Invest Clin 62(3): 247 - 275, 2021 https://doi.org/10.22209/IC.v62n3a06
Corresponding Author: José Núñez Troconis. Departamento de Obstetricia y Ginecología, Facultad de Medicina.
Universidad del Zulia. Maracaibo, Venezuela. E-mail: jtnunezt@gmail.com
Chlamydia trachomatis. Co-factor or factor
in cancer of the cervix?
José Núñez Troconis
Departamento de Obstetricia y Ginecología, Facultad de Medicina. Universidad del Zu-
lia. Maracaibo, Venezuela.
Key words: Chlamydia trachomatis; cancer of the cervix; inflammation mechanisms;
carcinogenesis; sexual transmitted infection.
Abstract. The objective of this article was to review and to analyze the
possible role that Chlamydia trachomatis has as a co-factor in the origin and
development of cervical cancer. For that purpose, the Latin-American and
international bibliography was reviewed using the Pub-Med, Google Scholar,
Springer, the Cochrane Library, Embase, Scielo, Imbiomed-L, Redalyc and Lat-
index databases. The searches included the key words: Chlamydia trachomatis,
epidemiology of Chlamydia trachomatis, epidemiology of cervical cancer, Chla-
mydia trachomatis and infection, Chlamydia trachomatis and inflammation
mechanisms, cervical cancer and co-factors, sexually transmitted infections
and cervical cancer, cancer and inflammation mechanisms, carcinogenesis, in-
flammation mechanisms. Publications from 1970 to June 2020 were reviewed
and analyzed. This review article analyzes the possible mechanisms that Chla-
mydia trachomatis could play in the carcinogenesis of the cervical cancer as a
co-factor with the human papilloma virus or as an independent factor.
248 Núñez Troconis
Investigación Clínica 62(3): 2021
Chlamydia trachomatis: ¿Co-factor o factor en el cáncer
del cuello uterino?
Invest Clin 2021; 62 (3): 247-275
Palabras clave: Chlamydia trachomatis; cáncer del cuello uterino; mecanismo de la
inflamación; carcinogénesis; infecciones de transmisión sexual.
Resumen. El objetivo de este artículo fue revisar y analizar el posible papel
que la Chlamydia trachomatis tiene como co-factor en el origen y desarrollo del
cáncer del cuello uterino. Para dicho propósito, se revisó la bibliografía latino-
americana e internacional en las bases de datos de Pub-Med, Google Scholar,
Springer, la biblioteca Cochrane, Embase, Scielo, Imbiomed-L, Redalyc and La-
tindex. La búsqueda incluyó las palabras claves: Chlamydia trachomatis, epide-
miología de la Chlamydia trachomatis, epidemiología del cáncer del cuello ute-
rino, cáncer del cuello uterino, Chlamydia trachomatis e infección, Chlamydia
trachomatis y mecanismo inflamatorios, cáncer del cuello uterino y co-factores,
enfermedades de transmisión sexual y cáncer del cuello uterino, cáncer y me-
canismos de la inflamación, carcinogénesis, mecanismos de la inflamación. Se
revisaron y analizaron publicaciones desde 1970 hasta junio 2020. Este artí-
culo de revisión analiza los posibles mecanismos que la Chlamydia trachomatis
pudiera jugar en la carcinogénesis del cáncer del cuello uterino tanto como co-
factor con el virus del papiloma humano o como factor independiente.
Received: 01-02-2021 Accepted: 10-04-2021
INTRODUCTION
Chlamydia trachomatis (Ct) infections
are the most commonly reported sexually
transmitted bacterial infections, and the
most common bacterium responsible for
sexually transmitted infections, globally (1).
Most (70%-80%) of these infections are as-
ymptomatic, often remain undetected and, if
not treated, can lead to severe complications,
mainly in young women. Advances in diag-
nostic techniques and methods of specimen
collection have made easier the detection,
treatment and prevention of these infections
of global public health significance (2).
Globally, more than 1 million curable
sexually transmitted infections (STIs) occur
each day. According to World Health Organi-
zation (WHO). The global estimate for 2016,
was roughly 376 million new infections (ex-
cluded the viral STI), more than 1 million
per day, of the four curable STIs: Chlamydia,
gonorrhoea, syphilis and trichomoniasis.
Trichomonas vaginalis (Tv): 156 millions in-
fections, Ct: 127 millions infections, gonor-
rhea: 87 millions infections and, syphilis: 6
millions infections (3). According to WHO,
the estimation of global prevalence of Ct
in 15-49 years old women was 3.8% and the
prevalence was found higher in upper and
middle income countries. For the American
continent, the prevalence was estimated in
7% (3.8%-6.6%). The global incidence rate
for Ct in 2016 was estimated to be 34 cases
per 1000 women. The American continent
had the highest incidence rate for Ct; like-
Chlamydia and Cervical Cancer 249
Vol. 62(3): 247 - 275, 2021
wise, the upper and middle income coun-
tries, had the higher incidence (4). In 2015,
Redmond et al. (5) reported a prevalence of
Ct in women of 3.6% (range: 3.0%-5.3%) in
countries member of European Union. In
our country, Venezuela, different authors
have reported an incidence among 12.8%
to 25% (4-6). One of the major problem is
that approximately 2/3 of the world’s pop-
ulation has limited access to Ct screening
and treatment programs. The predominantly
asymptomatic nature of Ct infection also re-
sults in many undiagnosed individuals who
go untreated, and hence, continue to spread
infection. These untreated infections could
also have other devastating consequences,
as Ct infection can also increase susceptibil-
ity to, and transmission of others STIs such
as gonorrhea, Tv, Human Immunodeficiency
Virus (HIV), Human Papillomavirus (HPV),
Hepatitis virus C (HVC), etc. (9).
Among the risk factors to get a Ct in-
fection, it could be mentioned: early age in
the initiation of sexual activity, especially in
women under 20 years old, unmarried status,
nulliparity, black race, poor socio-economic
conditions, multiple sexual partners, pro-
miscuous sexual partner, new sexual partner,
smoking habit, lack of use of barrier contra-
ceptive devices and concurrent gonococcal
infection (10-14). In addition, cervical chla-
mydial infections are found to be associated
with the use of oral contraceptives (14).
The objective of the present study was
to review and to analyze the relationship of
Ct infection with the cervical cancer (CC).
MATERIAL Y METHODS
Literature searches were performed
electronically in PubMed, Medline, ISI,
DOAJ, Springer, Embase, Web of Knowledge,
DOAJ, Google Scholar and the Cochrane
Library for original articles written in the
English language and in Scielo, Lantidex,
Imbiomed-L, Redalyc and Google Scholar
for original articles written in Spanish. Se-
lection criteria included randomized clinical
trials, observational trials, open-label non-
randomized trials, and case reports related
to Ct and CC. The Cochrane Library was
searched for reviews. Publications from 1970
to June 2020 were reviewed.
The searches included the key words:
Chlamydia trachomatis, epidemiology of
Chlamydia trachomatis, epidemiology of
cervical cancer, Chlamydia trachomatis and
infection, Chlamydia trachomatis and in-
flammation mechanisms, cervical cancer
and co-factors, sexual transmitted infections
and cervical cancer, cancer and inflamma-
tion mechanisms, carcinogenesis, inflamma-
tion mechanisms.
Chlamydia trachomatis
Microorganism
Chlamydiae are a Gram-negative,
spherical or ovoid obligate intracellular bac-
teria and have a unique, generally biphasic,
developmental cycle. Infectious, non-replica-
tive elementary bodies (EBs) infect genital
columnar epithelial cells and reside within a
membrane-bound vacuole termed an inclu-
sion but not able to divide. Here, EBs differ-
entiate into non-infectious vegetative reticu-
late bodies (RBs), which are metabolically
active and able to replicate, to grow into
the cell and to differentiate back into EBs,
and exit cells via lysis or extrusion mecha-
nisms; EBs released into the mucosal lumen
can then infect nearby epithelial cells or be
transmitted to sexual partners via genital
secretions (2,9,15). Within 8-12 divisions
(16), differentiation to EBs is initiated and
the cycle is complete when the cell releases
the contents of the inclusion to attach to
adjacent cells and reinitiate the cycle. Ct
has DNA and RNA, multiply by binary fission
rather than self-assembly, contain their own
ribosome, have a peptidoglycan free cell wall
(1,17). Chlamydiae can be classified into 19
serovars or serotypes (genovars) based on
antigenic variation in the major outer mem-
brane protein (MOMP) epitopes encoded
by ompA (1,17). Serovars or serotypes A, B,
Ba and C are associated with trachoma, se-
250 Núñez Troconis
Investigación Clínica 62(3): 2021
rovars or serotypes D-K are most commonly
with urogenital infection and serovars or se-
rotypes L1-L3 represent strains causing inva-
sive lymphoma granuloma venereum (LGV)
(see Table I).
Mode of transmission
The mode of transmission is by sexual
contact or intercourse. Chlamydial infection
can occur at any anatomical site of sexual
contact including endocervix, urethra, rec-
tum, and oropharynx (1).
Clinical manifestation
Infections in women
Chlamydial infection in women is
commonly asymptomatic (18). Women
with cervicitis/endocervicitis can be as-
ymptomatic or may complain of mucopu-
rulent vaginal discharge or throughout
the external orifice of the cervix and/or
postcoital bleeding. Edema, congestion
and bleeding of the cervix have been ob-
served. Urethral infection can be associ-
ated with cervicitis. A culture-negative
leucocyturia finding is suggestive of Ct
infection. Ascending infections can result
from cervicitis. Endometritis is frequently
associated with this and may produce ir-
regular uterine bleeding; the endometritis
can be acute or chronic. Other patholo-
gies, because of the ascending infections,
are salpingitis and pelvic inflammatory
disease (PID). They are often subclinical
and Ct is the cause of least 60% of cases
of acute PID (2,19). Salpingitis may lead
to tubal scarring and severe reproductive
complications. Two-thirds of all cases of
tubal factor infertility and 1/3 of all cases
of ectopic pregnancy could be due to chla-
mydial infection (2,20,21). Chronic pelvic
pain linked to the presence of peritoneal
adhesions may occur in more than 15%
of women with previous episodes of PID
(2,19). Salpingitis and peritoneal adhe-
sions are associate to infertility and ecto-
pic pregnancy (21). Ct is also considered a
leading cause of PID and female infertility
worldwide. Fitz-Hugh–Curtis syndrome, a
perihepatitis observed after or in conjunc-
tion with salpingitis, is more commonly
associated with Ct than with gonococcal
infections (2). There is little evidence,
and this is conflicting, to implicate Ct in
chorioamnionitis and adverse pregnancy
out-come (2,19). Postpartum endometri-
tis occurs in 30% of women with antena-
tal chlamydial infection. In both men and
women, Ct may be involved in conjuncti-
vitis by auto-inoculation from the genital
tract (2) (see Table I).
TABLE I
Chlamydia trachomatis
CLINICAL MANIFESTATIONS
Serovar Clinical Manifestation Complications
A-C Keratoconjunctivitis Scarring trachoma, blindness
D-K Male: urethritis, proctitis
Female: cervicitis, urethritis, proctitis,
endometritis, salpingitis. perihepatitis
Male and female: conjuntivitis
Male: epididymitis
Female: acute and chronic pelvic inflammatory
disease: pelvic pain, ectopic pregnancy, Fiztz-
Hugh-Curtos syndrome, infertility.
Male and female: Reiter’s syndrome, reactive
arthritis.
L1-L3 Lymphogranuloma venereum:
inguinal syndrome, proctitis
Fibrosis, rectal stricture
Chlamydia and Cervical Cancer 251
Vol. 62(3): 247 - 275, 2021
Infections in men
Ct is the major cause of non-gonococcal
urethritis and post-gonococcal urethritis.
Urethritis can be complicated by acute epi-
didymitis in young men. After 7–21 days of
incubation, the symptoms include dysuria,
and a moderate clear or whitish urethral
discharge (2,20). Acute proctitis can be as-
sociated with oculo-genital serovars or sero-
types, but is usually milder than that associ-
ated with LGV serovars. There is no evidence
of the role of Ct in prostatitis (22), and chla-
mydial infection does not significantly con-
tribute to male infertility (23). Reiter’s syn-
drome (urethritis, conjunctivitis, arthritis
and mucocutaneous lesions) or reactive ar-
thritis have also been associated with genital
Ct infections, with a high male/female ratio
(22). (see Table I).
Pathogenesis
Immunopathogenesis
Ct is a strong immunogenic, which
stimulates both humoral and cell mediated
immune responses. In addition to the immu-
nogenic antigens, the outcome of chlamydi-
al infection depends on the interaction and
balance of cytokines secreted by the activat-
ed lymphocytes. Interferon gamma (IFN-γ)
has been described as the single most impor-
tant factor in host defense against Ct, while
disease susceptibility has been linked with
enhanced expression of interleukin-10 (IL-
10). Immune system changes or disturbanc-
es induced by Ct may favor its own survival
in the infected host, and induce persistent
infections (11). Ct infection may be primary
or a chronic/recurrent/re-infection.
Primary infection: A serial infection of
the mucosal cells is seen during the primary
infection. The damaging and infected epi-
thelial cells secrete numerous pro-inflamma-
tory chemokines and cytokines, including
IL-1, IL-6, IL-8, granulocyte-macrophage
colony stimulating factor (GM-CSF), growth
regulated oncogene, and tumor necrosis fac-
tor alpha (TNF-α) (11/24,25). The released
cytokines cause vasodilatation, increased
endothelial permeability, activation and in-
flux of neutrophils, monocytes and T-lym-
phocytes, and elevated expression of adhe-
sion molecules. In addition, it stimulates
other cells to secrete cytokines. Neutrophils
appear to play a role in reducing the initial
amplification of Ct and possibly in limiting
the spread within the female genital tract.
IL-1 is secreted initially by the undamaged
cells and stimulates the secretion of other
cytokines from other non-infected cells,
like TNF-α (26). During the same period, Ct
passes via lymphatic vessels to local lymph
nodes. The decaying epithelial cells release a
few EBs, which are phagocytosed by neutro-
phils through phagolysosomes.
