ArticlePDF AvailableLiterature Review

HPV co‐infections with other pathogens in cancer development: A comprehensive review

Authors:

Abstract

High‐risk human papillomaviruses (HPVs) cause various malignancies in the anogenital and oropharyngeal regions. About 70% of cervical and oropharyngeal cancers are caused by HPV types 16 and 18. Notably, some viruses including herpes simplex virus (HSV), Epstein‐Barr virus (EBV) and human immunodeficiency virus (HIV) along with various bacteria often interact with HPV, potentially impacting its replication, persistence, and cancer progression. Thus, HPV infection can be significantly influenced by co‐infecting agents that influence infection dynamics and disease progression. Bacterial co‐infections ( e.g., Chlamydia trachomatis ) along with bacterial vaginosis‐related species also interact with HPV in genital tract leading to viral persistence and disease outcomes. Co‐infections involving HPV and diverse infectious agents have significant implications for disease transmission and clinical progression. This review explores multiple facets of HPV infection encompassing the co‐infection dynamics with other pathogens, interaction with the human microbiome, and its role in disease development. This article is protected by copyright. All rights reserved.
Received: 24 August 2023
|
Accepted: 2 November 2023
DOI: 10.1002/jmv.29236
REVIEW
HPV coinfections with other pathogens in cancer
development: A comprehensive review
Elahe Akbari |Alireza Milani |Masoud Seyedinkhorasani |Azam Bolhassani
Department of Hepatitis and AIDS, Pasteur
Institute of Iran, Tehran, Iran
Correspondence
Azam Bolhassani
Email: azam.bolhassani@yahoo.com and
A_bolhasani@pasteur.ac.ir
Abstract
Highrisk human papillomaviruses (HRHPVs) cause various malignancies in the
anogenital and oropharyngeal regions. About 70% of cervical and oropharyngeal
cancers are caused by HPV types 16 and 18. Notably, some viruses including herpes
simplex virus, EpsteinBarr virus, and human immunodeficiency virus along with
various bacteria often interact with HPV, potentially impacting its replication,
persistence, and cancer progression. Thus, HPV infection can be significantly
influenced by coinfecting agents that influence infection dynamics and disease
progression. Bacterial coinfections (e.g., Chlamydia trachomatis) along with bacterial
vaginosisrelated species also interact with HPV in genital tract leading to viral
persistence and disease outcomes. Coinfections involving HPV and diverse
infectious agents have significant implications for disease transmission and clinical
progression. This review explores multiple facets of HPV infection encompassing the
coinfection dynamics with other pathogens, interaction with the human micro-
biome, and its role in disease development.
KEYWORDS
cervical cancer, coinfection, HPV, HPVrelated cancers, microbiome, viral pathogens
1|INTRODUCTION
Human papillomaviruses (HPVs) are the most common sexually
transmitted viruses which cause different disorders in women and
men such as precancerous lesions and different cancers.
1
In 1933,
HPVs were described as a large family of DNA viruses.
2,3
Viral
genome encodes core and accessory proteins. Core proteins have a
major role in viral genome replication (E1 and E2: early proteins) and
also virus assembly (L1 and L2: late proteins). These proteins are
highly conserved among all types of papillomaviruses. In contrast,
the accessory proteins (E4, E5, E6, and E7: early proteins) possess
more variability in their expression time and functional properties.
These proteins modify the infected cells to facilitate viral replication
in different diseasespapillomavirus type relationships.
4
To date,
more than 200 HPV types have been identified. They were divided
into Alpha, Beta, Gamma, Mu, and Nu genera.
5
Notably, HPVs are
categorized as either 15 HRHPV/oncogenic types which can be
potentially carcinogenic, or 12 lowrisk (LR) HPV/nononcogenic
types which are often found in warts.
6
Although most HPV infections
are benign, persistent infection with one of the carcinogenic HR
HPV types is the main cause of cervical cancer. HPV types 16 and 18
are the most carcinogenic HPVs responsible for ~70% of cervical
cancer cases.
7
Moreover, several biological and environmental co
factors including tobacco usage, parity, hormonal changes, dietary
habits, immune level, and coinfection with other pathogens were
involved in the progression of HPVassociated cancers.
8
In 2020,
around 604 127 individuals received a new diagnosis of cervical
cancer, and 34 1831 lost their lives to this ailment on a global scale.
Regrettably, a significant majority of both new cases and fatalities,
ranging from 85% to 90%, have taken place in countries categorized
J Med Virol. 2023;95:e29236. wileyonlinelibrary.com/journal/jmv © 2023 Wiley Periodicals LLC.
|
1of21
https://doi.org/10.1002/jmv.29236
Elahe Akbari and Alireza Milani are the first authors.
as less developed.
9
In coinfection, the existence of one infectious
agent modifies the natural history of another one. Interactions of
HPVs with viruses or bacteria that share a similar epithelial niche
or transmission routes could increase HPV replication and
persistence, and accelerate cancer progression. Indeed, a history
of prior sexually or orally transmitted infections including human
immunodeficiency virus (HIV), herpes simplex virus (HSV),
EpsteinBarr virus (EBV), and different oral and cervicovaginal
bacteria led to a decreased ability for HPV clearance or an
increased risk of HPV infection.
8
Coinfection with multiple
genotypes was usually detected in HPV HPVpositive subjects,
as well.
10
In this review, the transmitted infectious agents as
potential risk factors in HPVrelated cancers are discussed.
2|EPIDEMIOLOGY
HPV infection is currently a global public health priority especially
among women.
11
The global HPV prevalence was estimated about
11.7%. The highest HPV prevalence was detected in Caribbean,
Eastern Africa, Eastern Europe, South Africa, and Western Europe.
Female sex workers are among the most susceptible groups to
HPV infections mainly HPV types 16, 52, and 53.
12
Table 1
represents the prevalence of HPV types 16/18 in women with
healthy cervical cell samples and precancerous cervical abnormali-
ties in some continents and subregions. The most prevalent HR
HPV types in the world include the HPV genotypes 16, 18, 59, 45,
31, 33, 52, 58, 35, 39, 51, 56, and 53, respectively. Also, the most
common LRHPV types are the HPV genotypes 6 and 11 causing
genital warts.
18
Table 2indicates the prevalence of typespecific
HPVs in women worldwide.
3|HPV LIFE CYCLE
ThelifecycleofHPVcontainsestablishment, maintenance, and
vegetative/productive amplification phases, respectively. The establish-
ment phase includes viral transcription and genome amplification in the
basal layer. After the entry into the cells, the virus requires the expression
of E1 and E2 genes to maintain a low number of copies of the genome.
24
After the initial establishment phase, the viral genome maintains a
constant copy number. Indeed, it is replicated approximately once during
the DNA synthesis phase (S phase) of infected cells and distributed to
daughter cells during cell division.
25
In this phase, the E6 and E7 proteins
are expressed in the suprabasal layer. The E7 protein degrades
retinoblastoma (Rb) family members (i.e., p105, p107, and p130) leading
to the release of the E2F transcription factor which promotes gene
expression in the S phase, and elicits hyperproliferation.
26
Viral assembly
occurs in the maturing squamous epithelium leading to the release of
amplified viruses from the terminally differentiated squamous cells. This
3week process refers to the maturation of a basal cell to the superficial
cells.
27
In the granular layer, the L1 and L2 proteins known as the major
and minor capsid proteins respectively, assemble to form new virions.
These new virions are released from the epithelial cornified layer.
28
4|HPV AND IMMUNE EVASION
HPVs possess several mechanisms to escape from host immunologic
responses and establish the HPVrelated lesions
29
including (a)
Coordination of viral replication to cellular differentiation: HPV
regulates its own replication with differentiation of the keratinocytes.
Moreover, virions are released through the mechanical breakage of
surface epithelium minimizing inflammatory responses
30
;(b)
TABLE 1 Incidence of HPV 16/18 in females with healthy cervical cell samples and precancerous cervical abnormalities in some continents
and subregions.
Continent/Subregion
Normal cytology Lowgrade lesions Highgrade lesions
Number
of tested
95% confidence
interval
Number
of tested
95% confidence
interval
Number
of tested
95% confidence
interval
African 19 726 3.8 465 24.9 399 38.6
Eastern Africa 4115 4.7 150 30.0 138 45.7
Americas 105 042 4.5 9893 26.7 13 590 56.9
South America 10 180 5.8 2191 35.6 2516 56.3
Asia 142 676 3.4 7959 21.2 13 444 42.1
Southern Asia 14 520 4.4 225 30.2 287 63.4
Europe 180 090 3.8 19 401 27.1 21 140 54.5
Eastern Europe 86 821 4.2 4,949 30.6 8448 54.9
Oceania 2997 8.3 473 27.1 1629 597.1
Australia & New Zealand 2271 8.5 473 27.1 1517 58.4
Abbreviation: HPV, human papillomavirus.
Source: Guan and colleagues.
1317
2of21
|
AKBARI ET AL.
Maintenance of viral antigens (i.e., early viral gene products) in low
levels controlled by E2 protein, and the lack of HPV proteins' secre-
tion: During disease progression, the HPV genome is integrated into
the host genome leading to the disruption of the E2 locus and thus
high expression of E6 and E7 proteins in highgrade lesions and
cancer.
31,32
The low levels of early viral proteins can hamper the
detection of HPVinfected cells by local antigenpresenting cells
named as Langerhans cells (LCs), and thus suppress the stimulation of
an effective adaptive immune response against these infected cells
33
;
(c) Direct inhibition of both innate and adaptive immune cell function:
Most cells of the innate immune system express pathogen
recognition receptors such as tolllike receptors (TLRs), NODlike
receptors (NLRs), and RIGIlike receptors.
3437
Furthermore, cyclic
GMPAMP synthase, a major cytosolic DNA sensor,
38
activates
STING (stimulator of interferon [IFN] genes) leading to transcription
of IFNs, chemokines, and cytokines, and thus induction of an antiviral
response.
39
For example, Hasan et al. showed that HPV16 E7
downregulates TLR9 in human epithelial cells by activating histone
demethylase JARID1B and histone deacetylase 1 and thus suppress-
ing IFN responses.
40
It was also reported that HPV18 E7 binds to
STING and inhibits upregulation of IFNs in the presence of cytosolic
DNA. In contrast, HPV16 E7 escapes from STINGinduced IFN
activation through the NLRX1 (NLR family member X1) protein.
24
5|CANCERRELATED HPVS
HPVs cause both premalignant and malignant lesions in different
tissues (e.g., cervical, anogenital, oropharyngeal cancers (OPC);
esophageal carcinoma)
3
(Table 3). Cervical cancer cases are often
related to HPV infections. For instance, the highest risk of cervical
intraepithelial neoplasia (CIN) was associated with HPV types 16 and
33 followed by HPV types 18, 31, and 45.
46
Head and neck, vulvar,
and esophageal squamous cell carcinoma (SCC) (i.e., head and neck
squamous cell carcinomas [HNSCC], vulvar squamous cell carcinoma,
and esophageal squamous cell carcinoma) are often related to HPV
type 16 followed by HPV type 18 and other strains (e.g., HPV types
31, 33, and 45).
4750
6|DIAGNOSIS AND TREATMENT
The diagnosis of HPV infections is the most important step for
control of HPVrelated diseases.
51,52
The effective techniques for
HPV detection and regular screening include Pap smear, biopsy,
acetic acid test and colposcopy, nucleic acid detection using
polymerase chain reaction, southern blot hybridization, and in situ
hybridization
53
as shown in Table 4. Serological tests are not useful
for HPV infection due to poor serological response of host.
54
The
HPV nucleic acid test is one of the accurate tests for HPV diagnosis in
women. On the other hand, it is worth noting that there is currently
no approved test specifically designed for HPV diagnosis in males.
Nevertheless, clinical diagnosis can utilize HPV mRNA or DNA in situ
for this purpose. Routine screening for HPVrelated diseases in men
is not presently recommended by the Centers for Disease Control
and PreventionUSA.
55,56
Sometimes, an anal Pap test may be
performed for men with a high risk (HR) of developing anal cancer.
1
On the other hand, the management and treatment of HPV
related diseases are largely influenced by various factors including
the specific types of HPVs, the availability of treatments, and the
TABLE 2 Typespecific HPV prevalence in women with normal cervical cytology, and precancerous cervical lesions in the World.
HPV type
Normal cytology Lowgrade lesions Highgrade lesions
Number of
tested
95% confidence
interval
Number of
tested
95% confidence
interval
Number of
tested
95% confidence
interval
Highrisk HPV type
16 453 184 2.8 38 177 19.3 50 202 45.1
18 440 810 1.1 37 748 6.5 49 743 6.8
31 415 367 1.2 36 170 7.7 48 538 10.4
33 413 075 0.7 35 733 4.7 48 592 7.3
35 396,307 2.8 31 095 3.0 44 703 3.3
Lowrisk HPV Type
6 418 946 0.9 26 981 6.2 34 563 2.3
11 406 162 0.5 26 179 2.9 33 547 1.3
40 186 634 0.3 4379 1.5 11 872 0.4
42 326 078 0.6 4932 7.1 9543 1.3
43 259 930 0.2 3258 1.7 5549 0.4
Abbreviation: HPV, human papillomavirus.
Source: Maranga and colleagues.
1923
AKBARI ET AL.
|
3of21
progression of the disease.
57
There are currently three HPV
prophylactic vaccines (e.g., Cervarix, Gardasil, and Gardasil9) that
are safe and effective in preventing cancerrelated HPV infections
58
;
but unfortunately, there is no licensed therapeutic vaccine. Thus, the
prophylactic HPV vaccination is considered as an additional
treatment in patients with recurrent HPVrelated diseases.
59
It is
hypothesized that such vaccination may stimulate cellmediated
immunity, which can play a role in preventing recurrent HPV
infections.
60
Several reports showed that adjuvant HPV vaccination
is related to a decreased risk of active HPVrelated diseases,
especially CIN recurrence.
59,61,62
The primary objective of treatment
is to alleviate symptoms, remove the transformation zone of warts or
lesions, and minimize the risk of future invasive cancers.
63
Unfortunately, there is currently no certain evidence regarding the
complete treatment of HPVrelated diseases.
57
In the cases of
infection with nononcogenic HPVs, recommended treatments for
external genital warts are topical medicine (e.g., Podophyllotoxin,
Imiquimod, Sinecatechins, and Trichloroacetic acid).
6366
Some
limited therapies such as 5fluorouracil, intralesional/topical IFN,
and photodynamic therapy are also recommended.
6769
Moreover,
surgery, chemotherapy, radiotherapy, targeted therapy, or their
combination are available therapies for treatment of HPVrelated
cancers.
62
In the recent years, several studies have suggested that
complementary treatments such as probiotics are useful in clearance
of HPV infections. Indeed, probiotics can stimulate the production of
antimicrobial peptides and antiinflammatory cytokines, prevent
bacterial adhesion and acidification, and subsequently reduce
bacterial vaginosis (BV) and sexually transmitted diseases.
7072
7|HPV COINFECTIONS
Different groups of pathogens (e.g., bacteria, viruses, protozoa, and
fungal parasites) infect human, and they often cooccur within
individuals.
73
Coinfection involves globally important infectious
agents such as HPV, HIV, tuberculosis, hepatitis, leishmaniasis, and
dengue fever.
74
The true prevalence of coinfection in infectious
diseases exceeds onesixth of the global population.
75
In coinfection,
pathogens can interact either directly with one another or indirectly
through the host's resources or immune system. These interactions
within coinfected hosts change the transmission, clinical progression
and control of multiple infectious diseases compared to single
pathogen infections.
76,77
HPV infection (a main risk factor for human
malignancies) often increases the risk of coinfection with other
infectious agents such as viruses and bacteria.
