on adhesion does
not imply cytotoxicity.
Zare and Sarzaeem reported the effect on HeLa cells of
the peptide ICD-85, generated from the venoms of the Ira‑
nian snake Agkistrodon halys and the scorpion Hemiscorpius
lepturus [67]. This peptide showed a dose-dependent cyto‑
toxic effect on the HeLa cell line and a nonsignificant effect
on the viability of nontumor lung cells when the dose was
less than 50 µg/mL. This cytotoxic effect was related to the
Table 2 Effects of peptides and peptide derivatives from snake venoms on cervical cancer cells
Peptide
Snake
Cervical
cancer cell
line
Principal effects on cervical cancer cells
References
Triflavin
Protobothrops flavoviridis
HeLa
[66]
ICD-85
Agkistrodon halys
HeLa
Nanoparticles Agkistrodon halys
of ICD-85
HeLa
Inhibition of cellular binding to extracellular matrix components,
without cytotoxicity
Cytotoxicity related to induction of apoptosis
Increase in levels of caspase-8 protein expression
Greater cytotoxicity than for ICD-85, related to induction of apoptosis
Increase in levels of caspase-8 protein expression
Less damage to the cytoplasmic membrane than for ICD-85 peptide
[67]
[68]
13
Molecular Biology Reports
Effects of isolated proteins on cervical
cancer cells
mitochondrial membrane permeabilizes and cytochrome C
is released. Additionally, the induction of apoptosis in HeLa
cells was accompanied by an increase in reactive oxygen
species (ROS) and the translocation of the Bad and Bax
proapoptotic proteins from the cytosol to the mitochondria,
promoting the release of apoptogenic mitochondrial pro‑
teins. Coimmunoprecipitation assays showed that there was
an interaction between the Bcl-xL and Bad proteins and a
reduction in the binding between Bcl-xL and Bak in HeLa
cells treated with ACTX-8. The increase in the interaction
between Bcl-xL and Bad blocked the antiapoptotic signal
promoted by Bcl-xL, while the reduction in the interaction
between Bcl-xL and Bak allowed Bak to participate in the
permeabilization of the mitochondrial membrane, promot‑
ing apoptosis.
Texeira et al. reported the characterization of an LAAO
(LAAOcdt) isolated from the venom of Crotalus durissus
terrificus [72]. The authors evaluated the cytotoxic effect
of the protein on nine tumor cell lines, including HeLa and
SiHa. LAAOcdt resulted in more cytotoxicity to HeLa cells,
and they found overexpression of p-H2AX, a protein related
to early DNA fragmentation. In SiHa cells, the cytotoxicity
was less evident, as flow cytometry showed no effects on the
cell cycle and western blot analyses showed no evidence of
the overexpression of apoptogenic proteins. These results
suggest that apoptosis may not be the mechanism of cell
death that explains the cytotoxic effect exerted by LAAOcdt
on both CC lines.
l‑Amino acid oxidases (LAAOs)
Phospholipase ­A2 ­(PLA2)
These flavoenzymes catalyze the oxidative deamination of
l-amino acids, generating alpha-ketoacids, ammonium, and
hydrogen peroxide. The enzymes are widely distributed in
several organisms, and the LAAOs of snake venoms are
among the most widely studied. The abundance of these
proteins in the venom of some snakes, as well as the ease
of purification, make them an interesting object of study in
enzymology, structural biology, and pharmacology [69]. To
date, three studies have reported the effects of this family of
proteins isolated from snake venoms on CC cells. The first
publication is a 1996 study from Yonsei University (Korea),
in which purified LAAOs from the venom of Agkistrodon
halys adhered to the cytoplasmic membrane and generated
cytotoxic effects on HeLa cells. However, this cytotoxic
effect was the lowest reported among the effects induced in
leukemic lines [70].
In 2004, Zhang and Wei reported the effect of an LAAO
isolated from the venom of Agkistrodon acutus, ACTX-8,
on HeLa cells [71]. This LAAO induced apoptosis through
the intrinsic pathway, evidenced by the alteration in the
mitochondrial membrane potential, as well as the activa‑
tion of caspase-3 and -9. This process occurs when the
These components are between the most abundant in snake
venoms and belong to a large family of enzymes that hydro‑
lyze glycerophospholipids in the sn-2 position of the glyc‑
erol backbone, releasing lysophospholipids, arachidonic
acid, and fatty acids. These proteins are distributed widely
in nature in different secretions and biological fluids, such
as those generated by inflammatory processes, pancreatic
secretions, and tears. They can also be found in venoms of
arthropods, mollusks, and snakes, fulfilling essential roles
for the biological performance of these animals in their natu‑
ral environment [73]. In the case of the P
­ LA2s present in
snake venoms, effects on cellular processes such as prolifera‑
tion, migration, angiogenesis, and cell death, among others,
have been widely explored in the search for pharmacological
applications and the design of new drugs [74].