T lymphocytes, mainly T helper cells
(Th1), play an important role during early
phase of infection, which, due to Ct antigen-
induced activation, secrete IFN-γ, necessary
for infection regression. It increases the po-
tential of various phagocytes to destroy Ct
and stimulates the secretion of other cyto-
kines, including IL-1. IL-1, in turn, by stimu-
lating the secretion of IL-2 by Th1 cells, it
causes increased replication of cytotoxic lym-
phocytes and natural killer cells.
.
The role of
secretory IgA has also been established in the
neutralization of primary infection (11,27).
It has been observed that a single acute epi-
sode of chlamydial infection cannot lead to
serious sequelae associated with this infec-
tion; persistent infection may be responsible
for the serious consequences (11).
Chronic infection/recurrent/reinfec-
tion: Chronic infection, associated with per-
sistence of Ct in the host cells, recurrent in-
fection or reinfection are more dangerous.
A delayed hypersensitivity reaction or rarely,
type 3 hypersensitivity reactions (Arthus re-
action) is observed in the long term or re-
current stimulatory action of chlamydial
antigens. Antibodies are not involved in the
delayed type of reaction developing within
24-48 hrs due to antigen interaction with
specifically sensitized Th1 lymphocytes. Pro-
cesses, which occur during these reactions,
lead to tissue damage, fibrosis and cicatriza-
252 Núñez Troconis
Investigación Clínica 62(3): 2021
tion within the affected organs. Irreversible
consequences, like PID leading to mechani-
cal infertility, ectopic pregnancy, chronic pel-
vic pains and chronic urethritis, may occur.
After a single episode of salpingitis about
one in 10 patients become infertile because
of tubal occlusion. After 2-3 episodes, infer-
tility ensues in about 35-70% cases. In sev-
eral studies, repeated chlamydial infection
was associated with PID and other reproduc-
tive sequelae, although it was difficult to
determine whether the risk per infection in-
creased with recurrent episodes (28). Lack
of treatment or improper therapeutic man-
agement may result in chronic infection. A
significant role of dietary factors like insuffi-
cient supply of tryptophan, L-isoleucine and
cysteine in diet, as well as certain cytokines
like INF-γ, TNF-α, have been observed and
reported (29). Formation of atypical chla-
mydial forms in vitro has been demonstrated
in INF-γ treated cells. The atypical forms
(29)
are large, non-infectious, have reduced
metabolic activity, and do not replicate, yet
remain alive. Such atypical forms display
decreased levels of MOMP and lipopolysac-
charide (LPS) antigens but continue with
high production of chlamydial heat shock
protein 60 (hsp60), which is capable of in-
ducing chronic inflammation and scarring.
Chronic and occult infections pose several
diagnostic and therapeutic problems. Due
to the variable antigenic structure of atypi-
cal forms, the routine diagnostic methods
do not always identify them. Moreover, these
forms have reduced MOMPs, which lead to
decreased transport of antibiotic across the
cell. Therefore, in case of chronic infections,
therapy frequently results in failure.
Reinfection is due to the repeated in-
fection, while recurrence is caused by the
presence of a Ct reservoir in the lymph node
and spleen (29).
Macrophages have been
found to play an important role in the recur-
rence of infection as Ct circulates within the
macrophages, finding a temporary shelter
in the lymph nodes, spleen and serous cavi-
ties. It has been observed that recurrences
were more frequent in young patients with
prolongation of the active period in compari-
son with patients in older age group (30).
The less common spread of infection in the
older age group has been attributed to low
exposure to Ct and by physiological changes
which reduce sensitivity to the acquisition
(29).
Inflammation and cancer
Inflammation is mediated by immune
cells as an immediate defense in response
to infection or injury by noxious stimuli. In-
nate immune cells such as neutrophils, mast
cells, and macrophages possess receptors
that signal the activation and production of
an array of biologically active proteins and
defense molecules in response to foreign
substances as well as to damaged or altered
self-molecules (31,32). The infiltration of
immune cells into sites of solid tumors, ob-
served first by Rudolf Virchow in 1863 (31)
has for many years been pursued as a failed
effort of the immune system to resist tumor
development. Though this is true and the
basis of tumor escape from immune surveil-
lance, Virchow’s idea that the immune cells
associated with tumors reflected a role for
these cells in the origination of cancer was
the first to suggest that the immune cells
‘themselves’ were active participants in tu-
mor development.
Inflammation and tumor development
It is now well recognized that the pres-
ence of inflammatory cells commonly pre-
cedes tumor development (33). Demon-
stration that inflammation plays a causal
role in tobacco-related carcinogenesis, vi-
ral carcinogenesis and asbestos-associated
carcinogenesis highlights the significance
of inflammation in tumorigenesis. Substan-
tial evidence from both experimental mod-
els and human studies have demonstrated
that inflammation fosters the development
of tumors by acting on or with the cancer
hallmarks identified by Hanahan et al. (34).
This includes effects on evasion of apoptosis,
Chlamydia and Cervical Cancer 253
Vol. 62(3): 247 - 275, 2021
uncontrolled growth and dissemination, as
well as altering/deregulating tumor immune
surveillance. In fact, Colotta et al. (33) sug-
gested that inflammation be considered a
separate cancer hallmark, an idea supported
in the update to the cancer hallmarks, where
because of the broad acting role of inflam-
matory cells in tumor development, Hana-
han et al. (34) conceptualized the role of
inflammation as one of ‘enabling’ or cause
of tumorigenesis.
Acute inflammation possesses two bal-
anced and biologically opposing effector
arms represented in a ‘yin’ (pro-apoptotic
or tumoricidal) and ‘yang’ (wound healing
or pro-tumorigenic) relationship model,
where immune cells participate with the
non-immune cells in the local environment
(e.g. epithelial, vasculature and neuronal)
(35). Unresolved and persistent inflamma-
tion has been described as the loss of or de-
regulation in the balance between the ‘yin’
and ‘yang’ responses. The role of persistent
inflammation as a contributing factor in tu-
morigenesis is well accepted and, in many
cancers, thought to be a necessary compo-
nent. Examples include a causal relation-
ship between inflammation and infectious
agent-associated cancers [e.g. hepatitis B
and C virus (liver), human papilloma virus
(e.g. cervix, anal) and the bacterium Helico-
bacter pylori (stomach)]. The relationship
between cancer and inflammation is also
supported by the elevated risk of cancer in
chronic inflammatory conditions, such as
colitis-associated colorectal cancer. Impor-
tantly, the cause-effect relationship between
inflammation and cancer is a challenging
concept as it implies that inflammation
precedes the processes. However, current
evidence widely suggests that in the case of
cancer, which is a multi-step and complex
process, inflammation is an integral compo-
nent of the overall pathogenesis of disease
at the microenvironment level that not only
contributes in a causal way but also sup-
ports a permissive state for tumors to grow
(34). As such, it is important to recognize
that tumor-associated inflammation (TAI)
in solid tumors is itself a complex patholog-
ic process, with contributions from classic
immune cells as well as poorly character-
ized, cancer-associated fibroblasts and the
epithelial tumor cell compartment (31).
Cellular mechanisms of inflammation
and tumorigenesis
Over the past two decades, our under-
standing of inflammation in tumorigenesis
has led to the identification of a number of
molecules that are strongly linked to the
development of human cancers (33,36,37).
Like tumorigenesis, tumor-promoting in-
flammation and TAI are the phenotypic
product of a complex set of cellular and mo-
lecular interactions that result in an imbal-
ance in local microenvironment that is most
analogous to an unresolved ‘wound-healing’
response (37). The cellular and molecular
composition of TAI has been the subject of
a number of extensive recent reviews (31-
33,35,37,38).
A number of the cellular and molecu-
lar mechanisms involved in inflammation
induced tumor initiation, promotion, and
progression are now well described (see Ta-
ble II). These inflammation-induced changes
occur at the cellular and tissue level. Among
the best characterized are the pro-inflamma-
tory and mutagenic reactive oxygen (ROS)
and reactive nitrogen species (RNS), cyto-
kines, chemokines and lipid-derived prod-
ucts of the inducible COX-2 in arachidonic
acid metabolism including the highly potent
prostaglandin (PG)-E
2
. At physiological lev-
els, ROS and RNS are important cell signal-
ing molecules (39), however, at high levels
or with aberrant production, ROS and RNS
are capable to cause considerable cellular
damage resulting in cell injury, DNA dam-
age and an inflammatory response (40,41).
During tumorigenesis, ROS and RNS have
been characterized for their ability to induce
a plethora of effects on cells and on the lo-
cal environment that include DNA damage,
adduct of cellular protein and lipids, and in
254 Núñez Troconis
Investigación Clínica 62(3): 2021
the absence of apoptosis at high levels, pro-
motion of abnormal cell proliferation and
transformation (39). High levels of ROS and
RNS are produced by the innate immune sys-
tem in response to tissue injury or damage.
ROS and RNS are produced in response to
cell-damage by inflammatory cells. The un-
resolved damage leads to a potential vicious
cycle producing chronic and high levels of
ROS and RNS. These high levels and chron-
ic exposure of cells to ROS and RNS from
macrophages and mast cells are linked to a
range of tissue pathologies, including neu-
rodegenerative and autoimmune diseases,
along with the propagation of mast cells that
are thought to promote myeloid-suppressor
cell expansion that inhibit tumor immuno-
surveillance as well as to maintain the tumor
promoting microenvironment (39,42,43).
This deregulation of ROS and/or RNS pro-
duction have been, and continue to be inves-
tigated as biological indicators as potential
exogenous and endogenous cause of cancer,
independent of the DNA damage (31).
Cyclooxygenase, prostaglandins and their
receptors
The cyclooxygenase (COX) enzymes
were among the first identified molecular tar-
gets of interest in TAI. There are three COX
TABLE II
MOLECULES, CELLS AND TISSUES ALTERATIONS WITH CHRONIC INFLAMMATION
AND TUMOR PROMOTING CONSENQUENCE
Genomic instability, chromosome remodeling, epigenetic changes and altered gene and miRNA
expression
Altered post-translational modification, activity and localization of cell proteins
Altered cell metabolism
Induction of cell growth and anti-apoptotic signals uncontrolled cell growth and retention of cells
with damaged genomes
Vasodilation, leakage of the vasculature and infiltration of leukocytes disrupted tissue integrity
and altered microenvironment and immunosuppression and recruitment of myeloid suppressor cells
Altered cell polarity disturbance in stroma/epithelial tissue matrix and loss of differentiation
signals
Tissue necrosis neovascularization and hypoxia
Induction of matrix metalloproteinases invasiveness and spread
Chlamydia and Cervical Cancer 255
Vol. 62(3): 247 - 275, 2021
isoforms: COX-1 or prostaglandin G/H syn-
thase 1 (PTGS1), which is constitutively ex-
pressed; COX-2 (PTGS2), the inducible form
of the COX enzymes; and COX-3, an alterna-
tive variant of COX-1. COX enzymes catalyze
the formation of lipid mediators, including
prostanoids, prostacyclins and thrombox-
anes. COX-2 is over-expressed in acute and
chronic inflammation as well as in tumors.
Extensive research efforts over the past three
decades have established a strong link be-
tween COX-2 expression, inflammation, and
cancer. COX-2 suppression prevents neopla-
sia in numerous rodent models of cancer as
well as in human clinical trials (44); several
epidemiological studies have reported lower
cancer rates in regular users of aspirin and
other non-steroidal anti-inflammatory agents
that are now explained by the inhibitory activ-
ity of these drugs on the pro-inflammatory/
pro-tumorigenic effects of PGE-2
(44,45).
COX-2 can be induced by a number of fac-
tors including cytokines, chemokines, ROS
and environmental chemicals. Induction of
COX-2 activates mPGES-1, the inducible en-
zyme that catalyzes the COX-2-derived lipid
intermediate PGH-2
to PGE-2, the biological
mediator of the tumorigenic effects of COX-
2. PGE-2 is the most abundant PG in solid tu-
mors and has been shown to influence tumor
cell growth, migration and invasiveness. The
tumorigenic actions of PGE-2
are numerous
and include the induction of angiogenesis,
transactivation of the epidermal growth fac-
tor receptor, inhibition of apoptosis and im-
munosuppression (46). The physiological and
pathological effects of PGE-2
are mediated
through interactions with specific PG recep-
tor subtypes present on multiple cell types,
including most immune cells and epithelial
cells. PGE-2
shows the highest affinity for the
EP receptor subtypes 1–4 (PTGER1-4 or EP1-
4). All four of the EP receptors are present
on the majority of cells involved in immune
responses (47, 48). Under normal conditions,
PGE-2
attenuates the activity of macrophages
and dendritic cells by inhibiting the produc-
tion of tumor necrosis factor (TNF)-α and
interleukin (IL)-10. The EP2 and EP4 recep-
tors mediate these activities as well as regu-
late the proliferation and differentiation of T
and B cells. And while it is clear that the bio-
logical activities of PGE-2
is determined by
the nature and distribution of the EP recep-
tors, very little is known about the EP recep-
tor subtype/PGE-2 interactions, interaction
with environmental chemicals, interaction
with bacterial and virus infections. It is im-
portant to recognize that COX-2 expression
is regulated by a number of transcription fac-
tors that its deregulation could lead to the
sustained induction of COX-2 as a co-factor
in TAI. These include the hypoxia inducible
factors (HIF-1α and HIF-2α), NF-κB, and sig-
nal transducer and activator of transcription
(STAT) (49,50).
STAT family proteins regulate cytokine-
dependent inflammation and immunity.
STAT protein family members, including
STAT 1–6, are over-expressed in a number of
human cancers. The role the STATs in TAI
has recently been well established in pros-
tate cancer where chronic inflammation is
believed to play a major role in tumor devel-
opment (51). STAT3 has been linked to the
induction and maintenance of an inflamma-
tory microenvironment in the prostate and
to the malignant transformation and pro-
gression due to the maintenance of a pro-in-
flammatory state. The pro-inflammatory cy-
tokine IL-6 is a potent inducer STAT3 where
binding to the IL6R induces activation of
the Janus tyrosine family kinase (JAK)-signal
transducer leading to a phosphorylation de-
pendent activation STAT3. STATs, like other
transcription factors, have a dual role in in-
flammation and is considered to be both a
friend and an enemy of tumorigenesis (52).
They can be induced by inflammation and
they can induce inflammation by activating
NFκB and IL-6 pathways. The activation of
IKKβ/NFκB is potent stimuli for IL-6 and
thus activation of the STAT3 protein. Inflam-
mation is an established risk factor for hepa-
tocellular cancer (HCC) from viral infection
and other environmental or drug insults.