74
A brief discussion of
these coinfections is described in the next sections.
8|COINFECTION OF HPVS WITH
OTHER VIRUSES
As mentioned earlier, development of HPVassociated dysplasia is
strongly associated with chronic or persistent HRHPVs infections.
8
However, it is worth noting that such infections typically resolve
spontaneously by the immune system. Moreover, the risk of
developing cancer in HRHPVs infection is low.
78,79
For example, in
the case of cervical cancer, only <1% of HPV
+
women will develop
neoplasia.
8
Thus, additional biological and environmental risk factors
like coinfection with other infectious agents may reduce the host
ability to clear HPV or increase the risk of HPV infectionrelated
malignancies.
80
Several studies reported that different viruses with
the same epithelial niche may interact with HPV leading to an
increased HPV replication and persistence, and thus accelerating
cancer progression.
8
We will review several viruses as potential risk
factors in HPVrelated neoplasia in the next sections. Among them,
HIV was known as the most important virus associated with HPV
infections.
8.1 |HPV and herpessimplex virus coinfection
The herpesviridae family includes the enveloped doublestranded
DNA viruses (i.e., HSV, EBV, Cytomegalovirus [CMV] and human
herpesvirus 6 and 8 [HHV6 & 8]).
81
They can establish lifelong latent
infections in the host.
82
The latent herpesviruses can be reactivated
in response to stress, and cause secondary infection in epithelium for
productive viral replication and shedding.
8
These viruses replicate
generally in the epithelial cells of the oral cavity and genital tract.
83
Different reports showed the implication of herpesviruses in
increasing the risks of cervical dysplasia.
82,83
Coinfection of HPV
with members of herpesviridae especially HSV and EBV was also
reported in different studies.
8385
Regarding HSV, there are two viral
TABLE 3 Some characteristics of HPVrelated cancers in the world.
Cancer Localization Sex Age (year) New cases of HPVsrelated cancers in 2020 References
Cervical cancer Cervix Women 1544 604 127 [41]
Oropharyngeal cancer Head and neck Men and women Mean age: 68 98 412 [42]
Vulvar squamous cell carcinoma Skin Men and women Mean age: 70 45 240 [43]
Penile cancer Penis Men 5070 36 068 [44]
Esophageal cancer Esophagus Men and women Mean age: 65 604 100 [45]
Abbreviation: HPV, human papillomavirus.
4of21
|
AKBARI ET AL.
genotypes of HSV1 and HSV2 that are distinguished by envelope
antigenic differences, and cause common oral and genital infections
worldwide.
80
Although both viruses are a significant source of
diseases all over the world, HSV1 is more seroprevalent.
86
HSV1
and HSV2 can be transmitted by close personal contact with an
individual shedding and attacking oral and genital mucosa, respec-
tively.
80
Thus, HSV2 is more frequently linked to recurrent genital
herpes.
81
Different epidemiological investigations have indicated the
correlation between HSV and HPV infections.
80,85
High incidence
rates for such coinfections were observed because of direct contact
with lesions or with HSVinfected oral or genital secretions during
asymptomatic shedding that may enable HPV to transmit and access
the basal cell layer more profoundly.
87
It was proposed that the
replication of HSV in tissues where HPV also replicates may have a
direct or indirect impact on the persistence, clearance, and/or
oncogenic potential of HPV.
8
According to different reports, HSV2
positive cases have a 2to 9fold higher chance of experiencing
cervical SCC or adenocarcinoma than HSV2 negative ones.
85
Furthermore, HPV/HSV2coinfection in cervical intraepithelial
neoplasia and SCC was strongly higher than in healthy women.
88
Comparatively to 0%4% of healthy cervical tissues, HSV2
coinfection with HPV types 16 and 18 was observed in 25%30%
of CIN and 13%25% of invasive cervical SCC and adenocarcino-
mas.
88
A crosssectional study in 2020 reported a significant
difference in HSV2 seroprevalence and HSV2 active infection rates
between negative and positive HRHPVs cases.
89
Additionally, HSV
2 can boost transmission of HIV1, EBV, or other sexually transmitted
pathogens, thus helping the HPV infection and persistence in this
way.
90
HSV2 and HIVinfected women were reported to have
cervicovaginal inflammation, and harbor a high diversity of microbes
leading to more susceptibility to HPV infection.
91
HPV/HSV2co
infection interferes with local immune responses, which increases the
likelihood of HPVrelated lesions progression.
92
A few studies
reported coinfection of HPV, EBV, and/or HSV in different
anatomical sites like anorectum, oral cavity, oropharynx, and
urethra.
90
A similar correlation between HSV1 and HPV was
observed in HNSCC. For example, HPV16/HSV1coinfection in
patients with HNSCC showed the worst disease outcome.
90
In
addition, HSV1 infection may increase the radiation resistance of
HPV16positive cancer cells by improving cell survival and preventing
apoptosis.
93
Previous studies demonstrated that HSV1 interferes
with DNA repair mechanism in the cells leading to some genetic
changes during the process of acute lymphoblastic leukemia.
88
Additionally, HSV infections induce permanent genetic alterations
through unexpected cellular DNA synthesis and chromosomal
amplifications that interfere with the differentiation of the cervical
epithelium, and subsequently induce abnormal proliferation as an
HPV cofactor.
83
In vitro studies suggested that HSV may contribute
to the process of HPV carcinogenesis without the need for continued
presence (hitandrun theory). This scenario explains that HSV
replication makes significant cytopathic effects in HPVinfected cells,
and also can reduce HPV E2, E6, and E7 expression and DNA
replication.
8
Transient infection by HSV leaves lasting molecular
TABLE 4 Screening and diagnosis of HPVrelated pathogenesis.
Type Characteristic Method Clinical sensitivity Principle
Nonmolecular techniques Cell morphology Visual inspection Low Visual inspection of the cervix
Colposcopy Moderate Stereoscopic and magnified viewing of the cervix
Cytology and histology High Examining of individual cells or an entire section of tissue
Molecular techniques HPV nucleic acids PCR High Amplification of viral sequences present in the biological specimen
Hybridization High Formation of specific HPV DNARNA hybrids, which are then captured by antibodies
Southern and Northern blot Moderate Hybridization (southern blot for DNA and northern blot for RNA molecules) with specific
HPV probes
In situ hybridization Moderate Identifying specific nucleotide sequences in cells or tissue sections with conserved
morphology
Serological assays AntiHPV antibodies Detection of capsid antibody Low Identifying HPV by VLPbased ELIZA
Neutralization assays Low Neutralizing epitopes by monoclonal antibodies
Detection of antibodies to HPV proteins Low Using HPV E6, E7 proteins as antigens in either ELIZA or western blot analysis
Abbreviation: HPV, human papillomavirus; VLP, virallike particles.
AKBARI ET AL.
|
5of21
changes that enhance oncogenic potential of HPV. For example, HSV
can activate AP1 (activator protein1) pathway by inducing the
expression of viral immediate early genes such as infected cell protein
0 which can directly interact with AP1 transcription factors. This
activation of AP1 can lead to the upregulation of genes involved in
cell cycle progression, antiapoptotic signaling, and angiogenesis, all of
which contribute to tumorigenesis.
94
Under this scenario, detailed
research is needed to better understand the biological mechanisms
underlying the HPV/HSV coinfection.
8.2 |HPV and EBV coinfection
EBV is a herpesvirus found in Bcells and epithelial cells that infects
over 90% of adults globally.
95
While primary infection often causes
no symptoms or mild mononucleosis, EBV establishes lifelong latent
infection in B lymphocytes and oropharyngeal and salivary gland
epithelial cells.
79,96
This latent infection is related to several
malignancies including Burkitt's lymphoma, Hodgkin's disease, non
Hodgkin's lymphoma, HNSCC.
90
According to different reports, EBV
DNA was detected in more than 60% of invasive SCC with a strong
association with lesion severity.
8
The mechanisms of EBV in
carcinogenesis are complex including induction of a local immune
suppression, and immortalization of infected cells via manipulation of
the cell cycle and apoptosis pathways.
79
Malignant transformation and
tumorigenesis in epithelial and lymphoid tissues are started by
targeting several host cellular pathways mainly through EBV proteins
such as latent membrane proteins (LMP1and2), BamHIA rightward
frame 1 (BARF1) and EBVencoded nuclear antigens (EBNAs) and small
noncoding ribonucleic acids (EBER1and2).
96,97
It is also hypothe-
sized that EBVrelated cancers may arise from the reactivation of the
virus, potentially triggered by the influence of cofactors such as
concurrent infections.
97
The immunosuppression and chronic antigenic
stimulation by EBV reactivation can result in viral replication, spread,
and establishment of new latent viruses in other cells that contribute
to the development of oncogenesis.
96
Interestingly, EBV and HRHPVs
coinfection is frequently detected in different cancers especially oral
cavity cancers (OCC).
96,98
In 2022, Rahman et al. reported the
prevalence of HPV/EBV coinfection in 11.9% of the combined oral
squamous cell carcinoma (OSCC) and oropharyngeal squamous cell
carcinoma (OPSCC) among a total of 1820 cases from different
studies.
96
In the case of OPSCC as the most common malignancy of
the head and neck, 15%20% of carcinomas are detected as HPV/EBV
coinfected.
96
However, there is a wide geographic variation of HPV
and EBV dual positivity. The highest HPV/EBV coinfection rates were
34.7% for OSCC in Sweden and 23.4% for OPSCC in Poland.
96
Co
infection rates were observed to vary from 25% to 70% for SCC of the
tonsils and base of the tongue.
99
HPV/EBV coinfection was also
reported in asymptomatic HPV
+
people,
83
and especially in oro-
pharynx, anorectum, and urethra of men who have sex with men
(MSM).
90
Moreover, EBV shedding was significantly correlated with
the prevalence and persistence of anal HRHPVs infection among HIV
+
MSM.
100
It seems likely to be a cofactor for development of anal and
penile cancers in these people.
101103
Furthermore, coinfection with
HPV and EBV in oral leukoplakia can be associated with severe
dysplastic changes.
84
Numerous studies reported the HRHPVs/EBV
coexistence ranging from 27.8% to 100% in HPVrelated cervical
cancer, and the potential cooperation of EBV as a cofactor in this
cancer.
82,83,104
Additionally, a published metaanalysis in 2018
demonstrated that coinfection with HPV increases the risk of cervical
cancer in EBV
+
women up to fourfold.
105
Precancerous cervical
lesions were associated with a twofold increase in EBV
+
women
compared to EBV
women.
83
Furthermore, a few studies reported the
HPV/EBV copresence in other epithelial cancers such as breast
cancer, prostate cancer, and nasopharyngeal carcinomas.
106
Figure 1
indicates a hypothetical model of HRHPVs/EBV cooperation for the
development of cancer. The frequent codetection of these two
oncogenic viruses in different types of cancers suggests their
cooperation in driving malignancy through different complementary
mechanisms that will be described in the next section.
8.2.1 |HPV promotes EBV entry, latency and lytic
cycle activation
The in vitro and in vivo studies suggested that complement
receptor type 2 or CD21 (expressed variably in epithelial cells and
detected mainly in B cells) is responsible for attachment and entry
of EBV.
104,107
Dysplastic changes in oral epithelial cells are
significantly dependent on CD21 expression level that is higher in
EBV and HPVinfected cases compared to HPV
/EBV
ones.
98
C3d is also another component of the complement system that is
widely expressed in the cervix and can bind to EBV. This
attachment may also protect the virus from complement
mediated lysis, and makes the cervical epithelium more sensitive
to various oncogenic stimuli.
106
In addition to CD21, the Ephrin
receptor A2 is the epithelial EBV receptor that is overexpressed
in HPVrelated cervical neoplasia (CN) compared to normal
cervical tissue.
96
These findings imply that HPV may help EBV
entrance into epithelial cells and increase the levels of proteins
involved in this process.
98
On the other hand, HPV infection and
its E6 and E7 oncogenes may play roles in the establishment of
latent EBV infection and reduction of EBV replication by changing
gene expression in EBV.
98
The HPV E7 can degrade the Rb, which
can stimulate cell cycle progression independent of p16 inhibition
of cyclin D/cyclindependent kinase complexes. This process
recapitulates the events required to establish EBV latency in
epithelial cells.
8
DNA damage and overexpression of cyclin D1
and human telomerase reverse transcriptase (hTERT) in HPV
infected cells can promote the establishment of EBV latency. It
also increases cell susceptibility to EBV latency which is a crucial
first step in the development of EBVdriven cancer.
96,98
Further-
more, the HPV E6 and E7 oncogenes can stimulate the expression
of an EBV immediateearly lytic gene named as BZLF1 (BamHI Z
fragment leftward open reading frame 1), which favors the
increased EBV genome maintenance and production of EBV lytic
6of21
|
AKBARI ET AL.
genes with oncogenic features.
107
It is interesting to note that
thepresenceofEBVincreasesrateoftheHPV16 and 18
integrations into the host genome. The reports showed that EBV
infection can promote the integration of HPV16 DNA up to
sevenfold.
98,103
It is important to consider that EBV can
permanently change gene expression even after a transient
infection, and the absence of the virus does not necessarily
indicate that it has not played a role in cancer development.
104
8.2.2 |HPV/EBV coinfection mediates immune
evasion, suppression, and modulation
Both HPV and EBV show a large variety of evasion strategies that
interfere with innate and adaptive host immune responses.
8
The
evasion strategies by HPV may generateafavorableenvironment
for EBV secondary infection, and conversely.
104,107
Persistent
HPV infection can inhibit the expression of TLRs such as TLR2,
TLR3, TLR7, TLR8, and TLR9.
96
Furthermore, EBV disrupts TLR
sensing by suppressing the expression of TLR2 through the
proteins expressed by lytic genes such as BGLF5 and BPLF1.
Interfering with TLR9 sensing is also conducted through LMP1
mediated NFκB activation.
96,98
Thus, synergistic effects of HPV
and EBV on downregulating TLRs disrupt the innate immune
recognition of the virus, and facilitate infection.
36
Moreover, HPV
and LMPmediated NFκB activation can induce chronic inflam-
mation in HPV/EBV coinfected organ sites.
108
While the
inflammation is aimed at generating a lethal environment for
pathogens, it paradoxically plays a major role in development of
EBV and HPVinduced malignancies by generating reactive
oxygen species (ROS), releasing growth factors and cytokines,
and causing DNA damage and alterations in critical cell path-
ways.
109
Inflammatory factors and ROS were found to be more
highly expressed in HPV/EBV coinfection compared to mono
FIGURE 1 A hypothetical model of highrisk (HR)human papillomavirus (HPVs)/EpsteinBarr virus (EBV) cooperation for the development
of cancer: HPV genome integration and concomitant amplification of E6/E7 induce genome instability by the degradation of tumor repressors
p53 and Rb; E6/E7 oncoproteins inhibit the antiviral immune responses and induce immune evasion; E6/E7 oncoproteins induce BZLF1
expression, favoring the expression of EBV lytic genes such as BamHIA rightward frame 1 (BARF1) and BCRF1. E6/E7 oncoproteins also
enhance EBV latency; HPV E6/E7 and EBV latent membrane proteins1/BARF1 oncoproteins increase cell immortalization and cell proliferation;
BARF1 and BCRF1 inhibit the antiviral immune responses and induce immune evasion; and HRHPV infection induces CD21 (CR2), which, in
turn, promotes EBV entrance. Purple shapes symbolize HRHPV oncoproteins, and yellow shapes represent EBV proteins. The figure was
created by BioRender.com.
AKBARI ET AL.
|
7of21
infection.
109,110
Downstream impacts of activating NFκBand
signal transducers and activators of transcription 3 (STAT3)
increased additional proinflammatory mediators leading to
inducing mutations, altering signaling pathways, and
promoting cell proliferation and survival, all of which contribute
to cancer development.