PLA2s found in snake venoms of the Viperidae family
belong to group II, which can be catalytically active if they
have an Asp residue in position 49 (catalytic region) or
devoid of this activity if the enzyme has a Lys residue in
this position, which affects its ability to bind the ­Ca2+ cofac‑
tor [75]. Cytotoxicity against cancer cells has been reported
for both types of ­PLA2s, suggesting that cytotoxicity is not
induction of apoptosis with no alterations in the integrity
of the cytoplasmic membrane or morphological changes in
cells characteristic of the apoptotic process (rounded cells
and granulation) when treating the cancer cells with the ­IC50
of the peptide. Additionally, in HeLa cells (and not noncan‑
cer cells), there was a dose-dependent increase in the expres‑
sion of the caspase-8 protein when cells were treated with
ICD-85; this caspase is an essential component of apoptosis
mediated by cell death receptors. These results allowed the
authors to conclude that ICD-85 selectively induces apopto‑
sis in HeLa cells and that one of the apoptotic mechanisms
induced by the peptide is the activation of caspase-8.
One year later, the same research group reported the
effects of a preparation of the peptide ICD-85 with nano‑
particles on HeLa cells. In this study, the cytotoxic effects
of the nanoparticles were significantly greater than those of
ICD-85 alone, and the alteration to the cytoplasmic mem‑
brane integrity was lower, which indicated less necrotic
effects. Nanoparticles induced the expression of the cas‑
pase-8 protein more potently than ICD-85, indicating that
treatment with the nanoparticles prepared from ICD-85 was
more effective than treatment with ICD-85 alone [68]. The
antitumor effects of peptides and peptide derivatives from
snake venoms on CC cells are summarized in Table 2.
13
Molecular Biology Reports
necessarily related to the enzymatic activity of phospholi‑
pases and that they are probably interacting with cell surface
receptors affecting intracellular pathways [76]. However, it
is still a matter of research to establish which type of recep‑
tors and signaling pathways P
­ LA2s of snake venoms interact
with cancer cells.
Most of the effects of P
­ LA2s on CC cells have been
assessed with Crotoxin, a heterodimeric protein purified
from the venom of the South American snake Crotalus
durissus terrificus [77]. Crotoxin generated a significant
dose-dependent cytotoxic effect on the ME-180R CC lines
with higher expression levels of EGFR compared to A431,
a line of squamous cell carcinoma. Muller et al. [78] also
evaluated the cytotoxicity of Crotoxin on nine tumor cell
lines, including two lines of CC (HeLa and SiHa). Despite
being cytotoxic for several carcinogenic cells, including
HeLa cells, at a dose of 30 µg/mL, SiHa cells were resist‑
ant to Crotoxin effects and did not present alterations in the
cell cycle, apoptosis, or the expression of proteins related to
proliferation or DNA damage. Interestingly, Crotoxin was
innocuous in nontumoral cells [78].
Recently, an acidic Asp49-PLA 2 isolated from the
Colombian Porthidium lansbergii lansbergii snake venom
showed cytotoxic activities on CC cell lines. This protein,
Pllans-II, inhibited the viability of HeLa and MCF-7 cells
(breast adenocarcinoma) in a dose-dependent manner, while
it was shown to be innocuous on mouse myoblasts, umbili‑
cal cord vascular endothelial cells (HUVECs), and breast
epithelial cells (MCF-10A). Treatment with one I­ C50 of
Pllans-II on HeLa cells generated cell cycle arrest in the
G0/G1 phase, apoptosis induction and a decrease in the
migratory capacity of these cells. HeLa cells treated with
Pllans-II presented a downregulation of BIRC5, Bax, and
BCL2 genes and upregulated BCL2L1 and caspase-8. These
results suggest the activation of the extrinsic apoptosis path‑
way and the possible interaction of P
­ LA2 with membrane
receptors involved in apoptosis induction. Pllans-II was
demonstrated to interact with α5 and β1 integrin subunits
on the HeLa cell membrane and showed an angiostatic effect
on HUVECs, with no changes in VEGF concentrations [62].