256 Núñez Troconis
Investigación Clínica 62(3): 2021
STAT3 is over-expressed in the majority of
HCC in human with high levels correlated
with IL-6 levels in the local tumor (53), find-
ings that support a role of IL-6 and STAT3
as a TAI phenomenon in HCC in humans.
Given the role of STATs in inflammation, and
evidence as an important signaling molecule
in TAI, the STAT transcription factors rep-
resent an important and unexplored family
of molecules as putative mediators of TAI in
the presence of environmental chemicals,
biological and other toxicants (31).
Cytokines as immune effector molecules
Cytokines are a large group of small
proteins (5–20 kD) that act as a paracrine
and autocrine messengers with a wide spec-
trum of biological functions across numer-
ous tissue and cell types. Collectively, the
cytokines include chemokines, interferons,
interleukins, lymphokines and tumor necro-
sis factors (TNF). Cytokines are produced
by cells of the immune system: B and T lym-
phocytes, macrophages, mast cells, stromal
cells: endothelial cells and fibroblasts, as
well as tumor cells. Cytokines exhibit para-
crine and autocrine effects on a wide range
of tissues and cells. The cytokine most con-
sistently associated with tumor cell killing is
TNFα. After TNFα interacts with its receptor,
a subsequent chain of cellular events leads
to the activation of the transcription fac-
tor, nuclear factor (NF)-κB and subsequent
production of IL-1β, IL-6, IL-8 and IL-17.
In the simplest mechanistic model, these
pro-inflammatory molecules are coupled to
each other via TNF-α binding to its receptor
(TNFR), which activates the NF-κB pathway
in the acute phase response. This results in
the up-regulation of a group of pro-inflam-
matory cytokines as a programmed response
to wounding or infection. It is this response
that is triggered in the initial response to
injury or infection (54) but when it is unre-
solved or chronic, is widely believed to pro-
mote tumorigenesis and contribute or en-
able tumor progression.
Under homeostatic conditions, two
membrane receptors, TNFR1 and TNFR2,
mediate the actions of the TNF family of mol-
ecules (55). While initially described as an
anti-tumor molecule, the role of TNF-α as pro-
tumorigenic is now well characterized. Tumor
and inflammatory cells within the tumor mi-
croenvironment produce TNFα, supporting
tumorigenesis and metastasis by promoting:
genomic instability through the production
of ROS and RNS, cell survival by deregulat-
ing apoptotic pathways, promoting invasion
through induction of matrix metalloprotein-
ases (MMPs), and angiogenesis via the induc-
tion of pro-angiogenic factors. Part of this re-
sponse may be due to the presence of TNFR1
on tumor, stromal and immune cells, thereby
allowing TNF-α to exert its activity both di-
rectly on the tumor and indirectly within the
tumor microenvironment to sustain local
inflammation and recruitment of cells with
inhibitory effects on tumor immunity. The ef-
fects of TNFα as a pro-tumor molecule have
been clearly demonstrated in TNFR1-deficient
mice, which are resistant to tumorigenesis.
The best-characterized mechanism of the
tumor-promoting effects of TNF-α are those
related to the tumor cell itself and molecular
alterations, such as mutation, deletion and
amplification, in key regulatory genes that
lead to the constitutive activation and dereg-
ulated activation of NF-κB. More recently, the
role of non-genetic factors in the overproduc-
tion of TNF-α has been is recognized (31). In
the presence of active NF-κB signaling, TNF-α
and NF-κB interact to induce cytokines: IL-1,
IL-6, COX- 2, adhesion proteins and MMPs.
The high levels of theses inflammatory cyto-
kines trigger uncontrolled NF-κB expression
and activation, preventing the resolution of
the response (33,36).The adaptation to the
local microenvironment stressors is thought
to place a selective pressure on tumor cells
that promotes angiogenesis and the escape of
tumor cells from the toxic environment; two
critical cancer hallmarks of metastasis (31).
Chlamydia and Cervical Cancer 257
Vol. 62(3): 247 - 275, 2021
Along with TNF-α, IL-6 is among the
most commonly over-expressed cytokine
in human tumors (56). IL-6 can act as a
double-edged sword. Induced in response to
injury or infection, IL-6 can induce COX-2
expression and PGE2 synthesis as well as
function in the resolution phase of an acute
response by inhibiting TNFα and IL-1 and
by inducing other anti-inflammatory cyto-
kines such as IL-10. Thus, IL-6 exhibits both
anti- and pro-inflammatory actions at the
site of a wound or lesion (31). In the tumor,
IL-6 has been shown to negatively regulate
apoptotic processes, making cells more re-
sistant to cell death. Two types of IL-6 re-
ceptors, membrane-bound and soluble (57).
The membrane bound IL-6 receptor is pre-
dominantly expressed in hepatocytes, lym-
phocytes, neutrophils, monocyte/macro-
phages and epithelial cells. After binding to
IL-6, the receptor associates with the signal-
transducing protein gp130 to initiate its sig-
naling cascade. The interaction with gp130
promotes a negative feedback loop responsi-
ble for the anti-inflammatory effect of IL-6.
The soluble IL6 receptor (IL-6R) is present
in body fluids and is linked to the inflamma-
tory action of IL-6 in cells not expressing IL-
6R. In this case, the IL-6/IL-6R complex can
bind to gp130, which is expressed in all cell
types, thus explaining the broad spectrum
and systemic action associated with IL-6 in
inflammation (31).
The diverse functions of IL-6 are linked
to interactions across distinct signaling
pathways, including the MAP/STAT pathway
and the AKT/PI3K signaling cascade, which
negatively regulates apoptosis and promotes
cellular proliferation. Recently, IL-6 has
been shown to play a key role in maintaining
the balance between the regulatory subclass
of T cells (Treg) and Th17, an effector T cells
that produces IL-17, IL-6, TNF-α and other
pro-inflammatory chemokines (58). This
function, which is a very important in im-
munity and immune pathology, is linked to
the inflammation process which, when chro-
nicity is maintained, promotes the onset of
malignancies in different organs and that
acts to suppress tumor immune surveillance
and tumor killing through the recruitment
of immunosuppressive myeloid suppressor
cells (59).
Along with IL-6, a number of other
cytokines that participate in inflammation
and present in TAI, have been implicated in
tumor metastasis. In the case of IL-8 and
IL-17 (60), these two pro-inflammatory cy-
tokines have the ability to induce neo-vas-
cularization and to enhance the activity of
the matrix-degrading enzymes MMP-2 and
MMP-9 (61). IL-8, which is also known as
CXCL8, is a potential therapeutic target for
a number of inflammatory diseases given its
critical role in innate immune responses and
as a chemoattractant for neutrophils (31).
The activity of IL-8 is mediated by binding
of monomeric or dimeric forms of CXCL8 to
one of its two receptors CXCR1 and CXCR2.
Expressed normally on the surface of leuko-
cytes, these receptors have also been shown
to be up-regulated on both tumor and tu-
mor-associated stromal cells in a variety
of cancers including lung, prostate and,
colorectal. Via CXCR1/2, IL-8 activates sev-
eral important signaling pathways that are
overactive in tumors (MAPK, PI3K, PKC, FAK
and Src) and which function in tumor cell
proliferation and migration. IL-8 pathway
signaling is induced by a number of factors
including inflammatory cytokines:TNF-α,
IL-1, ROS, and steroid hormones. There is
evidence that IL-8 and CXCR1/2 signaling
are major drivers in chronic inflammation
including TAI.
Like IL-8, the IL-17 molecule is a re-
cently recognized potent pro-inflammatory
cytokine that is produced by the Th17 sub-
population of T lymphocytes and is thought
to be involved in tumorigenesis (60). After
binding to its receptor, IL-17RA, IL-17 acti-
vates the MAPKs ERK1/2 and p38, PI3K/Akt
and NF-κB pathways, leading to the produc-
tion and secretion of IL-1β, IL-6, TNFα and
IL-8, as well as CXCL1 and CXCL6, which
attract neutrophils. The importance of IL-
258 Núñez Troconis
Investigación Clínica 62(3): 2021
17 in tumor development is supported by
observations that inhibition of IL-17 in ani-
mal models of colorectal carcinogenesis pre-
vents tumor formation, an effect that both
prevent the pro-inflammatory response and
the effect of the pro-inflammatory response
on tumor specific immunity (31).
Lipoxygenases and lipoxins
The lipoxygenases/lipoxins products
of polyunsaturated fatty acid metabolism
represent a more recently recognized of
bioactive metabolites in inflammation.
Th 5-lipoxygenase (5-LOX) has been im-
plicated in inflammation-related neopla-
sia. 5-LOX is a non-heme iron dioxygenase
that synthesizes leukotrienes, lipoxins, re-
solvins, and protectins from different sub-
strates belonging to the polyunsaturated
fatty acids (62). The 5-LOX is located
in the cytoplasm or nucleus and is acti-
vated in the nucleus where it translocates
to interact with 5-lipoxygenase activating
protein to mediate the transfer of arachi-
donic acid from the membrane to 5-LOX.
Besides its role in inflammation, the over-
expression of 5-LOX occurs in a number of
tumor tissues and cell lines (63). The final
products of 5-LOX, such as 5-hydroxye-
icosatetraenoic acid and leukotrienes A4
and B4 (LTA4 and LTB4) contribute to
cell survival and growth. The inhibition
of 5-LOX enzymatic activity or the si-
lencing of 5-LOX and leukotriene recep-
tor expression attenuates the metastatic
phenotype in colon cancer cells (64). As
with the COXs, there are anti-proliferative
effects with 5-LOX inhibitors such as AA-
861, zileuton, nordihydroguaiaretic acid
and 5-LOX activating protein inhibitors
such as MK 886, MK 591. These molecules
induce apoptosis in breast (65), leukemia
(66) and pancreatic (67) cell lines. As
such, much like the interest in COX2 and
PGE2, the LOX pathway is emerging as an
important mediator of tumorigenesis with
direct effects on tumor-associated and tu-
mor-promoting inflammation (31).
Chlamydia trachomatis and cervical
cancer
Epidemiology of cervical cancer
In 2018, according to the International
Agency for Research on Cancer/Globocan
(IARC) (68), the cervical cancer (CC) was
the ninth more frequent cancer worldwide in-
cluding both sexes, with 569.847 new cases,
representing an incidence of 3.2% among all
the cancers, and caused 311.365 deaths (in-
cidence: 3.3%), being the ninth type of can-
cer that caused more deaths worldwide (68).
CC represents the second most common
female organ cancer worldwide, after breast
cancer (68). The Pan-American Heath Orga-
nization/World Health Organization (PAHO/
WHO) (69) reported 72,000 new cases of CC
and almost 34,500 deaths in the American
continent during 2018. The mortality rate
was three times higher in Latin-America and
the Caribbean area than in North-America
(69).
Chlamydia trachomatis and inflammation
Ct has been found repeatedly to asso-
ciate with cervical intraepithelial neoplasia
(CIN) and CC (70,71),
although the associa-
tion has commonly been thought to be the
result of confounding by HPV (72). A possible
explanation for the association of Ct and CC
might be that the Ct-induced inflammation
results in an impaired ability to clear HPV
infections (72). The vaginal microenviron-
ment may be considered a co-factor in the
pathogenesis of CIN/CC (73-75). The influ-
ence of different infectious agents and their
association with HPV in cervical carcinogen-
esis has not yet been fully known (76). It is
believed that persistent HPV infection in the
cervical epithelium is facilitated by inflam-
matory processes caused by other STI patho-
gens. Among the main etiological agents re-
sponsible for STDs that may be potentially
involved in cervical carcinogenesis are Ct,
Herpes Simplex Virus (HSV) 1 and 2, Neis-
seria gonorrhoeae (Ng), Mycoplasma geni-
talium (Mg), Trichomonas vaginalis (Tv),
and Treponema pallidum (Tp), which cause
Chlamydia and Cervical Cancer 259
Vol. 62(3): 247 - 275, 2021
inflammatory processes and microabrasion
or microtrauma on the cervical epithelium,
deteriorating the infection scenario and pro-
moting the persistence of HPV (77,78).
Protective immunity to Ct infection is
limited and repeated episodes of infection
are common. It is believed that the inflam-
mation associated with genital tract disease
is immunologically mediated. Persistent an-
tigen synthesis and an ineffective immune
response largely contribute to chronic in-
flammation, tissue damage and immuno-
pathology (79,80). Previous exposure to Ct
offers limited protection against reinfection
because the protection is mostly serovar or
serotype specific and is due to antibody spe-
cific for MOMP, which defines the serovar
o serotype (81). Cell-mediated immunity
has to offer most of the protection against
Ct. Genital infection by Ct and the result-
ing cytokine environment together with the
route of antigen presentation determine the
outcome of infection and disease (82, 83).
The expression of cytokines within the tissue
regulates the recruitment of specific subsets
of lymphocytes to distinct parts of genital
tract (84, 85) so the outcome of genital
chlamydial infection depends on the Ct se-
rovar or serotype and the host immunologi-
cal responses to infection and the balance
between the pathogen specific Th1 and Th2
cell responses (86). While the critical role
of cytokines and lymphocyte subsets recruit-
ment in infection has been reported in dif-
ferent animal models, there is little informa-
tion available in humans about the nature
of immunological events occurring in the
female genital tract following infection with
Ct (83, 85). Local regulation of CD4+ and
CD8 + lymphocytes and the role of Th1/Th2
responses in the genital tract during Ct in-
fection are considered to be crucial for con-
trolling the duration of infection (79).
Different cytokines have been detected
in cervix and fallopian tube in response to Ct
infection; the major cytokines found up-reg-
ulated were: IFN-γ, IL-10, IL-12 and TNF-α.
High levels of IFN-γ have been reported in
the endocervical secretions of Ct positive
women (87). Further, the antichlamydial ac-
tivity of CD4+ and CD8+ T-cells is primarily
associated with the production of high lev-
els of IFN-γ (88-90). Beatty at al. (91) have
reported that INF-γ promotes the destruc-
tion of Ct and also triggers macrophage re-
lease of inflammatory mediators that cause
fibroblast proliferation, thereby enhancing
the synthesis of collagen. In addition, IFN-γ
delays the developmental cycle of Ct so that
chlamydial RBs persist longer, which might
result in persistent unapparent infection
and also, play a role in immunopathogenesis
by promoting inflammatory damage (92).