79
ROS can also inhibit the immune
response by disrupting Tcell receptor signaling and suppressing
cytokine production. It was also proven to develop radio
resistance in cancer cells, thus hindering the efficacy of
treatment.
98
As the HPV E2, E5, and especially E6 proteins
suppress the interferon regulatory factor signaling and reduce
IFNαand IFNβproduction, the HRHPVs E6 and E7 oncopro-
teins stimulate the BZLF1 expression in EBV that have an
additional effect on decreasing IFNs type 1 production.
111
Similarly, the EBV BRLF1 and BARF1 proteins inhibit both the
synthesis and release of IFNα. BARF1 also downregulates other
human cytokines such as IL8andIL1αthat are related to
antitumor immune responses.
108,112
EBV also expresses a viral IL
10 (vIL10) as a protein homolog encoded by BCRF1genethat
causes local suppression in the cellular immune responses to
HPVtransformed cells.
108
The vIL10 inhibits IFNγ,IL2, IL6,
TNFα,andgranulocytemacrophage colonystimulating factor
(GMCSF) production by CD4
+
T lymphocytes and monocytes
resulting in immune evasion of infected epithelial cells.
104
vIL10
impairs monocyte maturation and host natural defense system
against viral infections.
113
IL10inducedbyHPVE2,E6,andE7
proteins and vIL10 activate Janus kinase/STAT3 cascade that
leads to reduction of TNFα,IL6, and IL1βexpression
interfering with the NFκB signaling pathway with increased
tumorigenic and metastatic ability and epithelialmesenchymal
transition (EMT).
111,114
HPV E6 also interferes with the produc-
tion of other proinflammatory cytokines and chemokines such as
TNFαand GMCSF as well as IL18, IL1βthat are associated
with an increased risk of developing cervical cancer.
114
Thus, the
interaction and cooperation between HPV and EBV proteins
impair host natural antiviral defense system, and produce a
chronic inflammatory microenvironment leading to carcinogene-
sis and tumor progression.
8.2.3 |HPV/EBV coinfection mediates genome
instability and cell proliferation
Longterm expression of EBNA1 and LMP1 in EBVinfected cells
increased the ROS levels leading to DNA damage and oxidative
stress, and subsequently cell immortalization and malignant transfor-
mation.
110
Similarly, exposure of HPVinfected cells to ROS increased
the levels of E6 and E7 proteins which could interfere with the
normal function of tumor suppressor p53 and Rb proteins.
109
LMP1
is the most important EBV immunomodulatory oncoprotein that
enhances the effects of E6 and E7 proteins on p53 and Rb
disruption.
98
It was reported that combination of EBV LMP1 and
HPV16 E6 proteins leads to a decrease in the components of DNA
damage response (DDR) including p27, Rb, and p53.
115
EBNA1 also
has an important role in EBV persistency and maintaining the
EBV genome latently. It decreases p53 and increases EMT and
angiogenesis.
115,116
LMP1 also induces downregulation of
Ecadherin expression and also regulates some transcription factors
related to cell motility in collaboration with LMP2A.
110,117
More-
over, E6 and E7 oncoproteins upregulate the expression of EMT
markers such as Ncadherin, fibronectin, and vimentin that increase
cell migration and invasiveness.
115
In addition, LMP1mediated
NFκB activation induces the expression of DNA binding 1 (Id1)
inhibitors which negatively regulates tumor suppressor p16 thus
increasing cell replication. It also induces cell immortalization by
upregulation of the Bcl2 oncogene and promotion of telomerase
activity via hTERT.
115,116
AlThawadi et al. found an association
between the LMP1/E6 coexpression and upregulation of the Id1in
cervical cancer.
111
LMP1 mediates activation in cancer pathways
such as phosphoinositide 3Kinase/Akt (PI3K/AKT), extracellular
signalregulated kinase (ERK), and cJun Nterminal kinase, which
leads to an increased cell growth, survival, and motility.
118
LMP1/E6
coexpression also promotes cell survival by increasing the check-
point kinase 1, PI3K/AKT, mitogenactivated protein kinase (MAPK),
and NFκB signaling pathways in the HPV/EBV coinfection
cases.
98,115
Thus, coinfection of HPV and EBV cooperatively
expands dysregulation of shared oncogenic pathways.
115
According
to some reports, the expression levels of antiapoptotic proteins such
as survivin and Bcl2 were significantly higher in the HPV/EBV co
infected cases than in noninfected ones.
110,117
Actually, the expres-
sion levels of E6 and E7 proteins have a direct association with
survivin and Bcl2 expression levels. Also, a direct association was
found between the expression levels of LMP1 and survivin.
110
Thus,
the expression of EBNA1, LMPs, and HRHPVs E6 in HPV/EBV co
infection promotes cell proliferation, resistance to apoptosis, and
anchorageindependent growth associated with more aggressive
malignant tumors in cancer, and suggests a synergism between HPV
and EBV.
98
8.3 |Coinfection of HPV with HIV
HIV is a member of the retroviridae family, and the etiological agent
of acquired immunodeficiency syndrome (AIDS).
119
HIV shows
tropism for several cell types including monocytes, dendritic cells,
and epithelial cells; however, its primary target is the depletion of
CD4
+
T lymphocytes through different mechanisms.
120,121
A strong
relationship was observed between genital HPV infection and the risk
of HIV acquisition.
122
Although most HPV infections will spontane-
ously be resolved, but their considerable rates will persist leading to
an increased risk of anogenital dysplasia especially in patients
infected with HIV. HIV and HPV coinfection is common among
people living with HIV (PLWH).
78
Coinfection with HPV and HIV
showed that cervical cancer is the most common AIDSdefining
neoplasm in women. HIV changes the natural history of HPV
infection with decreased regression rates and more rapid progression
8of21
|
AKBARI ET AL.
to highgrade and invasive lesions.
123
Although molecular interac-
tions between these two viruses were not completely understood;
but several mechanisms were proposed for interaction of both
viruses with the immune system.
8.3.1 |Viralviral interaction
HPV infection of the cervix may enhance HIV infection
through induction of immune and inflammatoryrelated protein
production.
124
The identification of HIV within macrophages under-
scores their significance as a crucial reservoir and a possible avenue
for HPVHIV interaction. Furthermore, the interplay between viral
proteins could also play a role in this interaction.
124
HIV1 proteins
can directly lead to tumor growth by interfering with cellular
functions. For instance, HIV1 proteins such as Tat can interact with
the RB2/p130 tumor suppressor gene product and thus increase cell
proliferation.
125
The HIVTat protein was known to facilitate cell
cycle advancement and decrease the presence of cellcycle suppres-
sors.
126
Dolei et al. revealed that exposure to Tat protein led to a
dosedependent rise in the expression of E1 and L1 genes.
127
Meanwhile, Vernon et al. demonstrated that the HIV1 tat protein
enhanced E2dependent HPV16 transcription.
128
Furthermore, the
expression of the HIV rev gene in epithelial cells enabled the
expression of L1 protein in undifferentiated basal keratinocytes.
Generally, the activation of both early and late HPV genes may
account for the increased virulence of HPV in the milieu of HIV
infection. For instance, HIV viral protein R (Vpr) facilitates the
infection of macrophages, and also disrupts the G2/M checkpoint
with induction of apoptosis that is potentiated by HPV16 E6.
123
Toy
et al. suggested that Vpr may be useful as a cytostatic agent in
treatment.
129
Not only HPV may increase the susceptibility to HIV
infection, but also progression of HIV infection may be facilitated by
simultaneous HPV infection. Luque et al. showed the correlation of
active HPV infection with high HIV plasma RNA levels.
130
Moreover,
the HPVinduced inflammatory cytokines especially IL6 cytokine
may stimulate HIV p24 expression in monocytes by binding to the
CAAT/enhancer binding protein B followed by activation of a
cascade of acute phase reactants and cytokines.
123
HIV/HPV coinfection was related to an increased risk of
progression of CIN to cervical cancer. The results suggested that
coinfection of HPV and HIV led to a significant increase in the
vascular endothelial growth factor A, p27, and Elf1 expression
compared to HPV
+
/HIV
(i.e., single HPV infection) infection that
could facilitate viral persistence and invasive tumor development.
124
Also, expression of RANTES in HIV/HPV coinfection influenced the
development of CIN leading to the progression to cervical cancer.
Indeed, cervices from HIVpositive patients exhibited HIVNef
protein in the cells mainly around blood vessels, and a decreased
expression of cyclooxygenase1 (COX1) and TGFbRI, while RANTES
was highly expressed in the HIV/HPV and HPVinfected patients
compared to controls.
131
Moreover, HIV1 infection increased the
levels of COX2 and systemic prostaglandin E2 in women with
cervical HPV suggesting a positive correlation with plasma HIV1
RNA levels.
132
The host immune response to HPV is mediated by
Tlymphocytes.
133
This response may increase the risk of HIV since
T lymphocytes are primary target cells for HIV, and are upregulated in
HPVinfected cervical tissues.
3
For instance, the IL1βcytokine
directly acts on Tlymphocyte expansion and differentiation in vivo to
induce protective immunity against microbes and autoimmune
inflammation. Increased levels of IL1βwhich activates HIV
promoter
134
were detected in HPVassociated abnormal cervical
cytology.
135
8.3.2 |Relationship between the CD4 lymphocyte
count and HPV infection
Although low CD4 lymphocyte count is related to a higher
prevalence of HPV; but however, the correlation of the CD4
lymphocyte count with CIN or cervical dysplasia is still
controversial.
78,136,137
It was reported that lower CD4 counts
increased the risk of vulvar dysplasia.
138
In general, immuno-
suppression and more advanced stages of HIV infection enhanced
the risk of HPV infection. Several studies showed an increased
risk for HPV infection in PLWH with a large CD4 depletion (i.e.,
CD4 counts <200 cells/μL).
78
High HPV load was related to a 10fold
increased risk of CIN among HIVpositivewomenwithsevere
immunosuppression compared to women with higher CD4
+
counts.
139
Acrosssectional study in China reported an increased risk for anal HPV
infection in PLWH with CD4 counts <200 cells/μL. Moreover, a CD4
lymphocyte count <350 cells/μL was also related to an increased risk for
anal infection.
140
Similarly, a crosssectional study in Spain reported a
lower CD4 lymphocyte count among MSM with an HPV infection.
141
Several observational studies also showed an increased risk for cervical
infection in PLWH with a baseline CD4 lymphocyte count of <100 cells/
μL.
142,143
On the other hand, HIVpositive women in Africa showed
higher viral load for combined alpha9HPVspeciescomparedto
HIVnegative women. Moreover, HIVpositivewomenwithCD4counts
>350/μL had significantly lower viral loads for alpha7HPVspeciesthan
HIVpositivewomenwithCD4350/μL, but low CD4 count was not
significantly associated with increased viral load for other HPV
species.
144
8.3.3 |Relationship between antiretroviral therapy
(ART) and HPV
ART provides a normal life span to PLWH by maintaining an adequate
immune status along with the virological suppression of HIV. Several
studies showed the relationship between ART and the prevalence of
HPV. Different researchers reported a lower prevalence of cervix
HPV infection in PLWH receiving ART compared to untreated
patients.
145
In addition, a longer duration of ART among women was
associated with a lower prevalence of HRHPV infection in Kenya
146
suggesting higher immune control and clearance of HPV infection in
AKBARI ET AL.
|
9of21
the cervix.
145
Lower progression risk from lowgrade squamous
intraepithelial lesions to highgrade squamous intraepithelial
lesions was reported among women who used ART.
147
In contrast,
another study reported that ART had a protective effect against
CIN2
+
but not against invasive carcinoma.
148
Several studies
indicated a lower prevalence of highgrade anal dysplasia in patients
receiving ART than in ARTnaïve patients.
149,150
HRHPVpositive
women showed higher circulating levels of T cells expressing
activation/exhaustion markers (CD38, programmed cell death protein
1, CTLA4, Band Tlymphocyte attenuator, and CD160), Tregs, and
myeloid subsets expressing corresponding ligands (PDL1, PDL2,
CD86, CD40, herpesvirus entry mediator) than HRHPV negative
women. In ARTsuppressed HIVinfected women with HPV co
infection, the levels of T cell (i.e., CD4
+
T cell) and myeloid cell
activation/exhaustion were associated with the presence of HRHPV
genotypes.
151
It was also reported that the cellular immunity induced
by the bivalent or quadrivalent vaccine in PLWH was importantly
similar to that in people without HIV.
152
However, a stronger
humoral immune response was detected in patients receiving a
bivalent vaccine compared to those receiving a quadrivalent
vaccine.
153
However, HPV vaccination in HIVpositive individuals
seems to be a good idea, but its efficacy remains unproven.
154
8.3.4 |HPV genotypes found among coinfection
with HIV
Several HPV genotypes were found among HPV and HIVcoinfected
people. HPV16 was the most common oncogenic virus in cervical
cancer among HIVnegative and also HIVpositive women.
155157
However, HPV16 is responsible for a smaller proportion of invasive
cancers in HIVinfected women indicating that immunecompromise
does not further increase its oncogenic potential. However, other
genotypes (e.g., HPV types 18, 45, and 35) were also observed
among cervical cancer specimens
158160
that can be linked to
HIV.
159,161
For example, the most prevalent HPV types were HPV
56 and HPV16 in Brazilian patients,
136
and HPV52 and HPV58 in
Chinese patients.
162
Moreover, HPV genotyping from cervical
samples of HIVpositive patients in São Paulo showed that HPV56
was the predominant genotype followed by HPV16, HPV81, HPV
62, and HPV83. HPV types 6, 18, 26, 33, 52, 59, 72, 74, 90, and 114
were also observed at lower rates in these samples, respectively.
136
However, immunodeficiency can likely contribute to an increased
susceptibility to other types of HPVs in HIVinfected patients. The
interaction of HIV and nononcogenic HPV is unclear. It is possible
that immune suppression contributes to the development of warts
(large and problematic warts), especially in tobacco smokers.
163
Although ART may reduce the size and recurrence risk of warts,
but this is not consistent, and genetic factors may influence this
interaction.
164
More HPV types are present in warts of immunosup-
pressed women, but HPV6 and HPV11 types are the most reported
types.
154,163
8.3.5 |HPVrelated cancer and their interplay
with HIV
The International Agency for Research on Cancer classified both
HPV and HIV1 as carcinogens.
165
Indeed, HPV is a direct
carcinogen and HIV1 is an indirect carcinogen through immune
suppression.
165
PLWH are at a HR of developing HPVrelated
cancers. Indeed, HIV positivity was linked to an increased
prevalence of cervical HPV infection and CIN.
166
Treatment of
CIN in HIVpositivewomenfailsmoreoftenthaninHIVnegative
women. In contrast, highly active antiretroviral therapy (HAART)
showedaprotectiveeffectontherecurrenceofCIN.
167
Gilles
et al. reported higher recurrence rates after treatment of CIN in
HIVpositive women, and protection from recurrence after a
good viral response to HAART.
168
Moreover, high prevalence of
anal HPV infection, anal precancerous lesions, and anal cancer
in HIVpositive individuals were reported in men and
women.
165,167,169,170
Other HPVrelated cancers such as OPC have
also been linked to HIV infection.
170
The HIVinfected people
showed higher rates of OPC than HIVuninfected people.
165
Massad et al. studied the prevalence of genital warts and vulvar
intraepithelial neoplasia in HIV
+
and HIV
individuals, and showed
that the prevalence of warts was higher in HIV
+
than in HIV
subjects.
163
8.4 |HPV coinfection with other viruses from
different families
Some studies reported the copresence of HPV and CMV in both
cancerous and noncancerous cervical samples.
82,171
CMV has also
been observed in cervical cancer samples with an impact on
increasing the integrated or mixed HPV16 genome up to six
fold.