Pllans-II also affected the viability of CaSki cells in a dosedependent manner, with an ­IC50 very similar to that in HeLa
cells (100 µg/mL vs. 98 µg/mL, respectively, unpublished
data). Pllans-II also induced cell cycle arrest at the G2/M
phase, induced apoptosis, and inhibited adhesion and migra‑
tion. These effects on CaSki cells could also be related to
interactions with membrane receptors since treatment
with the protein did not alter the mitochondrial membrane
potential. The authors observed the interaction between the
C-terminal region of Pllans-II (noncatalytic) and the α5β1
integrin in silico by using protein–protein modeling analy‑
ses. The antitumor effects of snake venom proteins on CC
cells are summarized in Table 3.
Conclusion
Cervical cancer is a multifactorial disease that significantly
affects women worldwide. The studies included in this review
show that some snake venoms and their components can
induce cytotoxic effects on CC cells. Most of these studies
have been developed around the isolation of previously identi‑
fied molecules and the investigation of the mechanisms under‑
lying the cytotoxic effects, which include the alteration of the
cell cycle, induction of the cell death by extrinsic or intrinsic
apoptosis pathways, and inhibition of the metastatic ability
of cancer cells (cell adhesion, migration, and angiogenesis).
These findings provide new perspectives on drug discovery
and development for cancer treatment and can be used as a
basis for the design of more potent molecules. These studies
can also help to reveal cellular mechanisms that can be inves‑
tigated in greater depth for a better understanding of carcino‑
genesis and the development of metastasis. Given that there
are few studies on snake venom proteins in cervical cancer,
no preclinical or clinical studies are available in the literature
to determine their in vivo safety and efficacy. Therefore, more
research efforts are needed to delve into the mode of action of
the components of snake venoms in cervical cancer cells, thus
elucidating mechanisms that so far have not been described.
In addition, some limitations for future research on the
potential use of snake venom in CC treatment include the
correct delivery of the active components to the desired
anatomical sites and the limited amounts of purified com‑
ponents available for proof of concept and in vivo tests.
These problems could be assessed by evaluating the use of
nanomaterials for better bioavailability of the active com‑
pounds and the use of recombinant technology or chemical
synthesis for their design and large-scale production. The
use of these new technologies opens doors for increasing
future preclinical and clinical studies evaluating the use of
snake venom components in the treatment of CC and other
types of cancer.
13
Molecular Biology Reports
Table 3 Effects of proteins from snake venoms on cervical cancer cells
Protein
Snake
Cervical cancer cell line
Principal effects on cervical cancer cells
References
LAAOs
Agkistrodon halys
HeLa
[70]
Crotoxin ­[PLA2]
ACTX-8 [LAAO]
Crotalus durissus terrificus
Agkistrodon acutus
ME-180R
HeLa
LAAOcdt [LAAO]
Crotalus durissus terrificus
HeLa and SiHa
Crotoxin ­[PLA2]
Pllans-II
[Asp-49-PLA2]
Crotalus durissus terrificus
Porthidium lansbergii lansbergii
HeLa
HeLa
Pllans-II
[Asp-49-PLA2]
Porthidium lansbergii lansbergii
CaSki
Adheres to the cytoplasmic membrane and
generates a cytotoxic effect on HeLa cells
Cytotoxicity
Alteration in the mitochondrial membrane
potential, as well as the activation of
caspases 3 and 9, which induces intrinsic
pathway apoptosis
Activation of reactive oxygen species
Translocation of Bad and Bax proapoptotic
proteins from the cytosol to the mitochon‑
dria, which would promote the release of
apoptogenic mitochondrial proteins
Interaction between Bcl-xL and Bad proapop‑
totic proteins, and reduction in the interac‑
tion between Bcl-xL and Bak antiapoptotic
proteins
Cytotoxicity on both cell lines, but a stronger
effect on HeLa cells
In HeLa: overexpression of p-H2AX, a pro‑
tein related to early DNA fragmentation
In SiHa: overexpression of P21 protein
Cytotoxicity
Cytotoxicity
Cell cycle arrest in the G0/G1 phase of the
cell cycle
Apoptosis induction
Decrease on the migratory capacity
Downregulation of BIRC5, Bax and BCL2
genes
Up-regulation of BCL2L1 and CASP8 genes
Interaction with α5 and β1 integrins
Cytotoxicity
Arrest in the G2/M phase of the cell cycle
Apoptosis induction
Decrease on the adhesion and migratory
capacity
In Silico interaction with α5β1 integrin
Funding There is no funding source.
Compliance with ethical standards
2.
Conflict of interest The authors declare that they have no conflicts of
interest.
3.
Ethical approval This article does not contain any studies with human
participants or animals performed by any of the authors.
4.
References
1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A
(2018) Global cancer statistics 2018: GLOBOCAN estimates of
13
5.