Reddy et al. (79) reported that Ct infec-
tion also up-regulated the cervical produc-
tion of TNF-α which plays an important role
in the initiation of inflammatory response.
The same authors (79) found a marginal in-
crease in the cervix in their study. It is re-
ported that IL-1 is important both for the
recovery process and for causing inflamma-
tory response (93).
Increased levels of IL-12 have been ob-
served in cervical secretions of Ct positive
women (79). It was suggested that IL-12 is
important for the initial clearance of bacte-
ria (83, 84,94,95). Further, IL-12 is required
for promoting IFN-γ production by NK cells
(96). Phagocytosis of Ct induces dendritic
cells (DCs) to produce IL-12, which in turn
promotes Th-1 response and induces the pre-
sentation of chlamydial antigen to CD4+T-
cells (96), also there is an increase in IL-6
production in the cervix infected by Ct (79).
Levels of Th-2 cytokine, IL-10 has been
found to be high in cervical secretions,
thereby showing that the immune response
in Ct infection of a mixed type, with both
IFN-γ and IL-10 being up regulated. In ad-
dition, IL-10 is not always an inflammatory/
inhibitory cytokine; instead high levels of
IL-10 probably prevent the pathological ef-
fects of the inflammatory cytokines like, IL-
1, IFN-γ, TNF-α, etc. (97).
Ct infects squamous epithelial cells in
the cervix, often inducing an acute inflam-
260 Núñez Troconis
Investigación Clínica 62(3): 2021
matory reaction, followed by lymphocytic in-
filtration and the development of lymphoid
follicles. CD4+ and CD8+ T cell responses
are interdependent and are required for opti-
mal immunity to Ct infection. However, more
studies are needed to define the individual
contribution of CD4+ and CD8+ T-cells in
determining the T-helper cell response (Th1,
Th2 or Th0) during chlamydial pathogenesis
in the human female genitals (79).
Chlamydia trachomatis de-regulated
and damage host DNA
About 15% of human cancers can be
attributed to virus infection, and viruses
are second after tobacco as risk factors for
cancer. In the future, a major proportion
of these infections may be preventable by
immunization, significantly reducing the
worldwide cancer burden. The importance
of the experimental study of tumor viruses
in animals and human is illustrated by the
fact that oncogenes and tumor suppressor
genes were first identified through their in-
teraction with tumor virus proteins (98, 99,
100-102). There are two major mechanisms
by which oncogenic viruses induce tumors
(98,103-106): 1.- direct oncogenesis, the
virus infects a progenitor of the clonal tu-
mor cell population, and usually persists in
the tumor cells and, 2.- indirect oncogenesis
occurs when the virus does not necessarily
infect the tumor progenitor cell, but exerts
an indirect effect on cell and tissue turnover
or in the immune system, predisposing to
tumor development. However, assessing an
infectious etiology can be difficult (99) be-
cause of 1.- subclinical infections are com-
mon and this may lead to misclassification
bias; 2.- complex interactions can result be-
cause many sexually transmitted infections
do occur simultaneously; 3.- the presence of
a latency period between exposure and out-
come, which vary considerably; 4.- clinical
follow-up studies always remain inconclusive
(98,103,104,106-109). It was well known
and established that the human papilloma-
virus (HPV) is the principal etiological agent
in cervical neoplasia (103, 110-118), some
other sexually transmitted organisms may
either contribute to or protect against cervi-
cal carcinogenesis (70, 72,119,120).
It is well known the role that HPV espe-
cially high risk-HPV (hr-HPV) in the carcino-
genesis of CIN/CC. High oncogenic risk HPV
genotypes may infect the epithelium persis-
tently, inducing lesion progression and con-
tributing to carcinogenesis. Research has
shown that the hr-HPV genotypes detected
in carcinoma cases are HPV16, 18, 31, 33,
35, 39, 45, 51, 52, 56, 58, 59, 68, 73, and
82, also, it has been detected 3 HPV geno-
types that should be considered probably
carcinogenic: 26, 53 and 66 (118,121).
The molecular mechanisms underly-
ing cervical carcinogenesis induced by Ct
are not fully understood. Genetic damage
and neoplastic changes induced in vitro,
release of nitric oxide and the inhibition
of host cell apoptosis by blockade of mito-
chondrial cytochrome C release and caspase
activation might account, at least, in part
for such mechanisms (122,123). Chromatin
alterations, such as histone modifications,
may induce somatically heritable changes
of gene activity and thus have oncogenic
potential (124). Histone post-translational
modifications (PTMs) are typically induced
by signal transduction pathways activated in
response to cellular stimuli. One prominent
pathway implicated in histone PTMs is the
mitogen-activated protein kinase (MAPK)
cascade, which leads to histone H3 serine
10 (H3S10) phosphorylation in a promoter-
specific manner, targeting only a subset of
genes (125,126). More recently, the chla-
mydial nuclear effector protein was shown to
have histone methyltransferase activity that
targets histones H2B, H3, and H4 (127).
These data establish that bacterial patho-
gens induce multiple types of histone PTMs,
although the mechanisms and extent of this
phenomenon requires elucidation. (127).
Recently, the role of chromatin and his-
tone modifications in promoting DNA dam-
age responses (DDRs) and genome stability
Chlamydia and Cervical Cancer 261
Vol. 62(3): 247 - 275, 2021
have gained prominence (128). Upon detec-
tion of DNA double-strand breaks (DSBs),
cells activate DDR pathways that detect DNA
lesions and signal their presence by mediat-
ing responses such as cell-cycle arrest, DNA
repair, and, under some circumstances,
apoptosis. Phosphorylation of H2AX Ser139
(gH2AX) is a prominent chromatin modifica-
tion in response to DSBs that acts as a signal
for recruitment of repair proteins including
pATM and 53BP1 to DNA break sites. Defi-
ciencies in DNA damage signaling and repair
pathways lead to genetic instability, which in
turn might enhance oncogenesis (129).
Chumduri et al. (126) showed that Ct
infection of endocervical epithelial cells al-
ters global histone PTMs of host cells. The
infected cells showed a hypo-acetylation
and hipermethylation of lysine residues on
core histones, suggesting that an overall de-
crease chromatin accessibility and a higher
order chromatin structure. Host chromatin
perturbations occurred in the context of in-
fection-induced ROS-mediated DNA damage
and inhibited DDR. Moreover, infected cells
failed to activate cell-cycle checkpoints and
continued to proliferate. Thus, these data
provide evidence of a cellular mechanism
that supports the epidemiological observa-
tions associating Ct infection with cancers
of the female reproductive system.
Histone modifications are increasingly
implicated in DDRs and regulation of ge-
nome stability, in addition to their accepted
roles in transcriptional regulation (128).
Currently, there are a limited number of ex-
amples that describe the ability of bacterial
pathogens to perturb the host epigenome
(130). The prototypical carcinogenic bacte-
rium Helicobacter pylori has been shown to
alter histone H3 phosphorylation in a type-
IV-secretion-system-dependent mechanism
(131). More recently, the chlamydial effec-
tor protein Nue was shown to have histone
methyltransferase activity, which could di-
rectly modify mammalian histones (127).
Chumduri et al. (126) have shown that Ct
infection can lead to extensive alterations of
global host histone PTMs. Specifically, Ct in-
fection consistently increased levels of phos-
phorylated H2AX. gH2AX is a prominent
chromatin modification that is up-regulated
in response to DSB induction and is impor-
tant as a signal for the recruitment of repair
proteins to DSBs. Another histone modifica-
tion that is a constituent of the DNA damage
histone code is phosphorylation of H2B at
Ser14 (132), which was also found to be mod-
ulated in Ct-infected cells. Phosphorylation
of H4 at Ser1 (pH4Ser1) has been implicated
in the restoration of chromatin structure by
preventing re-acetylation and, thereby, shut-
ting down DNA damage signaling after DNA
repair (133). Ct infection induced elevated
levels of the phosphorylated form of H4Ser1
during acute infection, which might reflect
its role in DNA repair activities. Chumduri
et al. (126) have observed decreased levels
of H3K9Ac and H3K56Ac in infected cells,
which have previously been shown to be re-
duced in response to DNA damage in human
cells (134).
Chumduri et al. (126) have provided a
comprehensive analysis of global changes
to host chromatin induced upon infection
with a bacterial pathogen. They (126) have
shown that chlamydial infection alters his-
tone PTMs, leading to four distinct patterns
of histone marks, which vary between acute
and persistent infections. Among others,
gH2AX and H3K9me3, hallmarks of DSBs and
senescence associate to heterochromatin
foci (SAHF), respectively, showed sustained
up-regulation during Ct infection. Reactive
oxygen species induced by Ct were found to
contribute to persistent DSBs. SAHF forma-
tion was selectively induced in an ERK- de-
pendent manner in response to Ct-induced
DSBs, in contrast to DNA damage induced
by etoposide. They (126) demonstrated that
Ct infection suppressed DNA damage repair
activities despite the presence of extensive
DSBs in host cells. Infected cells containing
DSBs continued to proliferate, facilitated by
SAHF formation. The same authors (126)
have reported that Ct infection causes DSB
262 Núñez Troconis
Investigación Clínica 62(3): 2021
generation, which could predispose host
cells to genomic instability and transforma-
tion via the unusual combination of impaired
repair and pro-survival signaling.
According to Chamduri et al. (126) Ct
modulates host cell function in ways that
convey benefits to the pathogen but have se-
vere consequences for the fate of host cells.
These may be relevant during chronic in-
fection. Ct induces the formation of DSBs,
which leads to the induction of gH2AX. How-
ever, the induction of gH2AX is not followed
by a DDR that would either stimulate the
induction of appropriate repair processes
or lead to cellular senescence or apoptosis.
Instead, infection promotes cellular viability
by limiting the extent of DNA damage signal-
ing. Together, the orchestrated deregulation
of host cell signaling and perturbations to
host cell chromatin lead to the enforced sur-
vival of damaged host cells, which is likely to
predispose them to transformation.
Chlamydia trachomatis and human
papillomavirus
The pathogenesis of Ct in Cervical In-
traepithelial Neoplasia (CIN)/CC remains
unknown; however, different authors (135-
137) suggest that Ct may be involved in
cervical carcinogenesis. The cervical meta-
plasia induced by Ct can provide target cell
for acquisition of HPV, especially hr-HPV
(135,138). On the other hand, by causing
local immune-perturbation because it may
interfere with the immune surveillance of
HPV infections, especially hr-HPV types, in
patients with persistent or chronic infection
of Ct (119,139). These two alternatives are
supported by studies that show that Ct in-
fection is a risk factor for the new and per-
sistence of HPV DNA (72,140). However, evi-
dence of Ct infection increasing the risk of
the development of CIN/CC, among those
with or without hr-HPV infection at the base-
line, is still missing (119,122).
As it was mentioned before, it is well
known that Ct causes cervicitis and endocer-
vicitis, which becomes a chronic infection at
the endocervical cells of the transformation
zone. Such inflammation may predispose
women to other STDs, including HPV, HIV,
HSV 1 and 2, Ng, Mg, Tv and, Tp infection
by damaging epithelial integrity (120,141).
Different studies have suggested that a Ct in-
fection is associated with a persistence of hr-
HPV infection (72,140) and persistent HPV
infections are necessary for progression to
high-grade CIN and CC (142,143). So it is
believed that chronic cervical inflammation
by Ct could increase the risk of transforma-
tion of cervical cells that are persistently in-
fected with oncogenic types of HPV (120).
Several factors affect cervical carci-
nogenesis, from behavioral variables to the
presence of infectious agents linked with
STIs. This is true especially for high carcino-
genesis risk genotypes of the HPV (123,144).
There is a huge amount of reports that
evidence the incidence of HPV is higher in
women with secondary genital infections.
The vaginal microenvironment may be con-
sidered a co-factor in the pathogenesis of
CIN/CC (145,146). The influence of differ-
ent infectious agents and their association
with HPV in cervical carcinogenesis has not
yet been fully explained. It is believed that
inflammatory processes caused by other STI
pathogens facilitate persistent HPV infection
in the cervical epithelium. Among the main
etiological agents responsible for STIs that
may be potentially involved in cervical carci-
nogenesis are Ct, HSV 1 and 2, Neisseria Gn,
Mg, Tv, and Tp, which cause inflammatory
processes and micro-abrasion or micro-trau-
ma on the cervical epithelium, deteriorating
the infection scenario and promoting the
persistence of HPV (78,147). Ct has been
found repeatedly to associate with CIN/CC
in several cross-sectional case-control stud-
ies (148,149), although the association has
commonly been thought to be the result of
confounding by HPV. During recent years, an
association with Ct has also been found in
several biobank-based longitudinal studies
with invasive CC (72,150-154). A possible
explanation for the association of Ct and CC
Chlamydia and Cervical Cancer 263
Vol. 62(3): 247 - 275, 2021
might be that the Ct-induced inflammation
results in an impaired ability to clear HPV
infections (72).
The hypothesis establishes that the epi-
thelial cells infected by Ct become suscep-
tible to infection with hr-HPV and the syner-
gistic actions of the two infectious organisms
lead to development of neoplasia (155,156).
Persistent and recurrent Chlamydial infec-
tion, liberation of cytotoxic substances like
nitric oxide as well as anti-apoptotic mecha-
nisms come into play resulting in prolifera-
tion of damaged cells and initiating carcino-
genesis, the co-factor role of the organism
in HPV associated cervical lesions can be at-
tributed to immune-modulation (151). As a
result of a disturbances and under the influ-
ence of persistent infection, the cells escape
of the control of the cell signaling mecha-
nisms, DNA damage occurs leading to pro-
liferation of clones of cells carrying altered
genetic material with enhanced propensity
for neoplastic change (126). The sites of in-
fection by Ct are columnar epithelial cells of
the endocervix as is evident by the increased
prevalence of infection in cases with cervi-
cal ectropion; regions of squamous meta-
plasia of cervix are increasingly infected by
Ct for the high prevalence of squamous cell
carcinoma in association with the infection
(153,157). When CT infects a cell, entry of
HPV to the basal layer is facilitated by mi-
croscopic epithelial injuries, micro-trauma,
or micro-abrasion. HPV viral particles accu-
mulate and derangement of host immunity
occurs, which is manifested by shift of the
immune response from T-helper cell type
1(active in HPV control) to T-helper cell type
2 and plasma cell infiltrates (140).