171
It was hypothesized that the expression of immediateearly
genes such as IE1 and IE2 in CMV activates other viral and cellular
genes in infected cells.
171
In fact, CMV infection can serve as a
transformationinitiating factor in development of cervical cancer
according to hitandrun theory. Thus, CMV infection may increase
the susceptibility to subsequent HPV infections and the risk of
carcinogenesis.
172
Possible cooperation of HPV and HHV6 was
previously reported in development of intraepithelial cervical
lesions.
173
Moreover, HHV8, also called Kaposi's sarcoma
associated herpes virus was transmitted during oral, vaginal, and
anal sex, and associated with several malignancies.
174
Few studies
reported HHV8/HPV coinfection in immunocompromised patients
and cervical cancer up to 25%.
83,174,175
Based on the studies
conducted so far, the induction of chronic inflammation by HHV8
may contribute to development of a tumorpromoting micro-
environment through production of IL6, IL8, macrophage migration
inhibitory factor, and different chemokines.
83,175,176
Moreover,
several studies have suggested the potential role of oncogenic
polyomaviruses particularly BK virus, JC virus, and Merkel Cell
10 of 21
|
AKBARI ET AL.
Polyomavirus in development of HPVassociated malignancies such
as cervical and oral cancer.
177180
These viruses infect epithelial cells
in a latent state and have a large T antigen that can block the
functionality of p53 and Rb family members in the cell leading to
tumor development.
8
Additionally, human T lymphotropic virus type
1 (HTLV1) is a sexually transmitted pathogen that has shared
transmission routes with HPV.
181
The studies showed that HTLV1
infection is associated with a higher prevalence of HPV acquisition,
particularly HR types. However, there is limited evidence on the
impact of HTLV1/HPV coinfection in the development of cervical
cancer.
182,183
According to some studies, the prevalence of HIV,
HCV, and Treponema pallidum (causative organism of syphilis) was
relatively higher in HPV
+
patients than in the controls which showed
the increased possibility for the incidence of bloodborne infectious
diseases among HPVinfected individuals.
184,185
Similarly, higher
prevalence of Torque teno virus in HPV
+
patients was reported in
some studies that can be attributed to the same mode of
transmission and stimulation of the immune system by HPV
infection.
186,187
Thus, further research is needed to fully understand
the impact of coinfection of HPV with other viruses on the
development of HPVrelated malignancies.
9|MICROBIOME AND HPV
A robust and healthy human microbiome plays a crucial role in
protecting the host from a broad range of foreign pathogens and
diseases.
188
It reduces inflammation and allows normal mucosal
function.
79
Conversely, dysbiosis (imbalance) of the microbiome has
been linked to chronic inflammation and the pathogenesis of mucosal
diseases.
189
It has profound effects on epithelial surface integrity,
mucosal secretion, and immune regulation.
188
As mentioned above,
in the context of HPVrelated cancers, viral infection serves as a
necessary agent but inadequate cause of cancer development.
37
Co
factors such as the immune response to viral infection, the host
microbiome health, or other acquired infectious agents play
additional roles in carcinogenesis.
97
The microbiome is an important
contributor to chronic mucosal inflammation, thus changes in its
composition by overgrowth or undergrowth of different bacterial
populations in multiple organ sites have been associated with
progression to HPVassociated dysplasia.
79
It was proven that
bacterial shifts in the microbiome or coinfection with other bacterial
pathogens in the organ affected by HPV can modulate viral
proliferation and infection.
97
Coinfection/copresence of some
bacteria (either as part of the normal microbiome or as causative
agents of infections) with HPV can be assessed by DNAbased or
serology tests. To better understand the bacterialviral interactions
involved in the initiation, development, and progression of HPV
related cancers, it may be useful to compare the microbiome of
healthy individuals with HPVinfected patients.
78
The HPV, micro-
biome, and bacterial coinfections are described in the development
of HPVrelated diseases in the next sections.
9.1 |Microbiome and HPVrelated cervical cancer
The relationship between HPV and the cervicovaginal microbiome
has widely been studied in recent years. Cervicovaginal mucosa acts
as a protective barrier against pathogens entering the upper part of
the female sexual tract. However, it is often compromised by
pathogen spreading and dysbiosis.
190
In cases of HPV infection,
inflammation of the cervix can act as a cofactor for severe lesions.
Prolonged inflammation exposes the tissue to ongoing genotoxic
effects, leading to different forms of cancers.
191
Chronic inflamma-
tion can also accelerate the development of cervical cancer as well as
the initial alterations caused by HRHPVs.
192,193
Multiple studies
have proven that women with a specific cervicovaginal microbiota
composition may be more likely to acquire HPV or to show a faster
progression to CN.
194,195
Additional antigenic stimuli in the concur-
rent bacterial infection change the immunological responses, and
decrease the clearance of HPV.
185
In general, different cell stresses
such as high vaginal pH, production of nitrosamines by anaerobic
bacteria, and secretion of proinflammatory cytokines such as
interleukins (IL1β,IL6, and IL8) in cervicovaginal dysbiosis enhance
the risk of mutation, and the oncogenic potential of HPV.
193,196199
9.1.1 |BV and HPV
Multiple studies showed that the cervicovaginal microbiome of women
with persistent HPV infection is characterized by a high abundance of
anaerobic species from different genera.
188
An overgrowth of some
commensal bacteria was reported including Pseudomonas, Brevibacter-
ium, Peptostreptococcus, Delftia, Anaerococcus tetradius, Atopobium,
Shuttleworhia satelles, Megasphaera elsdenii, Fusobacteria,andSneathia
spp. with a decrease in Lactobacillus spp.population.
200202
Further-
more, changing in microbial diversity as a result of BV was more
pronounced in HPVinfected women.
188
BV is associated with major
changes in the vaginal environment by chronic inflammation because of
reduced levels of antiinflammatory molecule secretory leukocyte
protease inhibitor. It also induces epithelial barrier disruption by
producing epithelialliningdegrading enzymes that help HPV en-
trance.
203
In addition, the production of IL1βand IL10 in BV impairs
cytotoxic Tcell response and promotes HPV persistence and cervical
dysplasia.
193,195
Some reports suggested that lipopolysaccharide from
anaerobic bacteria in BV can interfere with tumor suppressors (e.g., p53
and Ecadherin) which are also targeted by HPV oncoproteins.
97
Comparative genomic analysis of several studies showed that Gardner-
ella vaginalis,
195,204
Mycoplasma spp.,
196,205,206
and Ureaplasma urealy-
ticum
195,206209
are the causal bacteria of BV that have a high
association with severe cervical lesions. These bacteria have been
identified as the main cofactors of HPVs.
193,196
Other lower genital
tract infections including aerobic vaginitis and desquamative inflamma-
tory vaginitis are more common in HPV
+
women.
210
These infections
caninduceinflammationwithanincreaseinvaginalleukocytesandIL
1βand IL6 levels leading to a progressive CIN.
210,211
AKBARI ET AL.
|
11 of 21
9.1.2 |Bacterial sexually transmitted infections
(STIs) and HPV
Studies have also identified a possible association between STIs and
HPV infection.
210
The imbalance in the resident microbiota increases
susceptibility to upper genital tract infections and bacterial STIs
demonstrated the correlation of Gemella that have a significant
association with HPV infection by mucosal barrier disruption.
212
Neisseria gonorrhea (NG), Mycoplasma spp., and Chlamydia trachoma-
tis are the main bacterial STIs that provide an ideal niche for HPV
persistence and CN development.
195,197,200
They can interact with
HPV to develop HPVrelated dysplasia lesions in the cervix or the
anus by causing further localized inflammation.
213215
The Myco-
plasmataceae family consists of two genera, Mycoplasma and
Ureaplasma, and is reported as the most significant differential
cervicovaginal bacteria between normal and precancerous cervical
cytology.
210,216
Mycoplasma hominis and Mycoplasma genitalium/HIV
coinfection are prevalent in women. Also, these HIV
+
women are
mostly infected with multiple HPV genotypes like HPV16 and HPV
56.
196,217
Mycoplasmataceae represents a family of intracellular
bacteria with persistent infection that makes a cytokinemediated
inflammatory environment. Inflammation diminishes the microvilli in
cervical epithelial cells which favors virus entrance, persistence, and
HPVassociated dysplasia.
218
They enhance carcinogenesis by
expressing proteins that can change biological mechanisms such as
programmed cell death and cellular metabolism.
97
According to some
reports, NG also increases the risk of cell transformation indepen-
dently, or in conjunction with HPV.
97
NG may interact with HPV by
dysregulation of multiple cellular signaling pathways and inducing
DNA doublestrand breaks (DSBs). It enhances the expression of pro
inflammatory cytokines and cyclindependent kinase inhibitors p21
and p27, and decreases p53 expression.
97,219
Moreover, Trichomonas
vaginalis as a protozoan parasite STI causes microlesions in the
cervical epithelium, and reduces the vaginal protective mucus layer
leading to the spread of HPV infection into the basal layer of the
cervical epithelium.
212
It also can increase the risk of cervical cancer
by developing an inflammatory environment through production of
nitric oxide (NO) by neutrophils in the cervix, which enhances DNA
damage, growth of abnormal cells, HPV entrance, and persistence.
220
TV infection can overexpress viral tumorigenesis which helps
activation of cancercausing pathways.
191
On the other hand, CT
has received more research interest as the most important pathogen
associated with HPV infection compared to other cervicovaginal
bacterial agents. Figure 2indicates the effects of C. trachomatis
infection and microbiome dysbiosis with HRHPVs in cancer
development. Therefore, we will further discuss its effects on the
increased risk of HPVdriven cervical abnormalities.
9.2 |HPV/C. trachomatis coinfection
C. trachomatis is a gramnegative intracellular bacterium and the most
common bacterial STI among various sexually transmitted agents in
young people.
221
Although it is frequently asymptomatic, genital CT
infection in women has been associated with several severe diseases
including tubal factor infertility, pelvic inflammatory disease, chronic
pelvic pain, ectopic pregnancy, venereal lymphogranuloma, uterine
cervical lesions, newborn pneumonia, trachoma, and conjunctivi-
tis.
194,222,223
The connection between CT infection and HPV retention
was studied in various regions of the world.
213,224227
These studies
suggested that CT is more prevalent in women who are infected with
HPV than in those who are not, so it makes HPV as a risk factor for
CT infection, and conversely.
224,225
Furthermore, several studies
demonstrated an association between HPV/CT coinfection and
developing abnormal cervical cytology and dysplasia.
213,226,227
The
HPV and CT DNAs can be detected in approximately 99% of cervical
cancer cases.
228,229
There is a mutual benefit in HPV/CT coinfection.
Indeed, CT affects the cervical microenvironment through promotion of
inflammatory conditions, and allows HPV penetration into the epithelial
cells. HPV also helps CT to spread and multiply by reduction of cell
mediated immunity.
228,230232
Dysregulation of metabolic signaling and
immune responses, chronic inflammation, epithelial barrier breach,
uncontrolled cellular proliferation and apoptosis, genome instability, and
angiogenesis activation are different processes that appear to be crucial
in HPV/CT coinfection and CN progression.
194
In the next sections, we
will review some mechanisms of HPV/CT coinfection that may evoke
cervical cancer.
9.2.1 |HPV/CT coinfection and genome instability
The correlation between HPV and CT infections with cervical tissue
transformation is based on the steady presence of both pathogens
that interfere with the host cells detection and repairing
mechanisms.
233
Thus, HPV/CT coinfection leads to tissue damage,
and reduces local regenerative capacity.
234
Various proteins ex-
pressed by CT can target different subcellular compartments such as
nucleus, endoplasmic reticulum, and mitochondria. These proteins
may have detrimental effects on essential biological functions leading
to cancer development.
235
The coexistence of DNAbinding proteins
from both human and CT within the nucleus creates a competitive
environment that may disrupt the binding of normal human proteins
to DNA, and increase the risk of malignancy.
233,235
Some of these
proteins recruit direct DSBs as the most dangerous form of DNA
damage due to their unrepaired nature.
226,236,237
Furthermore, CT
suppresses DNA repair activity by recruiting DDR proteins like
Ataxiatelangiectasiamutated (ATM) away from sites of DSBs.
238
ROS production also causes oxidative DSBs and senescence
associated heterochromatin foci by subverting the host histones in
CT infection.
228
Free radicals damage the DNA and DNA repair
factors, and prevent apoptosis resulting in genetic fragility.
236,237
In
contrast to CT, HPV E6, and E7 proteins stimulate ATMdependent
homologous recombination, and activate the mismatch repair (MMR)
system to maintain cellular and genome integrity, which is necessary
for viral replication.
236,237
In the case of HPV/CT coinfection, CT
impedes the HPVinduced MMR system at both the transcriptional
12 of 21
|
AKBARI ET AL.
and posttranslational levels.
239241
It reduces the transcription of the
MMRrelated genes by degrading the transcriptional factor E2F1.
242
CT infection also can modulate the protein serine/threonine
phosphatase 2A signaling pathway to suppress ATM activation,
which prevents cell cycle arrest. This contributes to a deficient high
fidelity HR pathway, and creates a conducive environment for further
mutagenesis.
242
9.2.2 |HPV/CT coinfection and cell proliferation
The replication and propagation of intracellular pathogens can be
limited by cell death process.
226
Thus, manipulation of cell survival
and death pathways by CT can cause further cellular transformation
in HPVinfected individuals.
234
CT creates a vacuole surrounded by a
membrane named as inclusionin which it can replicate and be ready
to infect other cells.
239241
Both CT infection and HPV/CT co
infection trigger the oncogenic MAPK pathway (RasRafMEKERK)
that regulates diverse cellular functions including cell proliferation,
survival, differentiation, and migration, thus promoting cancer cell
growth.
243,244
CT was also reported to activate PI3K/AKT that
enhances cellular proliferation, and blocks cell apoptosis.
238
Addi-
tionally, CT manipulates intrinsic apoptotic pathways by degrading
the MDM2 protein and inducing expression of antiapoptotic proteins
like Mcl1.
234
It also creates mitotic spindle defects causing the
premature host cells to exit from mitosis without the right
corrections.
226
Furthermore, cytokinemediated inflammatory
responses caused by chlamydial infection lead to fibrosis, tissue
dysfunction, and EMT by deposition of extracellular matrix
proteins.
245
These processes increase infected cell motility and
invasiveness, and decrease cell senescence and apoptosis, thus
promoting tumor progression.
194
On the other hand, EMT inducers
such as TGFβdownregulate protective modulators, and upregulate
fibrogenic and oncogenic modulators including miRNAs and tran-
scription factors which are associated with cellular transformation
and neoplasia.
246
It was reported that CT causes cervical epithelial
neoplasm by increasing Ki67 expression and decreasing p53 levels
due to the overexpression of E6 and E7 proteins. Ki67 is a cell
proliferation marker of cervical epithelium that is associated with
lesion intensity, and can be overexpressed during HPV/CT
coinfection.
247
Moreover, during CT persistent chronic infection, a
large quantity of 60kDa heat shock proteins (named as CHSP601)
are produced that interfere with host apoptosis and cellular
senescence processes.
248
Concomitant presence of HPV oncopro-
teins during CHSP601 expression may lead to survival of apoptotic
stimuli, uncontrolled proliferation, and finally neoplastic transforma-
tion. Thus, it can provide favorable conditions for HPV persistence
and proliferation of HPVinfected cells.
224
FIGURE 2 Possible outcomes of highriskhuman papillomavirus (HPVs) coinfection with a Chlamydia trachomatis. (A) Longterm silent
chlamydia infection and microbiome dysbiosis can contribute to the inflammation via cytokines and chemokines secretion or bacterial
metabolites directly. C. trachomatis also induces cell proliferation and chromosomal instability by dysregulating cellular pathways; (B) HPV can
infect the cells through epithelial barrier disruption, and cause further cellular transformation and immune suppression; and (C) These immune
activities contribute to the inflammation and tumor microenvironment, and further influence the cancer development. The figure was created by
BioRender.com.