[77]
[71]
[72]
[78]
[62]
MontoyaGómez
et al. [no
date]
incidence and mortality worldwide for 36 cancers in 185 coun‑
tries. CA: Cancer J Clin 68:394–424. https​://doi.org/10.3322/
caac.21492​
Pardo C, Cendales R (2018) Cancer incidence estimates and mor‑
tality for the top five cancer in Colombia, 2007–2011. Colombia
Médica 49:16–22. https​://doi.org/10.25100​/cm.v49i1​.3596
Munoz N, Bravo LE (2014) Epidemiology of cervical can‑
cer in Colombia. Salud Publica Mex 56:431–439. https​://doi.
org/10.21149​/spm.v56i5​.7368
American Cancer Society of Clinical Oncology (2008) Chemo‑
radiotherapy for Cervical Cancer Meta-Analysis Collaboration.
Reducing uncertainties about the effects of chemoradiotherapy
for cervical cancer: a systematic review and meta-analysis of
individual patient data from 18 randomized trials. J Clin Oncol
26:5802–5812. https​://doi.org/10.1200/JCO.2008.16.4368
Sturdza A, Potter R, Fokdal LU, Haie-Meder C, Tan LT,
Mazeron R et al (2016) Image guided brachytherapy in locally
Molecular Biology Reports
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
advanced cervical cancer: Improved pelvic control and sur‑
vival in RetroEMBRACE, a multicenter cohort study. Radio‑
ther Oncol 120:428–433. https ​ : //doi.org/10.1016/j.radon​
c.2016.03.011
Chaudary N, Pintilie M, Jelveh S, Lindsay P, Hill RP, Milosevic M
(2017) Plerixafor Improves Primary Tumor Response and Reduces
Metastases in Cervical Cancer Treated with Radio-Chemotherapy.
Clin Cancer Res 23:1242–1249. https​://doi.org/10.1158/10780432.CCR-16-1730
Bernardes-oliveira E, Gomes DL, Palomino GM, Juvenal K,
Farias S, Dias W et al (2016) Bothrops jararaca and Bothrops
erythromelas snake venoms promote cell cycle arrest and induce
apoptosis via the mitochondrial depolarization of cervical can‑
cer cells. Evid Based Complement Altern Med. https​://doi.
org/10.1155/2016/15749​71
Nurgali K, Jagoe RT, Abalo R (2018) Adverse effects of cancer
chemotherapy: anything new to improve tolerance and reduce
sequelae? Front Pharmacol 9:245. https​://doi.org/10.3389/fphar​
.2018.00245​
Li L, Huang J, Lin Y (2018) Snake venoms in cancer therapy:
past, present and future. Toxins. https​://doi.org/10.3390/toxin​
s1009​0346
Ma R, Mahadevappa R, Kwok HF (2017) Venom-based peptide
therapy: insights into anti-cancer mechanism. Oncotarget. https​
://doi.org/10.18632​/oncot​arget​.21740​
Vyas VK, Brahmbhatt K, Bhatt H, Parmar U (2013) Thera‑
peutic potential of snake venom in cancer therapy: current
perspectives. Asian Pac J Trop Biomed 3:156–162. https​://doi.
org/10.1016/S2221​-1691(13)60042​-8
World Health Organization (2019) Cancer. https​://www.who.
int/healt​h-topic​s/cance​r#tab=tab_1. Accessed 22 Sep 2019.
Mathers CD, Loncar D (2006) Projections of global mortality
and burden of disease from 2002 to 2030. PLoS Med 3:e442.
https​://doi.org/10.1371/journ​al.pmed.00304​42
Simms KT, Steinberg J, Caruana M, Smith MA, Bin LJ, Soer‑
jomataram I et al (2019) Impact of scaled up human papilloma‑
virus vaccination and cervical screening and the potential for
global elimination of cervical cancer in 181 countries, 2020–
99: a modelling study. Lancet Oncol 20:394–407. https​://doi.
org/10.1016/S1470​-2045(18)30836​-2
European Society for Medical Oncology (2018) ESMO Patient
Guide Series based on the ESMO Clinical Practice Guidelines.
https​: //www.esmo.org/for-patie​n ts/patie​n t-guide​s /cervi​c alcance​r. Accessed 22 Sep 2019.
Umar A, Dunn BK, Greenwald P (2012) Future directions in
cancer prevention. Nat Rev Cancer 12:835–848. https​: //doi.
org/10.1038/nrc33​97
American Cancer society (2016) What is cervical cancer? https​
://www.cance​r.org/cance​r/cervi​cal-cance​r/preve​ntion​-and-early​
-detec​tion/what-is-cervi​cal-cance​r.html. Accessed 22 Sep 2019.