Several authors (72,76,158,159) have
reported the relation between Ct and HPV
infection in the development and evolution
of CIN/CC. As it was mentioned before, Ct
infection may play a major role in the eti-
ology of CIN/CC by facilitating hr-HPV en-
trance and persistence. Probably, this is due
to the chronic inflammation induced by the
bacteria, and to the resistance to cell apop-
tosis that persistent Ct infections appear to
confer. Deluca et al. (158) say that the asso-
ciation between these two agents seems to
be more related to a combined potentiation
than to the fact that they share a common
route of transmission (160). Controversial
and discordant information exists on this
topic, and since the role of Ct in the natural
history of HPV infection is not sufficiently
clear, this particular issue merits further
study (158). Also, Gopalkrishna et al. (159)
mentioned that if there is a synergistic effect
when both Ct and HPV infection are present
in comparison with infection of HPV alone
during development of cervical cancer and
they found a slightly higher rate of chlamyd-
ial infection in patients with CC when they
are compared with control patients. That
indicates that Ct may play a role as a co-fac-
tor with regard to the pathological aggres-
siveness of the disease and the Ct chronic
infection in conjunction with HPV may be
a more pertinent factor mediating CC risk
(161). Although a carcinogenic interaction
between Ct and HPV has not been directly
demonstrated, in vitro data show that Ct
may inhibit cell apoptosis (162), a contribu-
tory element for carcinogenesis. Alternative-
ly, inflammatory cytokine responses during a
chlamydial infection may produce ROS that
might cause DNA damage or modification,
providing a mechanistic link between chron-
ic inflammation and malignant transforma-
tion (163). Other bacterial or parasitic in-
fections causing chronic inflammation have
also been implicated in human cancer, such
as Helicobacter pylori with stomach cancer
and Schistosoma haematobium with bladder
cancer. Smith et al. (161) based on a large
number of newly diagnosed CC patients,
consistently indicates a potential etiologic
role for Ct infection as an HPV cofactor in
the development of squamous CC.
In a meta-analysis, Zhu et al. (164)
found evidences that the chlamydial infec-
tion could be one of the risk factor of cervi-
cal cancer. Individuals infected with Ct have
a heightened risk of developing CC. There-
264 Núñez Troconis
Investigación Clínica 62(3): 2021
fore, it is necessary to expand Ct infection
screening and treat women with Ct infection
timely, particularly among women at a high-
er risk of HPV infections. This approach will
not only protect against PID and infertility,
but potentially also prevent cervical cancer
and reduce the incidence of CC.
The questions of which of the two infec-
tions, HPV and Ct: 1.- which of the two infec-
tion has to occur first; 2.- if Ct infection in-
creases the risk of acquiring HPV. Lehtinen
et al. (119) found that Ct associated relative
risk (RR) of developing CIN was statistically
significant comparable, both before and af-
ter the acquisition of HPV16/18, which sug-
gests that the order of the two infections is
not important in cervical carcinogenesis.
Several cohort studies have indicated that
Ct exposure increases the tendency for HPV
infection to associate an increased risk for
CC (72,140,165,166). An analysis of the
HPV-stratified and HPV-adjusted risk esti-
mates obtained both in the univariate and
multivariate analyses, suggests that the Ct
associated RR of developing CIN/CC is com-
parable both before and after acquisition of
HPV. Furthermore, no interaction between
the two microorganisms was observed. Thus,
Ct may or could not act in the cervical car-
cinogenesis by promotion of persistent HPV
infection (161). Lehtinen et al. (119) found
that Ct infection plays an independent co-
factor role in the development of cervical
neoplasia (167,168), the effect is likely to
take place at an early stage of cervical carci-
nogenesis. Lehtinen et al. (119) mentioned
that Ct infection might facilitate the devel-
opment of early cervical lesions. On the oth-
er hand, a proportion of CIN lesions regress
spontaneously. This and the fact that Ct in
other longitudinal studies have been associ-
ated with early stages of lesions developing
cervical cancer
suggest that Ct may have an
early role in cervical carcinogenesis in a pro-
portion of cases. The role of pathobiology is
a possible mechanism, however, this remains
open (151,152 154,157,169).
Anttilla et al. (170) found in a longitu-
dinal sero-epidemiologic study of an associa-
tion between exposure to specific serotypes
or serovars of Ct and CC, especially the
squamous (S) histological type. They (170)
reported that the presence of serum IgG
antibodies to Ct serotype or serovar G was
associated with the highest risk. Also, immu-
noglobulin G antibodies to more than one
serotype or serovar of Ct increased the risk
for subsequent development of SCC. Distri-
bution of the genital serotypes or serovars
varies from one geographic area to another,
suggesting that some serotypes have biologi-
cal advantages over others in defined popu-
lations (171). Serotypes or serovars D and
E are approximately 50% of all isolates, fol-
lowed by F and G serotypes or serovars which
represent 15% to 40%; other serotypes or se-
rovars represent less than 10% each. Sero-
types E and G have been found more often
in women than men, whereas serotype D has
been found more frequently in men than in
women (172-176). Lethinen et al. (168) re-
ported that a pool of GFK serotypes or se-
rovars was more common found in patients
with SCC than in control women.
Barnes et al. (177) mentioned that the
presence of mixed infections implies that
infection with one serotype does not induce
protective immunity against subsequent
infections caused by another serotype or
serovar. Antilla et al. (170) reported that
multiple exposures might increase the risk
of acquiring infections caused by the cancer-
associated serotypes. Therefore, antibodies
to multiple serotypes detected in patients
with cervical SCC may also suggest chronic
infection by a single serotype but they could
not distinguish between both possibilities.
Finally, a carcinogenic interaction be-
tween Ct and HPV has not been directly
demonstrated, in vitro data show that Ct
may inhibit cell apoptosis (161,178), a con-
tributory element for carcinogenesis. Alter-
natively, inflammatory cytokine responses
during a chlamydial infection may produce
Chlamydia and Cervical Cancer 265
Vol. 62(3): 247 - 275, 2021
ROS that might cause DNA damage or modi-
fication, providing a mechanistic link be-
tween chronic inflammation and malignant
transformation (163).
CONCLUSION
Further epidemiological studies are
needed to clarify the role of Ct in the etiol-
ogy of CC. Additional prospective data are
needed on the induction of inflammation by
Ct and other STIs and on the effect of their
relative timing, in conjunction with HPV in-
fection, on the risk of cervical neoplasia, in
addition to the effect of the treatment of Ct
infection on the progression of cervical neo-
plasia. Studies on a large number of newly
diagnosed CC patients, consistently indicate
a potential etiologic role for Ct infection as
an HPV cofactor in the development of squa-
mous CC (161). Beyond potentially aiding
the establishment or progression of HPV in-
fections, different authors (120,170) specu-
late that the inflammatory response and
metaplasia triggered by Ct infection may
encourage cell turnover and therefore the
number of non-dividing differentiating cells
that are needed for HPV replication and pro-
ductive HPV infections. Further, persistent
Ct infections may create an inflammatory
environment conductive to HPV-induced
carcinogenesis by increasing the chance of
DNA replication errors that have been shown
in vitro to lead to persistent disease and ac-
cumulation of genetically damaged cells.
As it is well known that, the development
of CC takes several years or decades. The link
between bacterial infections and carcinogen-
esis is not clear, but genetic damage and neo-
plastic changes can be induced in vitro by co-
culturing cells with activated inflammatory
cells (179). As it was mentioned before in the
manuscript, during a Ct infection, nitric ox-
ide is released (1) and is able to inhibit host
cell apoptosis (178). In a chronic chlamydial
infections, these mechanisms could initiate
or promote cervical carcinogenesis. The fact
that the exposure to a specific serotype or to
more than one serotype or serovar increases
the evidence for the role of Ct in cervical car-
cinogenesis by itself (170).
Future and further studies should address
the question of whether Ct plays as a co-factor
role or as an independent factor role in the car-
cinogenesis of CC; this effect is likely to take
place at an early stage of cervical carcinogen-
esis and/or restricted to some cases only.
REFERENCES
1. O’Connell CM, Ferone ME. Chlamydia tra-
chomatis genital Infections. Microb Cell.
2016;3(9):390-403. doi:10.15698/mic
2016. 09.525.
2. BéBéar C, de Barbe B. Genital Chlamydia
trachomatis infections. Clin Microbien In-
fect 2009:15(1):4-10.
3. Workowski KA, Bolan GA. Centers for
Disease Control and Prevention. Sexually
transmitted diseases treatment guideli-
nes, 2015. MMWR Recomm Rep. 2015 Jun
5;64(RR-03):1-137. Erratum in: MMWR Re-
comm Rep 2015 Aug 28;64(33):924.
4. Rowley J, Vander Hoorn S, Korenromp E,
Low N, Unemo M, Abu-Raddad LJ, Chico
RM, Smolak A, Newman L, Gottlieb S,
Thwin SS, Broutet N, Taylor MM. Chla-
mydia, gonorrhoea, trichomoniasis and
syphilis: global prevalence and incidence
estimates, 2016. Bull World Health Or-
gan. 2019 1;97(8):548-562. doi: 10.2471/
BLT.18.228486. Epub 2019 Jun 6.
5. Redmond SM, Alexander-Kisslig K, Wood-
hall SC, van den Broek IVF, van Bergen
J, Ward H, Uusküla A, Herrmann B, An-
dersen B, Götz HM, Sfetcu O, Low N.
Genital chlamydia prevalence in European
non-European high income countries: sys-
tematic review and meta-analysis. PLoS
One 2015;10(1):e0115753. doi:10.1371/
journal.pone.0115753.
6. Núñez-Troconis J, Gallegos B, Noriega C.
Incidencia de la Chlamydia trachomatis
en pacientes con esterilidad. Invest. Clin
1990;31(2): 91-104.
7. Núñez-Troconis J. Investigación de tres pa-
tógenos de transmisión sexual en diferen-
tes patologías ginecológicas. Rev Obstet
Ginecol Venez 1998;58(3):175-185.
266 Núñez Troconis
Investigación Clínica 62(3): 2021
8. Vivas K, Albarracin L, Ruiz E, Téllez, Mo-
reno Y, Noguera MU, Monsalve N, Mendoza
JA. Infección por virus del papiloma huma-
no y Chlamydia trachomatis y su implica-
ción en el desarrollo de las lesiones cervica-
les en mujeres de la ciudad de Mérida. Rev
Obstet Ginecol. 2018:78(2):113-121.
9. Albritton HL, Kozlowski PA, Lillis RA,
McGowan Cl, Siren D, Taylor SN, Iba-
na JA, Buckner LR, Shen L, Quayle AJ.
A novel whole-bacterial enzyme linked-
immunosorbant assay to quantify Chla-
mydia trachomatis specific antibodies
reveals distinct differences between syste-
mic and genital compartments. PLoS One.
2017;12(8):e0183101. doi: 10.1371/jour-
nal.pone.0183101
10. Molano M, Weiderpass E, Posso H, Mo-
rré SA, Ronderos M, Franceschi S, Arsian
A, Meijer CJLM, Miñoz N, van den Bru-
le AJC, HPV study group. Prevalence and
determinants of Chlamydia trachomatis in-
fections in women from Bogota, Colombia.
Sex Transm Infect. 2003;79(6):474-478.
doi:10.1136/sti.79.6.474.
11. Malhotra M, Sood S, Mukherjee A, Murali-
dhar S, Bala M. Genital Chlamydia tracho-
matis: an update. Indian J Med Res. 2013
Sep;138(3):303-316. PMID: 24135174; PM-
CID: PMC3818592.
12. Ward ME, Ridgway G. Chlamydia. In: Co-
llier L, Balows A, Sussman A editors. Topley
and Wilsons Microbiology and microbiology
infection. 9
th
edition. New York Press Inc.
1999. p. 1331-1336.
13. Novak M, Novak D. Risk factors for Chla-
mydia trachomatis infection among users
of an internet-based testing service in Swee-
den. Sex Reprod Health 2013; 4: 23-27.
14. Stamm WE, Batteiger BE. Chlamydia tra-
chomatis (Trachoma, Perinatal Infections,
Lymphogranuloma Venerum and other Ge-
nital Infections). 7th
edition. Mandell GL,
Bennett JE, Dolin R, editors. Philadelphia:
Churchill Livingstone Elsevier; 2010.
15. Wyrick PB. Chlamydia trachomatis persisten-
ce in vitro: An overview. J Infect Dis. 2010;
201 Suppl 2: S88–S95. Epub 2010 May 28.
16. Lambden PR, Pickett MA, Clarke IN. The
effect of penicillin on Chlamydia trachoma-
tis DNA replication. Microbiol 2006; 152(Pt
9): 2573- 2578.
17. Elwell C, Mirrashidi K, Engel J. Chlamydia
cell biology and pathogenesis. Nat Rev Mi-
crobiol. 2016;14(6):385-400. doi: 10.1038/
nrmicro.2016.30. Epub 2016 Apr 25. PMID:
27108705; PMCID: PMC4886739.
18. Hussen S, Wachamo D, Yohannes Z, Ta-
desse E. Prevalence of Chlamydia tracho-
matis infection among reproductive age
women in sub Saharan Africa: a systematic
review and meta-analysis. BMC Infect Dis.
2018;18(1):596. doi:10.1186/s12879-018-
3477.
19. Rogstad K. Complications in the female
and their management. In: Moss T, ed. In-
ternational handbook of Chlamydia, 3rd
edition. Haslemere, UK: Alden Press, 2008;
p. 111–121.
20. Peipert JF. Clinical practice. Genital chla-
mydial infections. N Engl J Med 2003; 349:
2424–2430.
21. Paavonen J, Eggert-Kruse W. Chlamydia
trachomatis: impact on human reproduc-
tion. Hum Reprod Update 1999; 5: 433–
447.
22. Hicks D. Complications of Chlamydia tra-
chomatis infection in men. In: Moss TR, ed.
International handbook of Chlamydia, 3rd
edition. Haslemere, UK: Alden Press, 2008;
p. 99–109.
23. Barbeyrac de B, Papaxanthos Roche A,
Mathieu C, Germain C, Brun JL, Gachet
M, Mayer G, Bébéar C, Chene G, Hock
C. Chlamydia trachomatis in subfertile
couples undergoing an in vitro fertiliza-
tion program: a prospective study. Eur J
Obstet Gynecol Reprod Biol 2006; 129:
46–53. PMID: 16701936 doi: 10.1016/j.ejo-
grb.2006.02.014.