AKBARI ET AL.
|
13 of 21
9.2.3 |HPV/CT coinfection and immune response
The coinfection of CT and HPV (especially the 16, 18, 31, 33, 53, and
56 genotypes) is considered the most important risk factor for the
development of cervical cancer.
228
During the initial stages of HPV
infection, LCs play a critical role in initiating the immune response
against the virus.
249
They present antigens to T cells, and help immune
system to recognize and eliminate the virus.
250
HPV infects the host cell
persistently by suppressing this process.
251
Lu et al. demonstrated that
decreasing antigen presentation ability of LCs and its density are
significantly higher in the HPV/CT coinfection cases than those in HPV
and CT single infections. In addition, chlamydia proteaselike activity
factor causes more immune dysfunction and viral persistence by
reducing the expression of MHCII and costimulatory molecules on LCs
in HPV/CT coinfection.
251
Indeed, activation of PI3K/AKT and MAPK
signaling pathways by HPV/CT coinfection can contribute to immune
evasion and promote viral progression.
243
Coinfection also leads to a
decrease in the number of CD4
+
and CD8
+
T cells, further suppressing
the cellular immune response against HPV and potentially leading to the
development of CN.
251
As HPV uses several molecular strategies to
evade innate and adaptive immunity, thus inflammatory patterns are
uncommon in monoHPV infection.
228
Instead, HPV infection is
characterized by a high number of regulatory Tcells and activated
Th2 cells. This immunosuppressive microenvironment (through macro-
phage type 2 induction and IL10expressionbyHPVE2,E6,andE7
proteins) can be enhanced by TGFβderived from bacteria, and creates
a positive feedback loop between microbiota and cytokine profile.
252
It
also suppresses cytotoxic functions that lead to T cell anergy and may
explain the increased acquisition rate of other pathogens such as
CT.
228,235
The cytokine profiles of HPV/CT cervical samples showed the
increased levels of proinflammatory cytokines including IL1β,IL6, and
TNFα, and the reduced levels of antiinflammatory cytokines including
IL4andIL10 compared to single infections or uninfected controls. This
cytokine profile of HPV/CT coinfected samples was associated with
the severity of cervical lesions.
253
Moreover, both infections increased
the TNFmediated immune response, but only CT specifically induced
the inflammatory responses of IL17 and NFκB.
250
CT could activate
the innate immune response through the TLR2 and TLR4 resulting in the
activation of NFκB.
228,249
NFκB then induced the synthesis of pro
inflammatory cytokines including IFNγ,IL6, IL8, IL10, and IL12 as
well as the recruitment of neutrophils/macrophages.
251
In addition, CT
infection increased the production of IL6andTGFβcytokines that are
often associated with tumor progression.
228,249
The expression of these
cytokines was further enhanced in the presence of HPV E6 and E7
oncogenes
226
indicating a synergistic effect of HPV and CT infections in
promoting cancer development.
9.3 |Microbiome and other HPVrelated cancers
The carcinogenic process induced by HPV has been extensively
studied in cervical cancer, but the findings have been extrapolated
to other HPVrelated cancers like HNSCC, based on similarities in
the epithelial cells.
188
The underlying conditions such as poor dental
status, especially in heavy alcohol consumers and/or smokers are
correlated with shifts in the oral microbiome.
97
Expression of NO
and proinflammatory cytokines such as TNFα,IL1β,andIL6in
oral dysbiosis induce chronic inflammation and modulation of
immune response. This compromised immune system and inflam-
matory environment make the host more susceptible to HPV
infection.
254
Furthermore, production of toxins and carcinogenic
metabolites such as acetaldehyde in oral dysbiosis activate signaling
pathways that promote cell proliferation and survival and damage
the DNA of oral epithelial cells leading to an increased risk of HPV
associated malignancies.
80
According to some epidemiological
studies, the oral microbiome in HPVinfected patients was enriched
in anaerobic bacterial families such as Prevotellaceae, Veillonellaceae,
Campylobacteraceae,andBacteroidetes.
255
Actinomycetaceae family
as a common agent in periodontal disease, and a risk factor for
HNSCC was also more abundant in HPV
+
patients.
79,255
Other
studies showed the association of Selenomonas spp., Haemophilus,
Fusobacterium naviforme, and STI pathogens (e.g., CT or NG) with
oral HPV infection.
79,256258
Additionally, some reports demon-
strated the correlation of Gemella and Leuconostoc with HPV
positive OCC and OPC, and the association of Streptococcus
anginosus with progression of OPSCC.
80,188
On the other hand,
Selenomonas noxia, Actinomyces, Granulicatella, Oribacterium, Cam-
pylobacter genera,Veillonella dispar, Rothia mucilaginosa,and
Haemophilus parainfluenzae were reported more prevalent in HPV
+
OPC patients compared to HPV
OPC.
254
Thus, oral bacterial
infections may act as an adjuvant risk factor and facilitate HPV
carcinogenic processes in some HNSCCs by altering the gene
expression directly (through virulence factors) or indirectly (through
oxidative stress and inflammation).
79
There is limited evidence to
support the interaction of anal and penile microbiome with HPV
infection. Furthermore, research characterizing the microbiome of
other HPVdriven dysplasia is relatively nonexistent. Regarding the
association between penile microbiota and HPV infection, Onywera
et al. reported higher relative abundances of BVrelated bacteria,
especially Prevotella, Peptinophilus,andDialister in HPVinfected
menincontrasttomenwithCorynebacteriumdominated penile
microbiota that are less likely to have HRHPVs.
259
10 |CONCLUSION
In conclusion, coinfections play a significant role in HPVassociated
diseases by influencing the dynamics of HPV infection and
subsequently disease progression. Different infectious agents espe-
cially viruses and bacteria can interact with HPV leading to
complicated outcomes. The intricate interplay of HPV with co
infecting agents in diverse human tissues, and its impact on
carcinogenesis and disease progression calls for continued research
to devise comprehensive preventive and therapeutic strategies,
ultimately improving the management of HPVrelated malignancies
and associated diseases. Understanding these interactions is crucial
14 of 21
|
AKBARI ET AL.
for comprehensive management and prevention strategies against
HPVassociated diseases. Thus, further research is needed to
uncover the precise mechanisms underlying these coinfections and
their implications for HPVrelated cancer development.
AUTHOR CONTRIBUTIONS
Elahe Akbari, Alireza Milani, and Masoud Seyedinkhorasani wrote the
original draft and designed the figures. Azam Bolhassani was
responsible for conceptualization, review, and editing. All authors
approved the final manuscript.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
DATA AVAILABILITY STATEMENT
All data are available in the manuscript.
ORCID
Azam Bolhassani http://orcid.org/0000-0001-7363-7406
REFERENCES
1. Szymonowicz KA, Chen J. Biological and clinical aspects of HPV
related cancers. Cancer Biol Med. 2020;17(4):864878.
2. Shope RE, Hurst EW. Infectious papillomatosis of rabbits. J Exp
Med. 1933;58(5):607624.
3. Khare P, Jain A. Immunopathology, Diagnosis and Treatment of HPV
Induced Malignancies. Academic Press; 2022:8197.
4. Schiffman M, Doorbar J, Wentzensen N, et al. Carcinogenic human
papillomavirus infection. Nat Rev Dis Primers. 2016;2(1):16086.
5. Shimizu A, Yamaguchi R, Kuriyama Y. Recent advances in
cutaneous HPV infection. J Dermatol. 2023;50(3):290298.
6. Omone OM, Gbenimachor AU, Kozlovszky M. Using Algorithms for
the Prediction of Lowrisk and Highrisk Human Papillomavirus
(HPV) in Males. 2020.17
7. Martinelli M, Musumeci R, Sechi I, et al. Prevalence of human
papillomavirus (HPV) and other sexually transmitted infections
(STIs) among Italian women referred for a colposcopy. Int J Environ
Res Public Health. 2019;16(24):5000.
8. Guidry JT, Scott RS. The interaction between human papilloma-
virus and other viruses. Virus Res. 2017;231:139147.
9. ViverosCarreño D, Fernandes A, Pareja R. Updates on cervical
cancer prevention. Int J Gynecol Cancer. 2023;33(3):394402.
10. Liao G, Jiang X, She B, et al. Multiinfection patterns and co
infection preference of 27 human papillomavirus types among
137,943 gynecological outpatients across China. Front Oncol.
2020;10:449.
11. Kombe Kombe AJ, Li B, Zahid A, et al. Epidemiology and burden of
human papillomavirus and related diseases, molecular pathogenesis,
and vaccine evaluation. Front Public Health. 2021;8:552028.
12. Farahmand M, Moghoofei M, Dorost A, et al. Prevalence and
genotype distribution of genital human papillomavirus infection in
female sex workers in the world: a systematic review and meta
analysis. BMC Public Health. 2020;20(1):1455.
13. Guan P, HowellJones R, Li N, et al. Human papillomavirus types in
115,789 HPVpositive women: a metaanalysis from cervical
infection to cancer. Int J Cancer. 2012;131(10):23492359.
14. Mejía L, Muñoz D, Trueba G, Tinoco L, Zapata S. Prevalence of
human papillomavirus types in cervical cancerous and pre-
cancerous lesions of Ecuadorian women. J Med Virol. 2016;88(1):
144152.
15. Swangvaree SS, Kongkaew P, Ngamkham J. Frequency and type
distribution of human papillomavirus from paraffinembedded
blocks of high grade cervical intraepithelial neoplasia lesions in
Thailand. Asian Pac J Cancer Prev. 2013;14(2):10231026.
16. Pista A, de Oliveira CF, Lopes C, Cunha MJ. Human papillomavirus
type distribution in cervical intraepithelial neoplasia grade 2/3 and
cervical cancer in Portugal: a CLEOPATRE II study. Int J Gynecol
Cancer. 2013;23(3):500506.
17. Callegari ET, Tabrizi SN, Pyman J, et al. How best to interpret
mixed human papillomavirus genotypes in highgrade cervical
intraepithelial neoplasia lesions. Vaccine. 2014;32(32):
40824088.
18. Soheili M, Keyvani H, Soheili M, Nasseri S. Human papilloma virus:
a review study of epidemiology, carcinogenesis, diagnostic meth-
ods, and treatment of all HPVrelated cancers. Med J Islam Repub
Iran. 2021;35:65.
19. Maranga IO. HIV infection alters the spectrum of HPV subtypes
found in cervical smears and carcinomas from Kenyan women.
Open Virol J. 2013;7:1927.
20. Martins AE, LucenaSilva N, Garcia RG, et al. Prevalence of human
papillomavirus infection, distribution of viral types and risk factors
in cervical samples from human immunodeficiency viruspositive
women attending three human immunodeficiency virusacquired
immune deficiency syndrome reference centres in northeastern
Brazil. Mem Inst Oswaldo Cruz. 2014;109:738747.
21. Khodakarami N, Clifford GM, Yavari P, et al. Human papillomavirus
infection in women with and without cervical cancer in Tehran,
Iran. Int J Cancer. 2012;131(2):E156E161.
22. Heard I, Tondeur L, Arowas L, Falguières M, Demazoin MC,
Favre M. Human papillomavirus types distribution in organised
cervical cancer screening in France. PLoS One. 2013;8(11):
e79372.
23. Garland SM, Brotherton JM, Condon JR, et al. Human papilloma-
virus prevalence among indigenous and nonindigenous Australian
women prior to a national HPV vaccination program. BMC Med.
2011;9(1):104.
24. Cosper PF, Bradley S, Luo Q, Kimple RJ, eds. Biology of HPV
mediated carcinogenesis and tumor progression. Semin Radiat
Oncol. 2021;31(4):265273.
25. Doorbar J, Quint W, Banks L, et al. The biology and lifecycle of
human papillomaviruses. Vaccine. 2012;30:F55F70.
26. Hatano T, Sano D, Takahashi H, Oridate N. Pathogenic role of
immune evasion and integration of human papillomavirus in
oropharyngeal cancer. Microorganisms. 2021;9(5):891.
27. Reich O, Regauer S. Elimination of reserve cells for prevention of
HPVassociated cervical cancer. Virus Res. 2023;329:199068.
28. Fernandes JV, Galvão de Araújo JM, Allyrio Araújo de Medeiros
Fernandes T. Biology and natural history of human papillomavirus
infection. OpenAccessJClinTrials. 2013;2013:112.
29. McBride AA. Human papillomaviruses: diversity, infection and host
interactions. Nat Rev Microbiol. 2022;20(2):95108.
30. Best SR, Niparko KJ, Pai SI. Biology of human papillomavirus
infection and immune therapy for HPVrelated head and neck
cancers. Otolaryngol Clin North Am. 2012;45(4):807822.
31. McBride AA, Warburton A. The role of integration in oncogenic
progression of HPVassociated cancers. PLoS Pathog. 2017;13(4):
e1006211.
32. Isaacson Wechsler E, Wang Q, Roberts I, et al. Reconstruction of
human papillomavirus type 16mediated earlystage neoplasia
implicates E6/E7 deregulation and the loss of contact inhibition
in neoplastic progression. J Virol. 2012;86(11):63586364.
33. Steinbach A, Riemer AB. Immune evasion mechanisms of human
papillomavirus: an update. Int J Cancer. 2018;142(2):224229.
34. Ma Z, Ni G, Damania B. Innate sensing of DNA virus genomes.
Annu Rev Virol. 2018;5:341362.
AKBARI ET AL.
|
15 of 21
35. Lo Cigno I, Calati F, Albertini S, Gariglio M. Subversion of host
innate immunity by human papillomavirus oncoproteins. Pathogens.
2020;9(4):292.
36. Vandermark ER, Deluca KA, Gardner CR, et al. Human papilloma-
virus type 16 E6 and E 7 proteins alter NFkB in cultured cervical
epithelial cells and inhibition of NFkB promotes cell growth and
immortalization. Virology. 2012;425(1):5360.
37. Fernandes JV, De Medeiros Fernandes TAA, De Azevedo JCV,
et al. Link between chronic inflammation and human
papillomavirusinduced carcinogenesis. Oncol Lett. 2015;9(3):
10151026.
38. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMPAMP synthase is a
cytosolic DNA sensor that activates the type I interferon pathway.
Science. 2013;339(6121):786791.
39. Chen Q, Sun L, Chen ZJ. Regulation and function of the
cGASSTING pathway of cytosolic DNA sensing. Nat Immunol.
2016;17(10):11421149.
40. Hasan UA, Zannetti C, Parroche P, et al. The human papillomavirus
type 16 E7 oncoprotein induces a transcriptional repressor
complex on the Tolllike receptor 9 promoter. J Exp Med.
2013;210(7):13691387.
41. Buskwofie A, DavidWest G, Clare CA. A review of cervical cancer:
incidence and disparities. J Natl Med Assoc. 2020;112(2):229232.
42. Lechner M, Liu J, Masterson L, Fenton TR. HPVassociated
oropharyngeal cancer: epidemiology, molecular biology and clinical
management. Nat Rev Clin Oncol. 2022;19(5):306327.
43. Singh N, Gilks CB. Vulval squamous cell carcinoma and its
precursors. Histopathology. 2020;76(1):128138.
44. Thomas A, Necchi A, Muneer A, et al. Penile cancer. Nat Rev Dis
Primers. 2021;7(1):11.
45. Rajendra S, Pavey D, McKay O, Merrett N, Gautam SD. Human
papillomavirus infection in esophageal squamous cell carcinoma
and esophageal adenocarcinoma: a concise review. Ann NY Acad
Sci. 2020;1482(1):3648.