National Cancer Institute at the National Institutes of Health
(2016) Cervical Cancer Treatment: Health Professional Version.
https​://www.ncbi.nlm.nih.gov/pubme​d/26389​493. Accessed 22
Sep 2019.
Sawaya GF, Smith-McCune K (2016) Cervical cancer screen‑
ing. Obstet Gynecol 127:459–467. https​: //doi.org/10.1097/
AOG.00000​00000​00113​6
Martínez JC, Pardo IFM, Medina RG (2015) Métodos actuales
de diagnóstico del cáncer de cuello uterino. Rev Ciencias Médi‑
cas La Habana 21:136–146
Giraudo E, Inoue M, Hanahan D (2004) An amino-bisphospho‑
nate targets MMP-9-expressing macrophages and angiogenesis
to impair cervical carcinogenesis. J Clin Invest 114:623–633.
https​://doi.org/10.1172/JCI22​087
22. De Palma M, Biziato D, Petrova TV (2017) Microenvironmental
regulation of tumour angiogenesis. Nat Rev Cancer 17:457–474.
https​://doi.org/10.1038/nrc.2017.51
23. Smith-McCune KK, Weidner N (1994) Demonstration and char‑
acterization of the angiogenic properties of cervical dysplasia.
Cancer Res 54:800–804
24. International Agency for Research on Cancer (2012) IARC
monographs on the evaluation of carcinogenic risks to humans
human papillomaviruses. https​://monog​raphs​.iarc.fr/wp-conte​
nt/uploa​ds/2018/06/mono9​0.pdf. Accessed 22 Sep 2019.
25. Trottier H, Franco EL (2006) The epidemiology of genital
human papillomavirus infection. Vaccine 24:S1–15. https​: //
doi.org/10.1016/j.vacci​ne.2005.09.054
26. Ault KA (2006) Vaccines for the prevention of human papillo‑
mavirus and associated gynecologic diseases: a review. Obstet
Gynecol Surv 61:S26–31. https​://doi.org/10.1097/01.ogx.00002​
21187​.63574​.5c
27. Schiffman M, Castle PE, Jeronimo J, Rodriguez AC, Wacholder
S (2007) Human papillomavirus and cervical cancer. Lancet
370:890–907. https​://doi.org/10.1016/S0140​-6736(07)61416​-0
28. Dunne EF, Unger ER, Sternberg M, McQuillan G, Swan DC, Patel
SS et al (2007) Prevalence of HPV infection among females in
the United States. JAMA 297:813–819. https​://doi.org/10.1001/
jama.297.8.813
29. Rodríguez AC, Schiffman M, Herrero R, Wacholder S, Hildesheim
A, Castle PE et al (2008) Rapid clearance of human papillomavi‑
rus and implications for clinical focus on persistent infections. J
Natl Cancer Inst 100:513–517. https​://doi.org/10.1093/jnci/djn04​
4
30. Jaisamrarn U, Castellsague X, Garland SM, Naud P, Palmroth J,
Del Rosario-Raymundo MR et al (2013) Natural history of pro‑
gression of HPV infection to cervical lesion or clearance: analysis
of the control arm of the large, randomised PATRICIA study.
PLoS ONE 8:e79260. https:​ //doi.org/10.1371/journa​ l.pone.00792​
60
31. Burd EM (2003) Human papillomavirus and cervical can‑
cer. Clin Microbiol Rev 16:1–17. https ​ : //doi.org/10.1128/
cmr.16.1.1-17.2003
32. Clifford GM, Smith JS, Plummer M, Munoz N, Franceschi S
(2003) Human papillomavirus types in invasive cervical cancer
worldwide: a meta-analysis. Br J Cancer 88:63–73. https​://doi.
org/10.1038/sj.bjc.66006​88
33. Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X,
Shah KV et al (2003) Epidemiologic classification of human pap‑
illomavirus types associated with cervical cancer. N Engl J Med
348:518–527. https​://doi.org/10.1056/NEJMo​a0216​41
34. Woodman CBJ, Collins SI, Young LS (2007) The natural history
of cervical HPV infection: unresolved issues. Nat Rev Cancer
7:11–22. https​://doi.org/10.1038/nrc20​50
35. Aguayo F, Anwar M, Koriyama C, Castillo A, Sun Q, Morewaya
J et al (2010) Human papillomavirus-16 presence and physical
status in lung carcinomas from Asia. Infect Agent Cancer 5:1–7.