24. Malinverni R. The role of cytokines in chla-
mydial infections. Curr Opin Infect Dis
1996; 9: 150-155.
25. Morton RS, Kinghorn GR. Genitourinary
chlamydial infection: a reappraisal and hy-
pothesis. Int J STD AIDS 1999; 10 : 765-75.
26. Rasmussen SJ, Eckmann L, Quayle AJ,
Shen L, Zhang Y, Anderson DJ, Fierer J,
Stephens RS, Kagnoff MF. Secretion of
pro-inflammatory cytokines by epithelial
cells in response to Chlamydial infection
suggests a central role for epithelial cells
in chlamydial pathogenesis. J Clin Invest
1997; 99: 77-87.
Chlamydia and Cervical Cancer 267
Vol. 62(3): 247 - 275, 2021
27. Morrison SG, Morrison RP. A predominant
role of antibody in acquired immunity to
chlamydial genital tract reinfection. J Im-
munol 2005; 73: 6183-6186.
28. Haggerty CL, Gottlieb SL, Taylor BD, Low
N, Xu F, Ness RB. Risk of sequelae after
Chlamydia trachomatis genital infection in
women. J Infect Dis 2010; 201 (Suppl 2):
S134-S155.
29. Zdrodowska-Stefanow B, Ostaszewska-
Puchalska I, Pucilo K. The immunology of
Chlamydia trachomatis Arch Immunol Ther
Exp 2003; 51: 289-294.
30. Burstein GR, Gaydos CA, Diener-West M.
Incidence of Chlamydia trachomatis infec-
tion among inner-city adolescents. JAMA
1998; 280: 521-526.
31. Thompson PA, Khatami M, Baglole CJ,
Sun J, Harris SA, Moon EY, Al-Mulla F, Al-
Temaimi R, Brown DG, Colacci A, Mon-
dello C, Raju J, Ryan EP, Woodrick J,
Scovassi AI, Singh N, Vaccari M, Roy R,
Forte S, Memeo L, Salem HK, Amedei A,
Hamid RA, Lowe L, Guarnieri T, Bisson
WH. Environmental immune disruptors,
inflammation and cancer risk. Carcinoge-
nesis. 2015;36(Suppl 1):S232-S253. doi:
10.1093/carcin/bgv038. PMID: 26106141;
PMCID: PMC4492068.
32. Khatami, M. Chronic inflammation: syner-
gistic interactions of recruiting macropha-
ges (TAMs) and eosinophils (Eos) with host
mast cells (MCs) and tumorigenesis in
CALTs. M-CSF, suitable biomarker for cancer
diagnosis! Cancers (Basel) 2014;6:297–322.
33. Colotta F, Allavena P, Sica A, Garlanda
C, Mantivani A. Cancer-related inflam-
mation, the seventh hallmark of cancer:
links to genetic instability. Carcinogenesis
2009;30:1073–1081.
34. Hanahan, D, Weinberg RA. The hallmarks
of cancer: the next generation. Cell 2000;
100:57–70.
35. Khatami, M. ’Yin and Yang’ in inflam-
mation: duality in innate immune cell
function and tumorigenesis. Expert Opin.
Biol. Ther 2008;8:1461–1472.
36. Candido J, Hageman T. Cancer-related
inflammation. J. Clin. Immunol 2013;33
(suppl. 1): S79–S84.
37. Costa A, Scholer-Dahirel A, Mecha-Grego-
riou F. The role of reactive oxygen species
and metabolism on cancer cells and their
microenvironment. Semin. Cancer Biol
2014;25: 23–32.
38. Khatami M. Unresolved inflammation: ‘im-
mune tsunami’ or erosion of integrity in
immune-privileged and immune-responsive
tissues and acute and chroniinflammatory
diseases or cancer. Expert Opin. Biol. Ther
2011;11:1419–1432.
39. Roberts RA, Smith RA, Safe S, Szabo C,
Tjalkens RB, Robertson FM.. Toxicologi-
cal and pathophysiological roles of reacti-
ve oxygen and nitrogen species. Toxicology
2010;276:85–94.
40. Naik E, Dixit VM. Mitochondrial reac-
tive oxygen species drive proinflamma-
tory cytokine production. J. Exp. Med
20111;208(3):417–420.
41. Kongara S, Karantza V. The interplay
between autophagy and ROS in tumori-
genesis. Front Oncol. 2012 Nov 21;2:171.
doi: 10.3389/fonc.2012.00171. PMID:
23181220; PMCID: PMC3502876.
42. Danelli L, Frossi B, Sri G, Mion F, Guar-
notta L, Tripodo C, Marluzzi L, Marzi-
notto S, Rigoni A, Blank U, Colombo
MP, Pucillo CE. Mast cells boost myeloid-
derived suppressor cell activity and contri-
bute to the development of tumor-favoring
microenvironment. Cancer Immunol. Res.
2015;3(1):85–95. doi: 10.1158/2326-6066.
CIR-14-0102. Epub 2014 Oct 28.
43. Grimm EA, Sikora AG, Ekmekcioglu
S. Molecular pathways: inflammation-
associated nitric-oxide production as a
cancer-supporting redox mechanism and
a potential therapeutic target. Clin Can-
cer Res 2013;19(20):5557-5563. doi: 10.
1158/1078-0432.CCR-12-1554.
44. Moran EM. Epidemiological and clinical
aspects of nonsteroidal anti-inflammatory
drugs and cancer risks. J. Environ. Pathol.
Toxicol. Oncol 2002;21:193–201.
45. Wakabayashi K. NSAIDs as cancer pre-
ventive agents. Asian Pac. J. Cancer Prev
2000:1:97–113.
46. Baglole CJ, Ray DM, Bernstein SH, Feldon
SE, Smith TJ, Sime PJ, Phipps RP. More
than structural cells, fibroblasts create and
orchestrate the tumor microenvironment.
Immunol. Invest. 2006;35(3-4):297-325.
doi: 10.1080/08820130600754960.
268 Núñez Troconis
Investigación Clínica 62(3): 2021
47. Nataraj C, Thomas DW, Tilley SL, Nguyen
MT, Mannon R, Koller BH, Coffman YM.
Receptors for prostaglandin E2 that regu-
late cellular immune responses in the mou-
se. J. Clin. Invest. 2001;108(8):1229–1235.
doi: 10.1172/JCI13640.
48. Tilley SL, Coffman TM, Koller BH. Mi-
xed messages: modulation of inflam-
mation and immune responses by prosta-
glandins and thromboxanes. J Clin Invest
2001;108(1):15-23. doi:10.1172/JCI1341.
49. Zhu Z, Zhong S, Shen Z. Targeting the
inflammatory pathways to enhance che-
motherapy of cancer. Cancer Biol Ther
2011;12(2):95-105. doi:10.4161/cbt. 12.2.
15952.
50. Bollrath J, Greten FR. IKK/NF-kappaB
and STAT3 pathways: central signalling
hubs in inflammation-mediated tumour
promotion and metastasis. EMBO Rep
2009;10(12):1314-1319. doi:10.1038/em-
bor.2009.243.
51. Nguyen DP, Li J, Tewari AK. Inflammation
and prostate cancer: the role of interleukin
6 (IL-6). BJU Int 2014;113(6):986-992.
doi:10.1111/bju.12452.
52. Zhang HF, Lai R. STAT3 in Cancer-Friend
or Foe? Cancers (Basel) 2014;6(3):1408-
1440. doi:10.3390/cancers6031408.
53. He G, Karin M. NF-κB and STAT3 - key play-
ers in liver inflammation and cancer. Cell
Res 2011;21(1):159-168. doi:10.1038/cr.
2010.183.
54. Feldmann M, Maini SR. Role of cytokines
in rheumatoid arthritis: an education in
pathophysiology and therapeutics. Im-
munol Rev 2008; 223:7-19. doi:10.1111/
j.1600-065X.2008.00626.x
55. Croft M. The TNF family in T cell differen-
tiation and function--unanswered questions
and future directions. Semin Immunol
2014;26(3):183-190. doi:10.1016/j.smim.
2014.02.005.
56. Ataie-Kachoie P, Pourgholami MH, Rich-
ardson DR, Morris DL. Gene of the
month: Interleukin 6 (IL-6). J Clin Pathol
2014;67(11):932-937. doi:10.1136 jclin-
path-2014-202493.
57. Rose-John S, Scheller J, Elson G, Jones
SA. Interleukin-6 biology is coordinated by
membrane-bound and soluble receptors:
role in inflammation and cancer. J Leukoc
Biol 2006;80(2):227-236. doi:10.1189/jlb.
1105674.
58. Taniguchi K, Karin M. IL-6 and related
cytokines as the critical lynchpins be-
tween inflammation and cancer. Semin
Immunol 2014;26(1):54-74. doi:10.1016j.
smim.2014.01.001.
59. Tsukamoto H, Nishikata R, Senju S,
Nishimura Y. Myeloid-derived suppressor
cells attenuate TH1 development through
IL-6 production to promote tumor progres-
sion. Cancer Immunol Res 2013;1(1):64-
76. doi:10.1158/2326-6066.CIR-13-0030.
60. Zarogoulidis P, Katsikogianni F, Tsiouda
T, Sakkas A, Katsikogiannis N, Zarogouli-
dis K. Interleukin-8 and interleukin-17 for
cancer. Cancer Invest 2014;32(5):197-205.
doi: 10.3109/07357907.2014.898156.
61. Campbell LM, Maxwell PJ, Waugh DJ.
Rationale and Means to Target Pro-Inflam-
matory Interleukin-8 (CXCL8) Signaling
in Cancer. Pharmaceuticals (Basel) 2013;
6(8):929-59. doi: 10.3390/ph6080929.
PMID: 24276377; PMCID: PMC3817732.
62. Schneider C, Pozzi A. Cyclooxygenases and
lipoxygenases in cancer. Cancer Metastasis
Rev 2011; 30(3-4):277-94. doi: 10.1007/
s10555-011-9310-3. PMID: 22002716; PM-
CID: PMC3798028.
63. Greene ER, Huang S, Serhan CN, Pani-
grahy D. Regulation of inflammation in
cancer by eicosanoids. Prostaglandins Oth-
er Lipid Mediat 2011;96(1-4):27-36. doi:
10.1016/j.prostaglandins.2011.08.004.
Epub 2011 Aug 16. PMID: 21864702; PM-
CID: PMC4051344.
64. Bishayee K, Khuda-Bukhsh AR. 5-lipoxy-
genase antagonist therapy: a new approach
towards targeted cancer chemotherapy.
Acta Biochim Biophys Sin (Shanghai).
2013;45(9):709-719. doi: 10.1093/abbs/
gmt064.
65. Gilmartin AG, Faith TH, Ritcher M, Groy
A, Seefeld MA, Darcy MG, Peng X, Federo-
wicz K, Yang J, Zhang SY, Minthon E, Ja-
worski JP, Schaber M, Marlens S, McNully
DE, Sinnamon RH, Zhang H, Kirkpatrick
RB, Nevins N, Cui G, Pietrak B, Diaz E,
Jones A, Brandt M, Schwartz B, Heerding
DA, Kumar R. Allosteric Wip1 phosphatase
inhibition through flap subdomain interac-
tion. Nat. Chem. Biol 2014;10(3):181-187.
Chlamydia and Cervical Cancer 269
Vol. 62(3): 247 - 275, 2021
doi: 10.1038/nchembio.1427. Epub 2014
Jan 5.
66. Datta K, Biswal SS, Kehrer JP. The 5-li-
poxygenase-activating protein (FLAP) in-
hibitor, MK886, induces apoptosis indepen-
dently of FLAP. Biochem J 1999;340 (Pt 2)
(Pt 2):371-375. PMID: 10333477; PMCID:
PMC1220259.
67. Schuller HM, Zhang L, Weddle DL, Cas-
tonguay A, Walker K, Miller MS. The cyclo-
oxygenase inhibitor ibuprofen and the FLAP
inhibitor MK886 inhibit pancreatic carci-
nogenesis induced in hamsters by transpla-
cental exposure to ethanol and the tobacco
carcinogen NNK. J Cancer Res Clin On-
col 2002;128(10):525-532. doi:10.1007/
s00432-002-0365-y
68. International Agency for Research on
Cancer. World Health Organization. Can-
cer 2018 Available at: https://gco.iarc.fr/
today/data/factsheets/populations/900-
world-fact-sheets.pdf. Consulted on
06/20/2020.
69. Pan-American Health Organization. Cáncer
Cervicouterino 2018. Available at: https://
www.paho.org/hq/index.php?option=com_
content&view=article&id=5420:2018-
cervical-cancer&Itemid=3637&lang=es.
Consultred on: 06/20/2020.
70. Smith JS, Bosetti C, Munoz N, Herrero
R, Bosch FX, Eluf-Neto J, Meijer CJ, Van
Den Brule AJ, Franceschi S, Peeling RW.
Chlamydia trachomatis and invasive cervi-
cal cancer: A pooled analysis of the IARC
multicentric case-control study. Int J Can-
cer 2004;111: 431–439.
71. Schachter J, Hill EC, King EB, Coleman
VR, Jones P, Meyer KF. Chlamydial infec-
tion in women with cervical dysplasia. Am J
Obstet Gynecol 1975;123:753–757.
72. Silins
I, Ryd W, Strand A, Wadell G, Tom-
berg S, Hansson BG, Wang X, Arnheim L,
Dahl L, Dahl V, Brenell D, Parson K, Die-
ner J, Rylander E. Chlamydia trachomatis
infection and persistence of human papil-
lomavirus. Int J Cancer 2005;116(1):110-
115. doi:10.1002/ijc.20970.
73. Murta EFC, de Souza MAH, Adad SJ,
Araújo Jr E. Infecção pelo papilomavírus
humano em adolescentes: relação com o
método anticoncepcional, gravidez, fumo e
achados citológicos. Rev Bras Ginecol Obs-
tet 2001;23: 217-221.
74. Martins MCL, Bôer CG, Svidzinski TIE,
Estivalet TI, Donida LG, Martins PFA,
Boscoli FNS, Consolaro MEL. Avaliação do
método de Papanicolaou para triagem de al-
gumas infecções cérvico-vaginais. Rev Bras
Anal Clin 2007;39: 217-221.