46. Rachel Skinner S, Wheeler CM, Romanowski B, et al. Progression of
HPV infection to detectable cervical lesions or clearance in adult
women: analysis of the control arm of the VIVIANE study. Int
J Cancer. 2016;138(10):24282438.
47. Rettig EM, D'Souza G. Epidemiology of head and neck cancer. Surg
Oncol Clin N Am. 2015;24(3):379396.
48. Tiwari PK, Kushwaha D, Kushwaha AK. Malignancies associated
with HPV. In: Khare P, Jain A, eds. Immunopathology, Diagnosis and
Treatment of HPV Induced Malignancies. Elsevier; 2022:4363.
49. Li X, Gao C, Yang Y, et al. Systematic review with metaanalysis:
the association between human papillomavirus infection and
oesophageal cancer. Aliment Pharmacol Ther. 2014;39(3):270281.
50. Zhang SK, Guo LW, Chen Q, et al. The association between human
papillomavirus 16 and esophageal cancer in Chinese population: a
metaanalysis. BMC Cancer. 2015;15(1):99.
51. Cho CY, Lo YC, Hung MC, Lai CC, Chen CJ, Wu KG. Risk of
cancer in patients with genital warts: a nationwide, population
based cohort study in Taiwan. PLoS One. 2017;12(8):e0183183.
52. Blomberg M, Friis S, Munk C, Bautz A, Kjaer SK. Genital warts and
risk of cancer: a Danish study of nearly 50 000 patients with genital
warts. J Infect Dis. 2012;205(10):15441553.
53. Marfatia Y, Dixit R, Bhavsar C. Laboratory diagnosis of human
papillomavirus virus infection in female genital tract. Indian J Sex
Transm Dis AIDS. 2011;32(1):50.
54. Fontham ETH, Wolf AMD, Church TR, et al. Cervical cancer screening
for individuals at average risk: 2020 guideline update from the
American Cancer Society. CA Cancer J Clin. 2020;70(5):321346.
55. Vives A, Cosentino M, Palou J. The role of human papilloma virus
test in men: first exhaustive review of literature. Actas Urol Esp
(Engl Ed). 2020;44(2):8693.
56. Centers for Disease Control and Prevention (US). [Last Reviewed:
April 18, 2022; cited 2023 Oct 10. Available from: https://www.
cdc.gov/std/hpv/stdfact-hpv-and-men.htm
57. Ranjeva SL, Baskerville EB, Dukic V, et al. Recurring infection with
ecologically distinct HPV types can explain high prevalence and
diversity. Proc Natl Acad Sci U S A. 2017;114(51):1357313578.
58. Williamson AL. Recent developments in human papillomavirus
(HPV) vaccinology. Viruses. 2023;15(7):1440.
59. Di Donato V, Caruso G, Bogani G, et al. HPV vaccination after
primary treatment of HPVrelated disease across different organ
sites: a multidisciplinary comprehensive review and metaanalysis.
Vaccines. 2022;10(2):239.
60. Saftlas AF, Spracklen CN, Ryckman KK, et al. Influence of a loop
electrosurgical excision procedure (LEEP) on levels of cytokines in
cervical secretions. J Reprod Immunol. 2015;109:7483.
61. Kechagias KS, Kalliala I, Bowden SJ, et al. Role of human
papillomavirus (HPV) vaccination on HPV infection and recurrence
of HPV related disease after local surgical treatment: systematic
review and metaanalysis. BMJ. 2022;378:e070135.
62. Kobayashi K, Hisamatsu K, Suzui N, Hara A, Tomita H, Miyazaki T.
A review of HPVrelated head and neck cancer. J Clin Med.
2018;7(9):241.
63. Khairkhah N, Bolhassani A, Najafipour R. Current and future
direction in treatment of HPVrelated cervical disease. J Mol Med.
2022;100(6):829845.
64. Lotfabadi P, Maleki F, Gholami A, Yazdanpanah MJ. Liquid nitrogen
cryotherapy versus 70% trichloroacetic acid in the treatment of
anogenital warts: a randomized controlled trial. Iran J Dermatol.
2015;18(4):151155.
65. Komericki P, AkkilicMaterna M, Strimitzer T, Aberer W. Efficacy
and safety of imiquimod versus podophyllotoxin in the treatment
of anogenital warts. Sex Transm Dis. 2011;38(3):216218.
66. Stockfleth E, Meyer T. Sinecatechins (Polyphenon E) ointment for
treatment of external genital warts and possible future indications.
Expert Opin Biol Ther. 2014;14(7):10331043.
67. Westfechtel L, Werner RN, Dressler C, Gaskins M, Nast A.
Adjuvant treatment of anogenital warts with systemic interferon:
a systematic review and metaanalysis. Sex Transm Infect.
2018;94(1):2129.
68. Cang W, Gu L, Hong Z, Wu A, Di W, Qiu L. Effectiveness of
photodynamic therapy with 5aminolevulinic acid on HPV clear-
ance in women without cervical lesions. Photodiagn Photodyn Ther.
2021;34:102293.
69. Desravines N, Hsu CH, Mohnot S, et al. Feasibility of 5fluorouracil
and imiquimod for the topical treatment of cervical intraepithelial
neoplasias (CIN) 2/3. Int J Gynaecol Obstet. 2023:16.
70. Ou YC, Fu HC, Tseng CW, Wu CH, Tsai CC, Lin H. The influence of
probiotics on genital highrisk human papilloma virus clearance and
quality of cervical smear: a randomized placebocontrolled trial.
BMC Womens Health. 2019;19:103.
71. Verhoeven V, Renard N, Makar A, et al. Probiotics enhance the
clearance of human papillomavirusrelated cervical lesions. Eur
J Cancer Prev. 2013;22(1):4651.
72. Mei Z, Li D. The role of probiotics in vaginal health. Front Cell Infect
Microbiol. 2022;12:963868.
73. Ashbolt NJ. Microbial contamination of drinking water and human
health from community water systems. Curr Environ Health Rep.
2015;2:95106.
74. Griffiths EC, Pedersen AB, Fenton A, Petchey OL. The nature and
consequences of coinfection in humans. J Infect. 2011;63(3):
200206.
75. Brooker S, Hotez PJ, Bundy DAP. The global Atlas of helminth
infection: mapping the way forward in neglected tropical disease
control. PLoS Neglected Trop Dis. 2010;4:e779.
16 of 21
|
AKBARI ET AL.
76. Mulherkar TH, Gómez DJ, Sandel G, Jain P. Coinfection and
cancer: hostpathogen interaction between dendritic cells and
HIV1, HTLV1, and other oncogenic viruses. Viruses. 2022;14(9):
2037.
77. Sternberg ED, Lefèvre T, Rawstern AH, de Roode JC. A virulent
parasite can provide protection against a lethal parasitoid. Infect
Genet Evol. 2011;11(2):399406.
78. PérezGonzález A, Cachay E, Ocampo A, Poveda E. Update on the
epidemiological features and clinical implications of human
papillomavirus infection (HPV) and human immunodeficiency virus
(HIV) coinfection. Microorganisms. 2022;10(5):1047.
79. McKeon M, Gallant JN, Kim Y, Das S. It takes two to
tango: a review of oncogenic virus and host microbiome associated
inflammation in head and neck cancer. Cancers. 2022;14(13):3120.
80. Malik S, Sah R, Muhammad K, Waheed Y. Tracking HPV infection,
associated cancer development, and recent treatment effortsa
comprehensive review. Vaccines. 2023;11(1):102.
81. Koelle DM, Norberg P, Fitzgibbon MP, et al. Worldwide circulation
of HSV HSV1 recombinant strains. Sci Rep. 2017;7(1):44084.
82. Ghosh S, Shetty RS, Pattanshetty SM, et al. Human papilloma and
other DNA virus infections of the cervix: a population based
comparative study among tribal and general population in India.
PLoS ONE. 2019;14(6):e0219173.
83. Sosse SA, Tadlaoui KA, Benhassou M, Elkarroumi M, Elmzibri M,
Ennaji MM. Viral coinfection of oncogenic human papillomavirus
with EpsteinBarr virus, human herpesvirus 8 and Herpes Simplex
Virus type 2 in malignant cervical cancer. Int Med J. 2022;3(2):115.
84. Erira AT, Navarro AFR, Robayo DAG. Human papillomavirus,
EpsteinBarr virus, and Candida albicans coinfection in oral
leukoplakia with different degrees of dysplasia. Clin Exp Dent Res.
2021;7(5):914923.
85. Castellsagué X, Bosch FX, Muñoz N. Environmental cofactors in
HPV carcinogenesis. Virus Res. 2002;89(2):191199.
86. Tavakolian S, Goudarzi H, Nazarian H, Raee P, Niakan S,
Faghihloo E. The evaluation of human papilloma virus and human
herpes viruses (EBV, CMV, VZV HSV1 and HSV2) in semen
samples. Andrologia. 2021;53(6):e14051.
87. Mosmann JP, Talavera AD, Criscuolo MI, et al. Sexually transmitted
infections in oral cavity lesions: Human papillomavirus,Chlamydia
trachomatis, and Herpes simplex virus.J Oral Microbiol.
2019;11(1):1632129.
88. Zhao Y, Cao X, Zheng Y, et al. Relationship between cervical
disease and infection with human papillomavirus types 16 and 18,
and herpes simplex virus 1 and 2. J Med Virol. 2012;84(12):
19201927.
89. BahenaRomán M, SánchezAlemán MA, ContrerasOchoa CO,
et al. Prevalence of active infection by herpes simplex virus type 2
in patients with highrisk human papillomavirus infection: a cross
sectional study. J Med Virol. 2020;92(8):12461252.
90. Chuerduangphui J, Proyrungroj K, Pientong C, et al. Prevalence and
anatomical sites of human papillomavirus, EpsteinBarr virus and
herpes simplex virus infections in men who have sex with men,
Khon Kaen, Thailand. BMC Infect Dis. 2018;18(1):509.
91. Keller MJ, Huber A, Espinoza L, et al. Impact of herpes simplex
virus type 2 and human immunodeficiency virus dual infection on
female genital tract mucosal immunity and the vaginal microbiome.
J Infect Dis. 2019;220(5):852861.
92. Li S, Wen X. Seropositivity to herpes simplex virus type 2, but not
type 1 is associated with cervical cancer: NHANES (19992014).
BMC Cancer. 2017;17(1):726.
93. Turunen A, Hukkanen V, Nygårdas M, Kulmala J, Syrjänen S. The
combined effects of irradiation and herpes simplex virus type 1
infection on an immortal gingival cell line. Virol J. 2014;
11(1):125.
94. Mirzaei H, Khodadad N, Karami C, Pirmoradi R, Khanizadeh S. The
AP1 pathway; A key regulator of cellular transformation modu-
lated by oncogenic viruses. Rev Med Virol. 2020;30(1):e2088.
95. Westhoff Smith D, Chakravorty A, Hayes M, Hammerschmidt W,
Sugden B. The EpsteinBarr virus oncogene EBNA1 suppresses
natural killer cell responses and apoptosis early after infection of
peripheral B cells. mBio. 2021;12(6):e0224321.
96. Rahman R, Shaikh MH, Gopinath D, Idris A, Johnson NW. Human
papillomavirus and EpsteinBarr virus coinfection in oral and
oropharyngeal squamous cell carcinomas: a systematic review and
metaanalysis. Mol Oral Microbiol. 2023;38(4):259274.
97. Kato I, Zhang J, Sun J. Bacterialviral interactions in human
orodigestive and female genital tract cancers: a summary of
epidemiologic and laboratory evidence. Cancers. 2022;14(2):425.
98. Blanco R, CarrilloBeltrán D, Corvalán AH, Aguayo F. Highrisk
human papillomavirus and EpsteinBarr virus coinfection: a poten-
tial role in head and neck carcinogenesis. Biology. 2021;10(12):
1232.
99. Guidry J, Birdwell C, Scott R. EpsteinBarr virus in the patho-
genesis of oral cancers. Oral Dis. 2018;24(4):497508.
100. Gianella S, Ginocchio CC, Daar ES, Dube MP, Morris SR. Genital
Epstein Barr Virus is associated with higher prevalence and
persistence of anal human papillomavirus in HIVinfected men on
antiretroviral therapy. BMC Infect Dis. 2015;16(1):24.
101. Guimarães AGDP, Araujo JR, Andrade RV, et al. Coinfection of
EpsteinBarr virus, cytomegalovirus, herpes simplex virus, human
papillomavirus and anal intraepithelial neoplasia in HIV patients in
Amazon, Brazil. J. Coloproctol. (Rio J). 2012;32:1825.
102. Afonso LA, Moyses N, Alves G, et al. Prevalence of human
papillomavirus and EpsteinBarr virus DNA in penile cancer cases
from Brazil. Mem Inst Oswaldo Cruz. 2012;107:1823.
103. Kahla S, Oueslati S, Achour M, et al. Correlation between ebv co
infection and HPV16 genome integrity in Tunisian cervical cancer
patients. Braz J Microbiol. 2012;43:744753.
104. Blanco R, CarrilloBeltrán D, Osorio JC, Calaf GM, Aguayo F. Role
of EpsteinBarr virus and human papillomavirus coinfection in
cervical cancer: epidemiology, mechanisms and perspectives.
Pathogens. 2020;9(9):685.
105. de Lima MAP, Neto PJN, Lima LPM, et al. Association between
EpsteinBarr virus (EBV) and cervical carcinoma: a metaanalysis.
Gynecol Oncol. 2018;148(2):317328.
106. Shi Y, Peng SL, Yang LF, Chen X, Tao YG, Cao Y. Coinfection of
EpsteinBarr virus and human papillomavirus in human tumori-
genesis. Chin J Cancer. 2016;35(1):16.
107. Van Sciver N. The Regulation of EBV Lytic Reactivation in Epithelial
Cells. The University of WisconsinMadison; 2021:1206.
108. Skinner CM, Ivanov NS, Barr SA, Chen Y, Skalsky RL. An Epstein
Barr virus microRNA blocks interleukin1 (IL1) signaling by
targeting IL1 receptor 1. J Virol. 2017;91(21):117.
109. StrycharzDudziak M, Fołtyn S, Dworzański J, et al. Glutathione
peroxidase (GPx) and superoxide dismutase (SOD) in oro-
pharyngeal cancer associated with EBV and HPV coinfection.
Viruses. 2020;12(9):1008.
110. Nahand JS, Khanaliha K, Mirzaei H, et al. Possible role of HPV/EBV
coinfection in anoikis resistance and development in prostate
cancer. BMC Cancer. 2021;21(1):926.
111. AlThawadi H, Ghabreau L, Aboulkassim T, et al. Coincidence of
epsteinBarr Virus and highrisk human papillomaviruses in cervical
cancer of Syrian women. Front Oncol. 2018;8:250.
112. Wiech T, Nikolopoulos E, Lassman S, et al. Cyclin D1 expression is
induced by viral BARF1 and is overexpressed in EBVassociated
gastric cancer. Virchows Arch. 2008;452:621627.
113. Feng M, Duan R, Gao Y, et al. Role of EpsteinBarr virus and human
papillomavirus coinfection in cervical intraepithelial neoplasia in
AKBARI ET AL.
|
17 of 21
Chinese women living with HIV. Front Cell Infect Microbiol.
2021;11:703259.
114. Shim AHR, Chang RA, Chen X, Longnecker R, He X. Multipronged
attenuation of macrophagecolony stimulating factor signaling by
EpsteinBarr virus BARF1. Proc Natl Acad Sci U S A. 2012;109(32):
1296212967.
115. Uehara K, Tanabe Y, Hirota S, et al. Coexpression of lowrisk HPV
E6/E7 and EBV LMP1 leads to precancerous lesions by DNA
damage. BMC Cancer. 2021;21(1):688.