https​://doi.org/10.1186/1750-9378-5-20
36. Zheng Z-M, Baker CC (2006) Papillomavirus genome structure,
expression, and post-transcriptional regulation. Front Biosci
11:2286–2302. https​://doi.org/10.2741/1971
37. Castillo A (2011) HPV infection and carcinogenesis in the
upper aero-digestive tract. Colomb Med 42:233–242. https​://doi.
org/10.25100​/cm.v42i2​.777
38. Yoon CS, Kim KD, Park SN, Cheong SW (2001) α6 Integrin is
the main receptor of human papillomavirus type 16 VLP. Biochem
Biophys Res Commun 283:668–673. https​://doi.org/10.1006/
bbrc.2001.4838
39. Ding L, Liu C, Zhou Q, Feng M, Wang J (2019) Association
of estradiol and HPV/HPV16 infection with the occurrence of
13
Molecular Biology Reports
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
cervical squamous cell carcinoma. Oncol Lett 17:3548–3554.
https​://doi.org/10.3892/ol.2019.10005​
Ramachandran B (2017) Functional association of oestrogen
receptors with HPV infection in cervical carcinogenesis. Endocr
Relat Cancer 24:R99–108. https​://doi.org/10.1530/ERC-16-0571
Chung S-H, Franceschi S, Lambert PF (2010) Estrogen and ERα:
culprits in cervical cancer? Trends Endocrinol Metab 21:504–511.
https​://doi.org/10.1016/j.tem.2010.03.005
Auborn KJ, Woodworth C, DiPaolo JA, Bradlow HL (1991) The
interaction between HPV infection and estrogen metabolism in
cervical carcinogenesis. Int J cancer 49:867–869. https​://doi.
org/10.1002/ijc.29104​90611​
Paavonen J, Koutsky LA, Kiviat N (1990) Cervical neoplasia and
other STD-related genital and anal neoplasias. Sex Transm Dis
2:561–592
Koutsky LA, Galloway DA, Holmes KK (1988) Epidemiology of
genital human papillomavirus infection. Epidemiol Rev 10:122.
https​://doi.org/10.1093/oxfor​djour​nals.epire​v.a0360​20
Mitrani-Rosenbaum S, Tsvieli R, Tur-Kaspa R (1989) Oestrogen
stimulates differential transcription of human papillomavirus type
16 in SiHa cervical carcinoma cells. J Gen Virol 70:2227–2232.
https​://doi.org/10.1099/0022-1317-70-8-2227
Plummer M, Herrero R, Franceschi S, Meijer CJLM, Snijders P,
Bosch FX (2003) Smoking and cervical cancer: pooled analysis of
the IARC multi-centric case–control study. Cancer Causes Control
14:805–814. https:​ //doi.org/10.1023/b:caco.000000​ 3811.​ 98261.​ 3e
Appleby P, Beral V, Berrington de Gonzalez A, Colin D, Franc‑
eschi S, Goodhill A (2007) Cervical cancer and hormonal con‑
traceptives: collaborative reanalysis of individual data for 16,573
women with cervical cancer and 35,509 women without cervical
cancer from 24 epidemiological studies. Lancet 370:1609–1621.
https​://doi.org/10.1016/S0140​-6736(07)61684​-5
Moreno V, Bosch FX, Munoz N, Meijer CJLM, Shah KV, Wal‑
boomers JMM et al (2002) Effect of oral contraceptives on risk of
cervical cancer in women with human papillomavirus infection:
the IARC multicentric case-control study. Lancet 359:1085–1092.
https​://doi.org/10.1016/S0140​-6736(02)08150​-3
Munoz N, Franceschi S, Bosetti C, Moreno V, Herrero R, Smith
JS et al (2002) Role of parity and human papillomavirus in cer‑
vical cancer: the IARC multicentric case-control study. Lancet
359:1093–1101. https:​ //doi.org/10.1016/S0140​-6736(02)08151​-5
International Collaboration of Epidemiological Studies of Cervi‑
cal Cancer (2006) Cervical carcinoma and reproductive factors:
collaborative reanalysis of individual data on 16,563 women with
cervical carcinoma and 33,542 women without cervical carcinoma
from 25 epidemiological studies. Int J cancer 119:1108–1124.
https​://doi.org/10.1002/ijc.21953​
Abraham AG, D’Souza G, Jing Y, Gange SJ, Sterling TR, Silver‑
berg MJ et al (2013) Invasive cervical cancer risk among HIVinfected women: a North American multicohort collaboration
prospective study. J Acquir Immune Defic Syndr 62:405–413.