75. Campos ACC, Freitas-Júnior R, Ribeiro
LFJ, Paulinelli RR, Reis C. Prevalence of
vulvovaginitis and bacterial vaginosis in pa-
tients with koilocytosis. Sao Paulo Med J
2008;126: 333-336.
76. Wohlmeister D, Vianna DR, Helfer VE,
Gimenes F, Consolaro ME, Barcellos RB,
Rossetti ML, Calil LN, Buffon A, Pilger
DA. Association of human papillomavirus
and Chlamydia trachomatis with intrae-
pithelial alterations in cervix samples. Mem
Inst Oswaldo Cruz 2016 Feb;111(2):106-
113. doi: 10.1590/0074-02760150330.
Epub 2016 Feb 2. PMID: 26841046; PM-
CID: PMC4750450.
77. Muvunyi CM, Dhont N, Verhelst R, Cru-
citti T, Reijans M, Mulders B, Simons
G, Temmerman M, Claeys G, Padalko E.
Evaluation of a new multiplex polymerase
chain reaction assay STDFinder for the si-
multaneous detection of 7 sexually trans-
mitted disease pathogens. Diagn Microbiol
Infect Dis 2011; 71: 29-37.
78. Rodríguez-Cerdeira C, Sánchez-Blanco E,
Alba A. Evaluation of association between
vaginal infections and high-risk human pa-
pillomavirus types in female sex workers in
Spain. ISRN Obstet Gynecol 2012: 240190.
doi:10.5402/2012/240190.
79. Reddy BS, Rastogi S, Das B, Salhan S,
Verma S, Mittal A. Cytokine expression
pattern in the genital tract of Chlamydia
trachomatis positive infertile women - im-
plication for T-cell responses. Clin Exp Im-
munol 2004;137(3):552-558. doi:10.1111/
j.1365-2249.2004.02564.x
80. Pal S, Hui W, Peterson EM, de la Maza LM.
Factors influencing the induction of infer-
tility in a mouse model of Chlamydia tra-
chomatis ascending genital tract infection.
J Med Microbiol 1998; 47:599–605.
81. Brunham RC. Human immunity to chla-
mydiae. In: Stephens RS, ed. Chlamydia:
270 Núñez Troconis
Investigación Clínica 62(3): 2021
Intracellular Biology, Pathogenesis and Im-
munity. Washington DC: ASM Press, 1999;
211–238.
82. Igietseme JU, Uriri IM, Kumar SN, Anaq-
naba GA, Ojior OO, Momodu IA, Caudal
DH, Black CM. Route of infection that in-
duces a high intensity of gamma interferon-
secreting T-cells in the genital tract produ-
ces optimal protection against Chlamydia
trachomatis infection in mice. Infect Im-
mun 1998; 66:4030–4035.
83. Kelly KA, Robinson E, Rank RG. Initial
route of antigen administration alters the
T-cell cytokine profile produced in response
to the mouse pneumonitis biovar of Chla-
mydia trachomatis following genital infec-
tion. Infect Immun 1996; 64:4976–4983.
84. Kelly KA 2, Walker JC, Jameel SH, Gray
HL, Rank RG. Differential regulation of
CD4 lymphocyte recruitment between the
upper and lower regions of the genital tract
during Chlamydia trachomatis infection.
Infect Immun 2000; 68:1519–1528.
85. Rank RG, Bowlin AK, Kelly KA. Charac-
terization of lymphocyte response in the
female genital tract during ascending chla-
mydial genital infection in the guinea pig
model. Infect Immun 2000; 68:5293–5298.
86. Openshaw P, Murphy EE, Hosken NA, Mai-
ne V, Davis K, Murphy K, O’Garra. Hetero-
geneity of intracellular cytokine synthesis
at the single cell level in polarized T helper-
1 and T helper-2 populations. J Exp Med
1995; 182:1357–1367.
87. Arno JN, Ricker VA, Batteiger BE. Interfe-
ron-g in endocervical secretions of women
infected with Chlamydia trachomatis. J In-
fect Dis 1990; 162:1385–1389.
88. Rank RG, Ramsey KH, Pack EA, Williams
DM. Effect of gamma interferon on resolu-
tion of murine chlamydial genital infection.
Infect Immun 1992; 60:4427–4429.
89. Zhong G, Peterson EM, Czarniecki CW,
Schreiber RD, de la Maza LM. Role of en-
dogenous gamma interferon in host defense
against Chlamydia trachomatis infections.
Infect Immun 1989; 57:152–157.
90. Perry LL, Feilzer K, Caldwell HD. Immuni-
ty to Chlamydia trachomatis is mediated by
T-helper 1 cells through IFN-gamma-depen-
dent and independent pathways. J Immunol
1997; 158:3344–3352.
91. Beatty WL, Byrne GI, Morrison RP. Mor-
phologic and antigenic characterization
of interferon-gamma mediated persistent
Chlamydia trachomatis infection in vitro.
Proc Natl Acad Sci USA 1993; 90:3998–
4002.
92. Rottenberg ME, Gigliotti-Rothfuchs A,
Wigzell H. The role of IFN- gamma in the
outcome of chlamydial infection. Curr Opin
Immunol 2002; 14:444–451.
93. Durum SK, Schimidt JA, Oppenheim JJ.
Interleukin-1: An immunological perspecti-
ve. Ann Rev Immunol 1985; 3:263–287.
94. MittalA,KapurS,GuptaS. Host Immuneres-
ponse in chlamydial cervicitis. Br J Biomed
Sci 1996; 53:941–947.
95. Kelly KA. Cellular immunity and Chla-
mydia genital infection: Induction, re-
cruitment and effector mechanisms.
Int Rev Immunol 2003; 22:3–41. doi:
10.1080/08830180305229.
96. Tseng CK, Rank RG. Role of NK cells in
early host immune response to chlamydial
genital infection. Infect Immun 1998;
66:5867–5875.
97. Stagg AJ, Elsley WAJ, Holland MJ. Den-
dritic cells (DC) in the initiation of immu-
ne responses to Chlamydia. In: Mardh PA,
La Placa M, Ward ME, eds. Proceedings Of
European Society for Chlamydia Research.
Town: Publisher, 1992: p.77–80.
98. Conti P, Kempuraj D, Kandere K, Di Gio-
acchino M, Barbacane RC, Castellani
ML, Felaco M, Bouchr W, Letourneau R,
Theoharides TC. IL-10, an inflammatory/
inhib- itory cytokine, but not always. Immu-
nol Lett 2003; 86:123–129. doi: 10.1016/
s0165-2478(03)00002-6.
99. Campbell K. Understanding how viruses
can cause malignant disease. Nurs Times
2006; 102:30-31.
100. Campbell K. The infectious causes of can-
cer. Nurs Times 2006;102:28-30.
101. Ledwaba T, Dlamini Z, Naicker S, Bhoo-
la K. Molecular genetics of human cervical
cancer: role of papillomavirus and the apop-
totic cascade. Biol Chem 2004;385:671-
682.
102. Nahar A, Azad AK. Sexually transmitted
diseases (STD) / reproductive tract infec-
tions (RTI) including acquired immunodefi-
ciency syndrome (AIDS) / human immuno-
Chlamydia and Cervical Cancer 271
Vol. 62(3): 247 - 275, 2021
deficiency virus (HIV) infections among the
women of reproductive age group: a review.
J Prev Soc Med 1999;18:84-88.
103. Thomas DJ. Sexually transmitted viral in-
fections: epidemiology and treatment. J
Obstet Gynecol Neonatal Nurs 2001;30:316-
323.
104. Kanodia S, Fahey LM, Kast WM. Mecha-
nisms used by human papillomaviruses to
escape the host immune response. Curr
Cancer Drug Targets 2007;7:79-89.
105. Klein E, Kis LL and Takahara M. Pathoge-
nesis of Epstein-Barr virus (EBV)-carrying
lymphomas. Acta Microbiol Immunol Hung
2006;53:441- 457.
106. Negro F. Mechanisms and significance of
liver steatosis in hepatitis C virus infection.
World J Gastroenterol 2006;12:6756-6765.
107. Moss SF, Blaser MJ. Mechanisms of disea-
se: Inflammation and the origins of cancer.
Nat Clin Pract Oncol 2005;2:90-97.
108. Rapp F. Current knowledge of mechanisms
of viral carcinogenesis. Crit Rev Toxicol
1984; 13:197-204.
109. Willecke K, Schafer R. Human oncogenes.
Hum Genet 1984;66:132-142.
110. Gilden RV, Rabin H. Mechanisms of viral
tumorigenesis. Adv Virus Res 1982;27:281-
334.
111. Cuschieri KS, Horne AW, Szarewski A, Cu-
bie HA. Public awareness of human papillo-
mavirus. J Med Screen 2006;13:201- 207.
112. Behtash N, Mehrdad N. Cervical cancer:
screening and prevention. Asian Pac J Can-
cer Prev 2006;7:683-686.
113. Dehn D, Torkko KC, Shroyer KR. Human
papillomavirus testing and molecular mar-
kers of cervical dysplasia and carcinoma.
Cancer 2007;111:1-14.
114. Weaver BA. Epidemiology and natural his-
tory of genital human papillomavirus infec-
tion. J Am Osteopath Assoc 2006;106:S2-8.
115. Zheng ZM, Baker CC. Papillomavirus ge-
nome structure, expression, and post-trans-
criptional regulation. Front Biosci 2006;
11:2286-2302.
116. Monsonego J. Cervical cancer prevention:
the impact of HPV vaccination. Gynecol
Obstet Fertil 2006;34:189-201.
117. Malik AI. The role of human papilloma vi-
rus (HPV) in the aetiology of cervical can-
cer. J Pak Med Assoc 2005;55:553-558.
118. Trottier H, Franco EL. The epidemiology
of genital human papillomavirus infection.
Vaccine 2006;24:S1-1S5.
119. González-Martinez G, Núñez-Troconis J.
Historia natural de la infección por el virus
del papiloma humano: una actualización.
Invest Clin 2014;55(1):82-91.
120. Lehtinen M, Ault KA, Lyytikainen E, Di-
llner J, Garland SM, Ferris DG, Koutsky
LA, Sings HL, Lu S, Haupt RM, Paavonen
J; FUTURE I and II Study Group. Chla-
mydia trachomatis infection and risk of cer-
vical intraepithelial neoplasia. Sex Transm
Infect 2011 Aug;87(5):372-6. doi: 10.1136/
sti.2010.044354. Epub 2011 Apr 6. PMID:
21471141; PMCID: PMC3252607.
121. Madeleine MM, Anttila T, Schwartz SM,
Saikku P, Leinonen M, Carter JJ, Wurs-
cher M, Johnson LG, Galloway DA, Da-
ling JR. Risk of cervical cancer associated
with Chlamydia trachomatis antibodies by
histology, HPV type and HPV cofactors.
Int J Cancer 2007;120(3):650-655. doi:
10.1002/ijc.22325. PMID: 17096345; PM-
CID: PMC4049152.
122. Muñoz N, Bosch FX, de Sanjosé S, Herrero R,
Castellsagué X, Shah K, Snijders PJF, Meijer
CJLM, International Agency for Research on
Cancer, Mukticenter Cervical Cancer Study
Group. Epidemiologic classification of human
papillomavirus types associated with cervical
cancer. N Engl J Med 2003;348(6):518-527.
doi:10.1056/NEJMoa021641.
123. Gravitt PE, Castle PE. Chlamydia tracho-
matis and cervical squamous cell carcino-
ma. JAMA 2001;285:1703-1704.
124. Al-Daraji WI, Smith JH. Infection and cer-
vical neoplasia: facts and fiction. Int J Clin
Exp Pathol 2009;2(1):48-64.
125. Fullgrabe J, Kavanagh E, Joseph B. Histo-
ne onco-modifications. Oncogene 2011;30:
3391–3403.
126. Clayton AL, Mahadevan LC. MAP kinase-
mediated phosphoacetylation of histone H3
and inducible gene regulation. FEBS Lett
2003;546, 51–58.
127. Chumduri C, Gurumurthy RK, Zadora
PK, Mi Y, Meyer TF. Chlamydia infection
promotes host DNA damage and prolifera-
tion but impairs the DNA damage respon-
se. Cell Host Microbe 2013;13(6):746-758.
doi:10.1016/j.chom.2013.05.010
272 Núñez Troconis
Investigación Clínica 62(3): 2021
128. Pennini ME, Perrinet S, Dautry-Varsat A,
Subtil A. Histone methylation by NUE, a
novel nuclear effector of the intracellular
pathogen Chlamydia trachomatis. PLoS
Pathog 2010;6, e1000995.
129. Miller KM, Jackson SP. Histone marks: re-
pairing DNA breaks within the context of
chromatin. Biochem. Soc. Trans 2012; 40:
370–376.
130. O’Driscoll M, Jeggo A. The role of double-
strand break repair- insights from human
genetics. Nat. Rev. Genet 2006;7: 45–54.
131. Lebreton A, Lakisic G, Job V, Fritsch
L, Tham TN, Camejo A, Matteı PJ, Reg-
nault B, Nahori MA, Cabanes D, Gau-
treau A, Ait-Si-Ali S, Dessen A, Cossart P,
Bierne H. A bacterial protein targets the
BAHD1 chromatin complex to stimulate
type III interferon response. Science 2001;
331:1319–1321.
132. Fehri LF, Rechner C, Janssen S, Mak TN,
Holland C, Bartfeld S, Bruggemann H,
Meyer FM. Helicobacter pylori induced mo-
dification of the histone H3 phosphoryla-
tion status in gastric epithelial cells reflects
its impact on cell cycle regulation. Epigene-
tics 2009; 4: 577–586.
133. Fernandez-Capetillo O, Allis CD, Nussen-
zweig A. Phosphorylation of histone H2B
at DNA double-strand breaks. J. Exp. Med
2004; 199:1671–1677.
134. Utley RT, Lacoste N, Jobin-Robitaille O, Al-
lard S, Coté J. Regulation of NuA4 histone
acetyltransferase activity in transcription and
DNA repair by phosphorylation of histone H4.
Mol. Cell. Biol 2005;25: 8179–8190.