116. SalimiJeda A, Badrzadeh F, Esghaei M, Abdoli A. The role of
telomerase and viruses interaction in cancer development, and
telomerasedependent therapeutic approaches. Cancer Treat Res
Commun. 2021;27:100323.
117. Al Moustafa AE, Chen D, Ghabreau L, Akil N. Association between
human papillomavirus and EpsteinBarr virus infections in human
oral carcinogenesis. Med Hypotheses. 2009;73(2):184186.
118. Shimabuku T, Tamanaha A, Kitamura B, et al. Dual expression of
EpsteinBarr virus, latent membrane protein1 and human
papillomavirus16 E6 transform primary mouse embryonic fibroblasts
through NFκB signaling. Int J Clin Exp Pathol. 2014;7(5):19201934.
119. Islam MR, Nowshin DT, Khan MR, Shahriar M, Bhuiyan MA.
Monkeypox and sex: sexual orientations and encounters are key
factors to consider. Health Sci Rep. 2023;6(1):e1069.
120. Perez R, Gibson S, Lopez P, Koenig E, De Castro M, Yamamura Y.
Distribution of HIV1 infection in different T lymphocyte subsets:
antiretroviral therapyNaïve vs. experienced patients. AIDS Res
Hum Retroviruses. 2011;27(4):399410.
121. Doitsh G, Galloway NLK, Geng X, et al. Cell death by pyroptosis
drives CD4 Tcell depletion in HIV1 infection. Nature.
2014;505(7484):509514.
122. Houlihan CF, Larke NL, WatsonJones D, et al. HPV infection and
increased risk of HIV acquisition. A systematic review and meta
analysis. AIDS. 2012;26(17). doi:10.1097/QAD.0b013e328
358d908
123. Clarke B. Postmodern cancer: the role of human immunodeficiency
virus in uterine cervical cancer. Mol Pathol. 2002;55(1):1924.
124. Nicol AF, Pires ARC, de Souza SR, et al. Cellcycle and suppressor
proteins expression in uterine cervix in HIV/HPV coinfection:
comparative study by tissue microarray (TMA). BMC Cancer.
2008;8(1):289.
125. Nyagol J, Leucci E, Onnis A, et al. The effects of HIV1 Tat protein
on cell cycle during cervical carcinogenesis. Cancer Biol Ther.
2006;5(6):684690.
126. Barillari G, Palladino C, Bacigalupo I, Leone P, Falchi M, Ensoli B.
Entrance of the Tat protein of HIV1 into human uterine cervical
carcinoma cells causes upregulation of HPVE6 expression and a
decrease in p53 protein levels. Oncol Lett. 2016;12(4):23892394.
127. Dolei A, Curreli S, Marongiu P, et al. Human immunodeficiency
virus infection in vitro activates naturally integrated human
papillomavirus type 18 and induces synthesis of the L1 capsid
protein. J Gen Virol. 1999;80(11):29372944.
128. Vernon SD, Hart CE, Reeves WC, Icenogle JP. The HIV1 tat
protein enhances E2dependent human papillomavirus 16 tran-
scription. Virus Res. 1993;27(2):133145.
129. Toy EP, RodríguezRodríguez L, McCance D, Ludlow J, Planelles V.
Induction of cellcycle arrest in cervical cancer cells by the human
immunodeficiency virus type 1 viral protein R. Obstet Gynecol.
2000;95(1):141146.
130. Luque AE, Demeter LM, Reichman RC. Association of human
papillomavirus infection and disease with magnitude of human
immunodeficiency virus type 1 (HIV1) RNA plasma level among
women with HIV1 infection. J Infect Dis. 1999;179(6):14051409.
131. Nicol AF, Nuovo GJ, SalomaoEstevez A, et al. Immune factors
involved in the cervical immune response in the HIV/HPV co
infection. J Clin Pathol. 2007;61(1):8488.
132. Fitzgerald DW, Bezak K, Ocheretina O, et al. The effect of HIV and
HPV coinfection on cervical COX2 expression and systemic
prostaglandin E2 levels. Cancer Prev Res. 2012;5(1):3440.
133. Ao C, Zeng K. The role of regulatory T cells in pathogenesis and
therapy of human papillomavirusrelated diseases, especially in
cancer. Infect Genet Evol. 2018;65:406413.
134. Osborn L, Kunkel S, Nabel GJ. Tumor necrosis factor alpha and
interleukin 1 stimulate the human immunodeficiency virus en-
hancer by activation of the nuclear factor kappa B. Proc Natl Acad
Sci USA. 1989;86(7):23362340.
135. Behbakht K, Friedman J, Heimler I, Aroutcheva A, Simoes J, Faro S.
Role of the vaginal microbiological ecosystem and cytokine profile
in the promotion of cervical dysplasia: a casecontrol study. Infect
Dis Obstet Gynecol. 2002;10:181186.
136. Badial RM, Dias MC, Stuqui B, et al. Detection and genotyping of
human papillomavirus (HPV) in HIVinfected women and its relationship
with HPV/HIV coinfection. Medicine. 2018;97(14):e9545.
137. Lehtovirta P, Paavonen J, Heikinheimo O. Risk factors, diagnosis
and prognosis of cervical intraepithelial neoplasia among HIV
infected women. Int J STD AIDS. 2008;19(1):3741.
138. Okoye JO, Erinle C, Ngokere AA, Jimoh A. Low CD4 cells and viral
coinfection increase the risk of VaIN: use of SCCA1 and ki67 as
diagnoprognostic biomarkers. Pathophysiology. 2018;25(1):5156.
139. Ferenczy A, Coutlée F, Franco E, Hankins C. Human papillomavirus and
HIV coinfection and the risk of neoplasias of the lower genital tract: a
review of recent developments. CMAJ. 2003;169(5):431434.
140. Duan R, Zhang H, Wu A, et al. Prevalence and risk factors for
anogenital HPV infection and neoplasia among women living with
HIV in China. Sex Transm Infect. 2022;98(4):247254.
141. HidalgoTenorio C, GilAnguita C, RamírezTaboada J, et al. Risk
factors for infection by oncogenic human papillomaviruses in HIV
positive MSM patients in the ART era (20102016). Medicine.
2017;96(39):e8109.
142. Grinsztejn B, Veloso VG, Levi JE, et al. Factors associated with
increased prevalence of human papillomavirus infection in a cohort
of HIVinfected Brazilian women. Int J Infect Dis. 2009;13(1):7280.
143. Camargo M, Del RíoOspina L, SotoDe León SC, et al. Association
of HIV status with infection by multiple HPV types. Trop Med Int
Health. 2018;23(11):12591268.
144. Mbulawa ZZ, Johnson LF, Marais DJ, et al. Increased alpha9
human papillomavirus species viral load in human immuno-
deficiency virus positive women. BMC Infect Dis. 2014;14:517.
145. Kelly H, Weiss HA, Benavente Y, et al. Association of antiretroviral
therapy with highrisk human papillomavirus, cervical intraepithe-
lial neoplasia, and invasive cervical cancer in women living with
HIV: a systematic review and metaanalysis. Lancet HIV.
2018;5(1):e45e58.
146. Ermel A, Tong Y, Tonui P, et al. Longer duration of antiretroviral
therapy is associated with decreased risk of human papilloma-
viruses detection in Kenyan women living with HIV. Int J STD AIDS.
2021;32(13):12121220.
147. Liu G, Sharma M, Tan N, Barnabas RV. HIVpositive women have
higher risk of human papilloma virus infection, precancerous
lesions, and cervical cancer. AIDS. 2018;32(6):795808.
148. Clifford GM, Franceschi S, Keiser O, et al. Immunodeficiency and
the risk of cervical intraepithelial neoplasia 2/3 and cervical cancer:
a nested casecontrol study in the Swiss HIV cohort study. Int
J Cancer. 2016;138(7):17321740.
149. HidalgoTenorio C, RiveroRodriguez M, GilAnguita C, et al.
Antiretroviral therapy as a factor protective against anal dysplasia
in HIVinfected males who have sex with males. PLoS ONE.
2014;9(3):e92376.
150. Franceschi S, Lise M, Clifford GM, et al. Changing patterns of
cancer incidence in the earlyand lateHAART periods: the Swiss
HIV cohort study. Br J Cancer. 2010;103(3):416422.
18 of 21
|
AKBARI ET AL.
151. Papasavvas E, Surrey LF, Glencross DK, et al. Highrisk oncogenic
HPV genotype infection associates with increased immune activa-
tion and T cell exhaustion in ARTsuppressed HIV1infected
women. Oncoimmunology. 2016;5(5):e1128612.
152. Zurek MunkMadsen M, Toft L, Kube T, et al. Cellular immunoge-
nicity of human papillomavirus vaccines cervarix and gardasil in
adults with HIV infection. Hum Vaccines Immunother. 2018;14(4):
909916.
153. Toft L, Storgaard M, Muller M, et al. Comparison of the
immunogenicity and reactogenicity of cervarix and gardasil human
papillomavirus vaccines in HIVinfected adults: a randomized,
doubleblind clinical trial. J Infect Dis. 2014;209(8):11651173.
154. Dreyer G. Clinical implications of the interaction between HPV and
HIV infections. Best Pract Res Clin Obstet Gynaecol. 2018;47:
95106.
155. De Vuyst H, Alemany L, Lacey C, et al. The burden of human
papillomavirus infections and related diseases in subSaharan
Africa. Vaccine. 2013;31:F32F46.
156. Menon S, Wusiman A, Boily MC, et al. Epidemiology of HPV
genotypes among HIV positive women in Kenya: a systematic
review and metaanalysis. PLoS One. 2016;11(10):e0163965.
157. Denny LA, Franceschi S, de Sanjosé S, Heard I, Moscicki AB,
Palefsky J. Human papillomavirus, human immunodeficiency virus
and immunosuppression. Vaccine. 2012;30:F168F174.
158. Clifford GM, de Vuyst H, Tenet V, Plummer M, Tully S,
Franceschi S. Effect of HIV infection on human papillomavirus
types causing invasive cervical cancer in Africa. J Acquir Immune
Defic Syndr. 2016;73(3):332339.
159. Denny L, Adewole I, Anorlu R, et al. Human papillomavirus
prevalence and type distribution in invasive cervical cancer in
subSaharan Africa. Int J Cancer. 2014;134(6):13891398.
160. Blitz S, Baxter J, Raboud J, et al. Evaluation of HIV and highly active
antiretroviral therapy on the natural history of human papillomavirus
infection and cervical cytopathologic findings in HIVpositive and high
risk HIVnegative women. JInfectDis. 2013;208(3):454462.
161. Van Aardt MC, Dreyer G, Pienaar HF, et al. Unique human
papillomavirustype distribution in South African women with
invasive cervical cancer and the effect of human immunodeficiency
virus infection. Int J Gynecol Cancer. 2015;25(5):919925.
162. Wang Q, Ma X, Zhang X, et al. Human papillomavirus infection and
associated factors for cervical intraepithelial neoplasia in women
living with HIV in China: a crosssectional study. Sex Transm Infect.
2019;95(2):140144.
163. Massad LS, Xie X, Darragh T, et al. Genital warts and vulvar
intraepithelial neoplasia: natural history and effects of treatment
and human immunodeficiency virus infection. Obstet Gynecol.
2011;118(4):831839.
164. Pernot S, Terme M, Zaanan A, Tartour E, Weiss L, Taieb J.
Immunity and squamous cell carcinoma of the anus: epidemiolo-
gical, clinical and therapeutic aspects. Clin Res Hepatol
Gastroenterol. 2014;38(1):1823.
165. Lekoane KMB, Kuupiel D, MashambaThompson TP, Ginindza TG.
The interplay of HIV and human papillomavirusrelated cancers in
subSaharan Africa: scoping review. Syst Rev. 2020;9(1):88.
166. Denny L, Boa R, Williamson AL, et al. Human papillomavirus
infection and cervical disease in human immunodeficiency Virus
1infected women. Obstet Gynecol. 2008;111(6):13801387.
167. Palefsky J. Human papillomavirusrelated tumors in HIV. Curr Opin
Oncol. 2006;18(5):463468.
168. Gilles C, Manigart Y, Konopnicki D, Barlow P, Rozenberg S.
Management and outcome of cervical intraepithelial neoplasia
lesions: a study of matched cases according to HIV status. Gynecol
Oncol. 2005;96(1):112118.
169. Heard I. Human papillomavirus, cancer and vaccination. Curr Opin
HIV AIDS. 2011;6(4):297302.
170. JedyAgba EE, Dareng EO, Adebamowo SN, et al. The burden of
HPV associated cancers in two regions in Nigeria 20122014.
Cancer Epidemiol. 2016;45:9197.
171. Szostek S, Zawilinska B, Kopec J, KoszVnenchak M. Herpesviruses
as possible cofactors in HPV16related oncogenesis. Acta Biochim
Pol. 2009;56(2):337342.
172. McGalie CE. Cytomegalovirus infection of the cervix: morphologi-
cal observations in five cases of a possibly underrecognised
condition. J Clin Pathol. 2004;57(7):691694.
173. Broccolo F, Cassina G, Chiari S, et al. Frequency and clinical
significance of human βherpesviruses in cervical samples from
Italian women. J Med Virol. 2008;80(1):147153.
174. Jalilvand S, Shoja Z, MokhtariAzad T, Nategh R, Gharehbaghian A.
Seroprevalence of human herpesvirus 8 (HHV8) and incidence of
Kaposi's sarcoma in Iran. Infect Agent Cancer. 2011;6(1):5.
175. ChavoshpourMamaghani S, Shoja Z, MollaeiKandelous Y,
Sharifian K, Jalilvand S. The prevalence of human herpesvirus 8
in normal, premalignant, and malignant cervical samples of Iranian
women. Virol J. 2021;18(1):144.
176. Dai L, Zhao M, Jiang W, Lin Z, Del Valle L, Qin Z. KSHV co
infection, a new cofactor for HPVrelated cervical carcinogenesis?
Am J Cancer Res. 2018;8(11):21762184.
177. Saber Amoli S, Hasanzadeh A, Sadeghi F, et al. Prevalence of co
infection by human papillomavirus, EpsteinBarr virus and merkel
cell polyomavirus in Iranian oral cavity cancer and premalignant
lesions. Int J Mol Cell Med. 2022;11(1):6477.
178. Falchook GS, Rady P, Hymes S, et al. Merkel cell polyomavirus and
HPV17 associated with cutaneous squamous cell carcinoma
arising in a patient with melanoma treated with the BRAF inhibitor
dabrafenib. JAMA Dermatol. 2013;149(3):322326.
179. Comar M, Bonifacio D, Zanconati F, et al. High prevalence of BK
polyomavirus sequences in human papillomavirus16positive
precancerous cervical lesions. JMedVirol. 2011;83(10):
17701776.
180. Alosaimi B, Hampson L, He X, Maranga IO, Oliver AW,
Hampson IN. Increased prevalence of JC polyomavirus in cervical
carcinomas from women infected with HIV. J Med Virol.
2014;86(4):672677.
181. Rosadas C, Taylor GP. HTLV1 and coinfections. Front Med.
2022;9:812016.
182. Lôpo SS, Oliveira PM, Santana IU, et al. Evidence of a higher
prevalence of HPV infection in HTLV1infected women: a cross
sectional study. Rev Soc Bras Med Trop. 2012;45:305308.
183. Blas MM, Alva IE, Garcia PJ, et al. Association between human
papillomavirus and human Tlymphotropic virus in indigenous
women from the Peruvian Amazon 2012;7(8):16.
184. Soares CC, Georg I, La mpe E, et al. HIV1, HBV, HCV, HTLV,
HPV16/18, and Treponema pallidum infections in a sample of
Brazilian men who have sex with men. PLoS One. 2014;
9(8):e102676.