https​://doi.org/10.1097/QAI.0b013​e3182​8177d​7
Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM (2007)
Incidence of cancers in people with HIV/AIDS compared with
immunosuppressed transplant recipients: a meta-analysis. Lancet
370:59–67. https​://doi.org/10.1016/S0140​-6736(07)61050​-2
Hoover RN, Hyer M, Pfeiffer RM, Adam E, Bond B, Cheville AL
et al (2011) Adverse health outcomes in women exposed in utero
to diethylstilbestrol. N Engl J Med 365:1304–1314. https​://doi.
org/10.1056/NEJMo​a1013​961
Ospina M, Martinez E, Pacheco OE, Bonilla H, Martínez V, Mar‑
tinez J (2017) Protocolo de Vigilancia en Salud Pública: Cáncer
de Mama y de Cuello Uterino. https​://www.ins.gov.co/busca​dorevent​os/Linea​mient​os/PRO%20C%C3%A1nce​r%20de%20mam​
a%20y%20cue​llo%20ute​rino-.pdf. Accessed 15 oct 2019
13
55. Serman F (2002) Cancer cervicouterino. Epidemiologia, historia
natural y rol del virus papiloma humano: Perspectivas en preven‑
ción y tratamiento. Rev Chil Obstet Ginecol 67:318–323. https​://
doi.org/10.4067/S0717​-75262​00200​04000​11
56. Dreyer G, Snyman LC, Mouton A, Lindeque BG (2005) Man‑
agement of recurrent cervical cancer. Best Pract Res Clin
Obstet Gynaecol 19:631–644. https​://doi.org/10.1016/j.bpobg​
yn.2005.03.003
57. Kim JH, Dougherty PM, Abdi S (2015) Basic science and
clinical management of painful and non-painful chemotherapyrelated neuropathy. Gynecol Oncol 136:453–459. https​://doi.
org/10.1016/j.ygyno​.2015.01.524
58. Fry BG (1999) Structure-function properties of venom compo‑
nents from Australian elapids. Toxicon 37:11–32. https​://doi.
org/10.1016/s0041​-0101(98)00125​-1
59. Markland FS (1998) Snake venoms and the hemostatic sys‑
tem. Toxicon 36:1749–1800. https​: //doi.org/10.1016/s0041​
-0101(98)00126​-3
60. Calvete JJ, Juarez P, Sanz L (2007) Snake venomics Strategy
and applications. J Mass Spectrom 42:1405–1414. https​://doi.
org/10.1002/jms.1242
61. Braud S, Bon C, Wisner A (2000) Snake venom proteins acting
on hemostasis. Biochimie 82:851–859. https​://doi.org/10.1016/
S0300​-9084(00)01178​-0
62. Jimenez-Charris E, Lopes DS, Gimenes SNC, Teixeira SC, Mon‑
tealegre-Sanchez L, Solano-Redondo L et al (2019) Antitumor
potential of Pllans-II, an acidic Asp49-PLA2 from Porthidium
lansbergii lansbergii snake venom on human cervical carcinoma
HeLa cells. Int J Biol Macromol 122:1053–1061. https​://doi.
org/10.1016/j.ijbio​mac.2018.09.053
63. Lee HL, Park MH, Hong JE, Kim DH, Kim JY, Seo HO et al
(2016) Inhibitory effect of snake venom toxin on NF-kappaB
activity prevents human cervical cancer cell growth via increase
of death receptor 3 and 5 expression. Arch Toxicol 90:463–477.
https​://doi.org/10.1007/s0020​4-014-1393-5
64. Park MH, Song HS, Kim KH, Son DJ, Lee SH, Yoon DY et al
(2005) Cobrotoxin inhibits NF-kappa B activation and target gene
expression through reaction with NF-kappa B signal molecules.
Biochemistry 44:8326–8336. https​://doi.org/10.18632​/oncot​arget​
.4192
65. Son DJ, Park MH, Chae SJ, Moon SO, Lee JW, Song HS et al
(2007) Inhibitory effect of snake venom toxin from Vipera
lebetina turanica on hormone-refractory human prostate can‑
cer cell growth: induction of apoptosis through inactivation of
nuclear factor kappaB. Mol Cancer Ther 6:675–683. https​://doi.
org/10.1158/1535-7163.MCT-06-0328
66. Sheu JR, Lin CH, Peng HC, Huang TF (1994) Triflavin, an
Arg-Gly-Asp-containing peptide, inhibits human cervical car‑
cinoma [HeLa] cell-substratum adhesion through an RGDdependent mechanism. Peptides 15:1391–1398. https​: //doi.