135. Tjeertes JV, Miller KM, Jackson SP. Screen
for DNA-damage- responsive histone modifi-
cations identifies H3K9Ac and H3K56Ac in
human cells. EMBO J 2009;28;1878–1889.
136. Paavonen J, Purola E. Cytologic findings
in cervical chlamydial infection. Med Biol
1980;58(3):174-178.
137. Luostarinen T, Lehtinen M, Bjorge T,
Abeler V, Hakana M, Hellman’s G, Jellum
E, Koskela T, Lenner P, Lie AK, Paavonen
J, Pukkala E, Saikku P, Sigstad E, Thore-
sen S, Thoresen S, Yuongman LD, Diller
J, Hakulinen T. Joint effects of different
human papillomaviruses and Chlamydia
trachomatis infections on risk of squamous
cell carcinoma of the cervix uteri. Eur J
Cancer 2004;40:1058e65.
138. Arnheim L. Immunological Responses in
Genital HPV Infections and Etiology of Cer-
vical Cancer. Stockholm, Sweden: Karolin-
ska Institute, 2005:1e75. Available http://
diss. kib.ki.se/2005/91-7140-266-7/thesis.
pdf. Accesed on: June 10, 2020.
139. Quint KD, de Koning MN, Geraets DT,
Quint WG, Pirog EC. Comprehensive anal-
ysis of Human Papillomavirus and Chla-
mydia trachomatis in in-situ and invasive
cervical adenocarcinoma. Gynecol Oncol
2009;114(3):390-394. doi:10.1016/j.ygy-
no.2009.05.013
140. Agrawal T, Vats V, Wallace L. Role of cer-
vical dendritic cell subsets, co-stimulatory
molecules, cytokine secretion profile and
beta-estradiol in development of sequalae
to Chlamydia trachomatis infection. BMC
Reproduct Biol Endocrinol 2008;6:46.
141. Samoff E, Koumans EH, Markowitz LE,
Sternberg M, Sawyer MK, Swan D, Papp
JR, Black CM, Unger ER. Association of
Chlamydia trachomatis with persistence of
high-risk types of human papillomavirus in
a cohort of female adolescents. Am J Epide-
miol 2005;162(7):668-675. doi: 10.1093/
aje/kwi262. Epub 2005 Aug 24.
142. Stamm WE. Chlamydia trachomatis infec-
tions of the adult. In: Holmes KK, Mardh
PA, Sparlin PF, Lemon SM, Stamm WE, Piot
P, Wasserheit JN, editors. Sexually trans-
mitted diseases, 3rd edition. New York: Mc-
Graw-Hill, 1999. p:407–422.
143. Nobbenhuis MA, Walboomers JM, Helmer-
horst TJ, Rozendaal L, Remmink AJ,
Risse EK, van der Linden HC, Voorhorst
FJ, Kenemans P, Meijer CJ. Relation of
human papillomavirus status to cervical
lesions and consequences for cervical-can-
cer screening: a prospective study. Lancet
1999; 354:20–25.
144. Kjaer SK, van den Brule AJ, Paull G, Svare
EI, Sherman ME, Thomsen BL, Suntum
M, Bock JE, Poll PA, Meijer CJ. Type spe-
cific persistence of high risk human papil-
lomavirus (HPV) as indicator of high grade
cervical squamous intraepithelial lesions in
young women: population based prospec-
tive follow up study. BMJ 2002; 325:572.
Chlamydia and Cervical Cancer 273
Vol. 62(3): 247 - 275, 2021
145. Schiffman M, Solomon D. Cervical-can-
cer screening with human papillomavirus
and cytologic co-testing. New Engl J Med
2013;369: 2324-2331.
146. Martins MCL, Bôer CG, Svidzinski TIE,
Estivalet TI, Donida LG, Martins PFA,
Boscoli FNS, Consolaro MEL. Avaliação
do método de Papanicolaou para triagem
de algumas infecções cérvico-vaginais. Rev
Bras Anal Clin 2007;39: 217-221.
147. Campos ACC, Freitas-Júnior R, Ribeiro
LFJ, Paulinelli RR, Reis C. Prevalence of
vulvovaginitis and bacterial vaginosis in pa-
tients with koilocytosis. Sao Paulo Med J
2008;126: 333-336.
148. Muvunyi CM, Dhont N, Verhelst R, Cru-
citti T, Reijans M, Mulders B, Simons G,
Temmerman M, Claeys G, Padalko E. Eval-
uation of a new multiplex polymerase chain
reaction assay STDFinder for the simulta-
neous detection of 7 sexually transmitted
disease pathogens. Diagn Microbiol Infect
Dis 2011;71: 29-37.
149. Schachter J, Hill EC, King EB, Coleman
VR, Jones P, Meyer KF. Chlamydial infec-
tion in women with cervical dysplasia. Am J
Obstet Gynecol 1975;123:753–757.
150. Smith JS, Bosetti C, Munoz N, Herrero
R, Bosch FX, Eluf-Neto J, Meijer CJ, Van
Den Brule AJ, Franceschi S, Peeling RW.
Chlamydia trachomatis and invasive cervi-
cal cancer: A pooled analysis of the IARC
multicentric case-control study. Int J Can-
cer 2004;111: 431–439.
151. Hakama M, Lehtinen M, Knekt P, Aromaa
A, Leinikki P, Miettinen A, Paavonen J,
Peto R, Teppo L. Serum antibodies and
subsequent cervical neoplasms: a prospec-
tive study with 12 years of follow-up. Am J
Epidemiol 1993;137:166–170.
152. Anttila T, Saikku P, Koskela P, Bloigu A,
Dillner J, Ikaheimo I, Jellum E, Lehtinen
M, Lenner P, Hakulinen T, Narvanen A, Puk-
kala E, Thoresen S, Youngman L, Paavonen
J. Serotypes of Chlamydia trachomatis and
risk for development of cervical squamous
cell carcinoma. JAMA 2001;285:47–51. doi:
10.1001/jama.285.1.47.
153. Wallin KL, Wiklund F, Luostarinen T,
Angstrom T, Anttila T, Bergman F, Hall-
mans G, Ikaheimo I, Koskela P, Lehtinen
M, Stendahl U, Paavonen J, Diener J. A
population-based prospective study of Chla-
mydia trachomatis infection and cervical
carcinoma. Int J Cancer 2002; 101:371–
374. doi: 10.1002/ijc.10639.
154. Koskela P, Anttila T, Bjørge T, Brunsvig
A, Dillner J, Hakama M, Hakulinen T,
Jellum M, Lethinen M, Lenner P, Luos-
tarinen T, Pukkala, Saikku E, Thore-
sen S, Youngman J, Paavonen J. Chla-
mydia trachomatis infection as a risk
factor for invasive cervical cancer. Int J
Cancer. 2000; 85:35–39. doi: 10.1002/
(sici)1097-0215(20000101)85:1<35::aid-
ijc6>3.0.co;2-a.
155. Quint KD, de Koning MN, Geraets DT,
Quint WG, Pirog EC. Comprehensive
analysis of Human Papillomavirus and
Chlamydia trachomatis in in-situ and inva-
sive cervical adenocarcinoma. Gynecol On-
col 2009;114(3):390-394. doi:10.1016/j.
ygyno.2009.05.013.
156. Ghosh I, Mandal R, Kundu P, Biswas J.
Association of genital infections other
than human papillomavirus with pre-Inva-
sive and invasive cervical neoplasia. J Clin
Diagn Res. 2016 Feb;10(2):XE01-XE06.
doi: 10.7860/JCDR/2016/15305.7173.
Epub 2016 Feb1.
157. Johnston VJ, Mabey DC. Global epidemio-
logy and control of Trichomonas vaginalis.
Curr Opin Infect Dis 2008;21(1):56-64.
doi:10.1097/QCO.0b013e3282f3d999
158. Paavonen J. Chlamydia trachomatis and can-
cer. Sex Transm Infect 2001; 77: 154-156.
159. Deluca GD, Basiletti J, Schelover E, Díaz-
Vásquez N, Alonso JM, Marin HM, Lucero
RH, Picconi MA. Chlamydia trachomatis
as a probable cofactor in human papilloma-
virus infection in aboriginal women from
northeastern Argentina. Braz J Infect Dis
2011;15(6):567-572. doi:10.1016/s1413-
8670(11)70252-5.
160. Gopalkrishna V, Aggarwal N, Malhotra
VL, Koranne RV, Mohan VP, Mittal A, Das
BC. Chlamydia trachomatis and human
papillomavirus infection in Indian women
with sexually transmitted diseases and cer-
vical precancerous and cancerous lesions.
Clin Microbiol Infect 2000;6(2):88-93.
doi:10.1046/j.1469-0691.2000.00024.x
161. Verhoeven V, Baay M, Weyler J, Avonts D,
Lardon F, Van Royen P, Vermorken JB.
274 Núñez Troconis
Investigación Clínica 62(3): 2021
Concomitant Chlamydia trachomatis and
human papilloma virus infection cannot be
attributed solely to sexual behavior. Eur J
Clin Microbiol Infect Dis 2004;23(9):735-
737.
162. Smith JS, Muñoz N, Herrero R, Eluf-Neto
J, Ngelangle C, Francheschi S, Bosch FX,
Walboomers JMM, Peeling RW. Evidence
for Chlamydia trachomatis as a human pa-
pillomavirus cofactor in the etiology of in-
vasive cervical cancer in Brazil and the Phi-
lippines. J Infect Dis 2002;185(3):324-331.
doi:10.1086/338569.
163. Fan T, Lu H, Hu H,Shi L, McCarty GA,
Nance DM, Greenberg AH, Zhong G. In-
hibition of apoptosis in chlamydia-infected
cells: blockade of mitochondrial cytochro-
me c release and caspase activation. J Exp
Med 1998;187:487–496.
164. Jaiswal M, LaRusso NF, Burgart LJ, Gores
GJ. Inflammatory cytokines induce DNA
damage and inhibit DNA repair in cholan-
giocarcinoma cells by a nitric oxide–de-
pendent mechanism. Cancer Res 2000;60:
184–190.
165. Zhu H, Shen Z, Luo H, Zhang W, Zhu X.
Chlamydia Trachomatis Infection-Associa-
ted Risk of Cervical Cancer: A Meta-Analysis.
Medicine (Baltimore). 2016;95(13):e3077.
doi:10.1097/MD.0000000000003077.
166. Velicer C, Zhu X, Vuocolo S, Liaw KL,
Saah A. Prevalence and incidence of HPV
genital infection in women. Sex Transm
Dis. 2009;36(11):696-703. doi:10.1097/
OLQ.0b013e3181ad25ff
167. Shew ML, Fortenberry JD, Tu W, Juliar BE,
Batteiger BE, Qadadri B, Brown DR. As-
sociation of condom use, sexual behaviors,
and sexually transmitted infections with the
duration of genital human papillomavirus
infection among adolescent women. Arch
Pediatr Adolesc Med 2006;160(2):151-156.
doi: 10.1001/archpedi.160.2.151.
168. Koutsky LA, Holmes KK, Critchlow CW,
Stevens CE, Paavonen J, Beckmann AM,
DeRouen TA, Galloway DA, Veton D, Ki-
vat NB. A cohort study of the risk of cer-
vical intraepithelial neoplasia grade 2 or
3 in relation to papillomavirus infection.
N Engl J Med. 1992;327(18):1272-1278.
doi:10.1056/NEJM199210293271804.
169. Lehtinen M, Dillner J, Knekt P, Luostari-
nen T, Aromaa A, Kirnbauer R, Koskela P,
Paavonen J, Peto R, Schiller JT, Hakama
M. Serologically diagnosed infection with
human papillomavirus type 16 and risk for
subsequent development of cervical car-
cinoma: nested case-control study. BMJ
1996;312(7030):537-539. doi: 10.1136/
bmj.312.7030.537.
170. Naucler P, Chen HC, Persson K, You SL,
Hsieh CY, Sun CA, Dillner J, Chen CJ.
Seroprevalence of human papillomaviru-
ses and Chlamydia trachomatis and cer-
vical cancer risk: nested case-control stu-
dy. J Gen Virol 2007;88(Pt 3):814-822.
doi:10.1099/vir.0.82503-0.
171. Anttila T, Saikku P, Koskela P, Bloigu A, Di-
llner J, Ikaheimo I, Jellum E, Mehtinen M,
Lenner P, Hakulinen T, Narvanen A, Pukka-
la E, Thoresen S, Youngman L, Paavonwn
J.. Serotypes of Chlamydia trachomatis and
risk for development of cervical squamous
cell carcinoma. JAMA 2001;285(1):47-51.
doi:10.1001/jama.285.1.47.
172. Workowski KA, Suchland RJ, Pettinger
MB, Stamm WE. Association of genital
infection with specific Chlamydia tracho-
matis serovars and race. J Infect Dis 1992;
166:1445-1449.
173. Kuo CC, Wang SP, Holmes KK, Grayston
T. Immunotypes of Chlamydia trachomatis
isolates in Seattle, Washington. Infect Im-
mun 1983;41:865-868.
174. Saikku P, Wang SP. Chlamydia tracho-
matis immunotypes in Finland. APMIS
1987;95:131-134.
175. Moncan T, Eb F, Orfila J. Monoclonal anti-
bodies in serovar determination of 53 Chla-
mydia trachomatis isolates from Amiens,
France. Res Microbiol 1990; 141:695-701.
176. Barnes RC, Rompalo AM, Stamm WE.
Comparison of Chlamydia trachomatis
serovars causing rectal and cervical infec-
tions. J Infect Dis 1987;156:953-958.
177. Poole E, Lamont I. Chlamydia trachoma-
tis serovar differentiation by direct sequen-
ce of the variable segment 4 region of the
major outer membrane protein gene. Infect
Immun 1992; 60:1089-1094.
Chlamydia and Cervical Cancer 275
Vol. 62(3): 247 - 275, 2021
178. Barnes RC, Suchland RJ, Wang S-P, Kuo
CC, Stamm WE. Detection of multiple se-
rovars of Chlamydia trachomatis in geni-
tal infections. J Infect Dis 1985;152:985-
989.
179. Fan T, Lu H, Hu H, McCarty GA, Nance DM,
Greenberg AH, Zhong G. Inhibition of apop-
tosis in chlamydia-infected cells: blockade of
mitochondrial cytochrome c release and cas-
pase activation. J Exp Med 1998;187(4):487-
496. doi:10.1084/jem.187.4.487.