org/10.1016/0196-9781(94)90114​-7
67. Zare-Mirakabadi A, Sarzaeem A (2012) Extracellular caspase-8
dependent apoptosis on HeLa cancer cells and MRC-5 normal
cells by ICD-85 (venom derived peptides). Iran J cancer Prev
5:194–202
68. Moradhaseli S, Zare Mirakabadi A, Sarzaeem A, Kamalzadeh M,
Haji Hosseini R (2013) Cytotoxicity of ICD-85 NPs on human
cervical carcinoma HeLa cells through caspase-8 mediated path‑
way. Iran J Pharm Res IJPR 12:155–163
69. Du X-Y, Clemetson KJ (2002) Snake venom l-amino acid oxi‑
dases. Toxicon 40:659–665. https ​ : //doi.org/10.1016/s0041​
-0101(02)00102​-2
70. Suhr SM, Kim DS (1996) Identification of the snake venom sub‑
stance that induces apoptosis. Biochem Biophys Res Commun
224:134–139. https​://doi.org/10.1006/bbrc.1996.0996
Molecular Biology Reports
71. Zhang L, Wei LJ (2007) ACTX-8, a cytotoxic L-amino acid oxi‑
dase isolated from Agkistrodon acutus snake venom, induces
apoptosis in Hela cervical cancer cells. Life Sci 80:1189–1197.
https​://doi.org/10.1016/j.lfs.2006.12.024
72. Teixeira TL, Oliveira Silva VA, da Cunha DB, Polettini FL,
Thomaz CD, Pianca AA et al (2016) Isolation, characterization
and screening of the in vitro cytotoxic activity of a novel l-amino
acid oxidase [LAAOcdt] from Crotalus durissus terrificus venom
on human cancer cell lines. Toxicon 119:203–217. https​://doi.
org/10.1016/j.toxic​on.2016.06.009
73. Mora R, Valverde B, Diaz C, Lomonte B, Gutierrez JM (2005) A
Lys49 phospholipase A[2] homologue from Bothrops asper snake
venom induces proliferation, apoptosis and necrosis in a lympho‑
blastoid cell line. Toxicon 45:651–660. https​://doi.org/10.1016/j.
toxic​on.2005.01.008
74. Kini RM (2003) Excitement ahead : structure, function and
mechanism of snake venom phospholipase A2 enzymes. Toxicon
42:827–840. https​://doi.org/10.1016/j.toxic​on.2003.11.002
75. Rodrigues RS, Fernando L, Izidoro M, De ORJ, Sampaio SV,
Soares AM et al (2009) Snake venom phospholipases A2: a new
class of antitumor agents. Protein Pept Lett 16:894–898. https​://
doi.org/10.2174/09298​66097​88923​266
76. Osipov AV, Utkin YN (2015) Antiproliferative effects of snake
venom phospholipases A2 and their perspectives for cancer treat‑
ment. In: Gopalakrishnakone P (ed) Toxins and drug discovery.
Springer, Dordrecht, pp 1–15
77. Donato NJ, Martin CA, Perez M, Newman RA, Vidal JC, Etch‑
everry M (1996) Regulation of epidermal growth factor recep‑
tor activity by crotoxin, a snake venom phospholipase A2 toxin.
A novel growth inhibitory mechanism. Biochem Pharmacol
51:1535–1543. https​://doi.org/10.1016/0006-2952(96)00097​-4
78. Muller SP, Silva VAO, Silvestrini AVP, de Macedo LH, Caetano
GF, Reis RM et al (2018) Crotoxin from Crotalus durissus terrificus venom: in vitro cytotoxic activity of a heterodimeric phospho‑
lipase A2 on human cancer-derived cell lines. Toxicon 156:13–22.
https​://doi.org/10.1016/j.toxic​on.2018.10.306
79. Jiménez-Charris E, Montealegre-Sanchez L, Solano-Redondo L,
Mora-Obando D, Camacho E, Castro-Herrera F, Lomonte B et al
(2015) Proteomic and functional analyses of the venom of Porthidium lansbergii lansbergii (Lansberg’s hognose viper) from
the Atlantic Department of Colombia. J Proteomics 114:287–299.
https​://doi.org/10.1016/j.jprot​.2014.11.016
80. Jiménez-Charris E, Montealegre-Sánchez L, Solano-Redondo L,
Castro-Herrera F, Fierro-Pérez L, Lomonte B (2016) Divergent
functional profiles of acidic and basic phospholipases A2 in the
venom of the snake Porthidium lansbergii lansbergii. Toxicon
119:289–298. https​://doi.org/10.1016/j.toxic​on.2016.07.006
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
13
Descargar