185. LiuJ,LiuW,LiuY,ZhouX,ZhangZ,SunZ.Prevalenceof
microorganisms coinfections in human papillomaviruses infected
womeninNorthernChina.Arch Gynecol Obstet. 2016;293:595602.
186. Saláková M, Němeček V, Tachezy R. TTV and HPV coinfection in
cervical smears of patients with cervical lesions. BMC Infect Dis.
2009;9(1):118.
187. Siahpoush M, Noorbazargan H, Kalantari S, Shayestehpour M,
Yazdani S. Coinfection of torque teno virus (TTV) and human
papillomavirus (HPV) in cervical samples of women living in Tehran,
Iran. Iran J Microbiol. 2022;14(2):181.
188. Lin D, Kouzy R, Abi Jaoude J, et al. Microbiome factors in HPV
driven carcinogenesis and cancers. PLoS Pathog. 2020;16(6):
e1008524.
189. Cullin N, Azevedo Antunes C, Straussman R, SteinThoeringer CK,
Elinav E. Microbiome and cancer. Cancer Cell. 2021;39(10):13171341.
AKBARI ET AL.
|
19 of 21
190. Gholiof M, AdamsonDe luca E, Wessels JM. The female
reproductive tract microbiotas, inflammation, and gynecological
conditions. Front Reprod Health. 2022;4:124.
191. Hamar B, Teutsch B, Hoffmann E, et al. Trichomonas vaginalis
infection is associated with increased risk of cervical carcinogene-
sis: a systematic review and metaanalysis of 470 000 patients. Int
J Gynaecol Obstet. 2023;163:113.
192. Zhou ZW, Long HZ, Cheng Y, Luo HY, Wen DD, Gao LC. From
microbiome to inflammation: the key drivers of cervical cancer.
Front Microbiol. 2021;12:767931.
193. Klein C, Kahesa C, Mwaiselage J, West JT, Wood C, Angeletti PC.
How the cervical microbiota contributes to cervical cancer risk in
subSaharan Africa. Front Cell Infect Microbiol. 2020;10:23.
194. Kumari S, Bhor VM. Association of cervicovaginal dysbiosis
mediated HPV infection with cervical intraepithelial neoplasia.
Microb Pathog. 2021;152:104780.
195. Lv P, Zhao F, Xu X, Xu J, Wang Q, Zhao Z. Correlation between
common lower genital tract microbes and highrisk human papilloma-
virus infection. Can J Infect Dis Med Microbiol. 2019;2019:16.
196. Ye H, Song T, Zeng X, Li L, Hou M, Xi M. Association between genital
mycoplasmas infection and human papillomavirus infection, abnormal
cervical cytopathology, and cervical cancer: a systematic review and
metaanalysis. Arch Gynecol Obstet. 2018;297:13771387.
197. Tamarelle J, Thiébaut ACM, De Barbeyrac B, Bébéar C, Ravel J,
DelarocqueAstagneau E. The vaginal microbiota and its associa-
tion with Human papillomavirus,Chlamydia trachomatis,Neisseria
gonorrhoeae and Mycoplasma genitalium infections: a systematic
review and metaanalysis. Clin Microbiol Infect. 2019;25(1):3547.
198. Ekiel AM, Friedek DA, Romanik MK, źwiak J, Martirosian G.
Occurrence of Ureaplasma parvum and Ureaplasma urealyticum in
women with cervical dysplasia in Katowice, Poland. J Korean Med
Sci. 2009;24(6):11771181.
199. McGowin CL, Popov VL, Pyles RB. Intracellular Mycoplasma
genitalium infection of human vaginal and cervical epithelial cells
elicits distinct patterns of inflammatory cytokine secretion and
provides a possible survival niche against macrophagemediated
killing. BMC Microbiol. 2009;9:13911.
200. Kaliterna V, Kaliterna P, Pejkovic L, Vulic R, Zanchi L, Cerskov K.
Prevalence of human papillomavirus (HPV) among females in the
general population of the split and dalmatia county and association
with genital microbiota and infections: a prospective study. Viruses.
2023;15(2):443.
201. Mortaki D, Gkegkes ID, Psomiadou V, et al. Vaginal microbiota and
human papillomavirus: a systematic review. J Turk Ger Gynecol
Assoc. 2020;21(3):193200.
202. So KA, Yang EJ, Kim NR, et al. Changes of vaginal microbiota during
cervical carcinogenesis in women with human papillomavirus
infection. PLoS ONE. 2020;15(9):e0238705.
203. Ağar E, Aker SS. Association of HPV and sexually transmitted
infections among patients with genital warts and asymptomatic
individuals: a crosssectional study. Eur J Gynaecol Oncol. 2023;1:11.
204. Wei ZT, Chen HL, Wang CF, Yang GL, Han SM, Zhang SL.
Depiction of vaginal microbiota in women with highrisk human
papillomavirus infection. Front Public Health. 2021;8:587298.
205. Adebamowo SN, Ma B, Zella D, et al. Mycoplasma hominis and
Mycoplasma genitalium in the vaginal microbiota and persistent
highrisk human papillomavirus infection. Front Public Health.
2017;5:140.
206. Verteramo R, Pierangeli A, Mancini E, et al. Human papilloma-
viruses and genital coinfections in gynaecological outpatients.
BMC Infect Dis. 2009;9(1):16.
207. Kim SI, Yoon JH, Park DC, et al. Coinfection of Ureaplasma ur-
ealyticum and human papilloma virus in asymptomatic sexually
active individuals. Int J Med Sci. 2018;15(9):915920.
208. Adamopoulou M, Avgoustidis D, Voyiatjaki C, et al. Impact of
combined mycoplasmataceae and HPV coinfection on females
with cervical intraepithelial neoplasia and carcinoma. Off J Balk
Union. Oncol. 2021;1313:261319.
209. Kim HS, Kim TJ, Lee IH, Hong SR. Associations between sexually
transmitted infections, highrisk human papillomavirus infection,
and abnormal cervical Pap smear results in OB/GYN outpatients.
J Gynecol Oncol. 2016;27(5):111.
210. Bi H, Zhang D, Xiao B. Association between human papillomavirus
infection and common sexually transmitted infections, and the
clinical significance of different Mycoplasma subtypes. Frontiers in
cellular and infection. Microbiology. 2023;13:238.
211. Plisko O, Zodzika J, Jermakova I, et al. Aerobic vaginitis
underestimated risk factor for cervical intraepithelial neoplasia.
Diagnostics. 2021;11(1):97.
212. FazlollahpourNaghibi A, Bagheri K, Almukhtar M, et al. Tricho-
monas vaginalis infection and risk of cervical neoplasia: a systematic
review and metaanalysis. PLoS One. 2023;18(7):e0288443.
213. Ssedyabane F, Amnia DA, Mayanja R, et al. HPVchlamydial
coinfection, prevalence, and association with cervical intraepithelial
lesions: a pilot study at Mbarara regional referral hospital. J Cancer
Epidemiol. 2019;2019:17.
214. Koskela P, Anttila T, Bjørge T, et al. Chlamydia trachomatis
infection as a risk factor for invasive cervical cancer. Int J Cancer.
2000;85(1):3539.
215. Cancer IAfRo. IARC Working Group on the Evaluation of Carcinogenic
Risk to Humans. Biological Agents. Lyon (FR): International Agency for
Research on Cancer; 2012. IARC Monographs on the Evaluation of
Carcinogenic Risks to Humans. 100, 2012:1441.
216. Klein C, Samwel K, Kahesa C, et al. Mycoplasma coinfection is
associated with cervical cancer risk. Cancers. 2020;12(5):1093.
217. Yin YP, Li HM, Xiang Z, et al. Association of sexually transmitted
infections with highrisk human papillomavirus types. Sex Transm
Dis. 2013;40(6):493495.
218. McGowin CL, Radtke AL, Abraham K, Martin DH, HerbstKralovetz
M. Mycoplasma genitalium infection activates cellular host defense
and inflammation pathways in a 3dimensional human endocervical
epithelial cell model. J Infect Dis. 2013;207(12):18571868.
219. Morgan EL, Scarth JA, Patterson MR, et al. E6mediated activation
of JNK drives EGFR signalling to promote proliferation and viral
oncoprotein expression in cervical cancer. Cell Death Differ.
2021;28(5):16691687.
220. Zhang Z, Li D, Li Y, et al. The correlation between Trichomonas
vaginalis infection and reproductive system cancer: a systematic
review and metaanalysis. Infect Agent Cancer. 2023;18(1):15.
221. Wilson JS. A systematic review of the prevalence of Chlamydia
trachomatis among European women. Hum Reprod Update.
2002;8(4):385394.
222. FernándezBenítez C, MejutoLópez P, OteroGuerra L, Margolles
Martins MJ, SuárezLeiva P, Vazquez F. Prevalence of genital
Chlamydia trachomatis infection among young men and women in
Spain. BMC Infect Dis. 2013;13(1):388.
223. Ruelle J, Debaisieux L, Vancutsem E, et al. HIV1 lowlevel viraemia
assessed with 3 commercial realtime PCR assays show high
variability. BMC Infect Dis. 2012;12(1):100.
224. Seraceni S, De Seta F, Colli C, et al. High prevalence of hpv multiple
genotypes in women with persistent chlamydia trachomatis
infection. Infect Agent Cancer. 2014;9(1):30.
225. Silva J, Cerqueira F, Ribeiro J, Sousa H, Osório T, Medeiros R. Is
Chlamydia trachomatis related to human papillomavirus infection in
young women of southern European population? A selfsampling
study. Arch Gynecol Obstet. 2013;288:627633.
226. Kumari S, Bhor VM. A literature review on correlation between
HPV coinfection with C. trachomatis and cervical neoplasia
20 of 21
|
AKBARI ET AL.
coinfection mediated cellular transformation. Microb Pathog.
2022;168:105587.
227. Ferrera L, Rogua H, El Mansouri N, et al. The association of
Chlamydia trachomatis and human papillomavirus coinfection with
abnormal cervical cytology among women in south of Morocco.
Microb Pathog. 2023;175:105971.
228. Gargiulo Isacco C, Balzanelli MG, Garzone S, et al. Alterations of
vaginal microbiota and Chlamydia trachomatis as crucial co
Causative factors in cervical cancer genesis procured by HPV.
Microorganisms. 2023;11(3):662.
229. Syrjänen K, Väyrynen M, Saarikoski S, et al. Natural history of
cervical human papillomavirus (HPV) infections based on prospec-
tive followup. Br J Obstet Gynaecol. 1985;92(11):10861092.
230. Suehiro TT, Gimenes F, Souza RP, et al. High molecular prevalence
of HPV and other sexually transmitted infections in a population of
asymptomatic women who work or study at a Brazilian university.
Rev Inst Med Trop Sao Paulo. 2021;63:110.
231. Mosmann JP, Zayas S, Kiguen AX, Venezuela RF, Rosato O,
Cuffini CG. Human papillomavirus and Chlamydia trachomatis in
oral and genital mucosa of women with normal and abnormal
cervical cytology. BMC Infect Dis. 2021;21(1):422.
232. Sangpichai S, Patarapadungkit N, Pientong C, et al. Chlamydia
trachomatis infection in highrisk human papillomavirus based on
cervical cytology specimen. Asian Pac J Cancer Prev. 2019;20(12):
38433847.
233. Chumduri C, Gurumurthy RK, Zadora PK, Mi Y, Meyer TF.
Chlamydia infection promotes host DNA damage and proliferation
but impairs the DNA damage response. Cell Host Microbe.
2013;13(6):746758.
234. Challagundla N, ChrisopheBourdon J, AgrawalRajput R. Chla-
mydia trachomatis infection cooperatively enhances HPV E6E7
oncogenes mediated tumorigenesis and immunosuppression.
Microb Pathog. 2023;175:105929.
235. Yang X, Siddique A, Khan AA, et al. Chlamydia trachomatis
infection: their potential implication in the etiology of cervical
cancer. J Cancer. 2021;12(16):48914900.
236. Castle PE, Giuliano AR. Chapter 4: genital tract infections, cervical
inflammation, and antioxidant nutrientsassessing their roles as human
papillomavirus cofactors. Jnci Monographs. 2003;2003(31):2934.
237. Hawes SE, Kiviat NB. Are genital infections and inflammation
cofactors in the pathogenesis of invasive cervical cancer? Oxford
University Press; 2002:15921593.
238. Koster S, Gurumurthy RK, Berger H, et al. Chlamydia coinfection
inhibits HPVinduced safeguards of the cellular and genomic integrity in
patientderived ectocervical organoids. bioRxiv. 2021:146.
239. Anacker DC, Moody CA. Modulation of the DNA damage response
during the life cycle of human papillomaviruses. Virus Res.
2017;231:4149.
240. Moody CA, Laimins LA. Human papillomavirus oncoproteins:
pathways to transformation. Nat Rev Cancer. 2010;10(8):550560.
241. Ngoi NY, Sundararajan V, Tan DS. Exploiting replicative stress in
gynecological cancers as a therapeutic strategy. Int J Gynecol
Cancer. 2020;30(8):12241238.
242. Mi Y, Gurumurthy RK, Zadora PK, Meyer TF, Chumduri C.
Chlamydia trachomatis inhibits homologous recombination repair
of DNA breaks by interfering with PP2A signaling. mBio. 2018;9(6):
14651518.
243. Zhang L, Wu J, Ling MT, Zhao L, Zhao KN. The role of the PI3K/
Akt/mTOR signalling pathway in human cancers induced by
infection with human papillomaviruses. Mol Cancer. 2015;14(1):87.
244. Bridge G, Rashid S, Martin S. DNA mismatch repair and oxidative
DNA damage: implications for cancer biology and treatment.
Cancers. 2014;6(3):15971614.
245. RajićJ, InicKanada A, Stein E, et al. Chlamydia trachomatis
infection is associated with Ecadherin promoter methylation,
downregulation of Ecadherin expression, and increased expres-
sion of fibronectin and αSMAimplications for epithelial
mesenchymal transition. Front Cell Infect Microbiol. 2017;7:253.
246. Igietseme JU, Omosun Y, Stuchlik O, et al. Role of epithelial
mesenchyme transition in Chlamydia pathogenesis. PLoS ONE.
2015;10(12):e0145198.
247. Silva J, Cerqueira F, Medeiros R. Chlamydia trachomatis infection:
implications for HPV status and cervical cancer. Arch Gynecol
Obstet. 2014;289:715723.
248. Paavonen J. Chlamydia trachomatis infections of the female genital
tract: state of the art. Ann Med. 2012;44(1):1828.
249. Lechien JR, Seminerio I, Descamps G, et al. Impact of HPV infection on
the immune system in oropharyngeal and nonoropharyngeal squa-
mous cell carcinoma: a systematic review. Cells. 2019;8(9):1061.
250. Dai W, Gui L, Du H, Li S, Wu R. The association of cervicovaginal
Langerhans cells with clearance of human papillomavirus. Front
Immunol. 2022;13:918190.
251. Lu Y, Wu Q, Wang L, Ji L Chlamydia trachomatis enhances HPV
persistence through immune modulation. 2022.117
252. Castanheira CP, Sallas ML, Nunes RAL, Lorenzi NPC, Termini L.
Microbiome and cervical cancer. Pathobiology. 2021;88(2):
187197.
253. Lugo LZA, Puga MAM, Jacob CMB, et al. Cytokine profiling of
samples positive for Chlamydia trachomatis and Human papillo-
mavirus. PLoS ONE. 2023;18(3):e0279390.
254. Gonzalez K, Watts TL. Understanding th e link between the oral
microbiome and the development and progression of head and
neck squamous cell carcinoma. Curr Opin Physiol. 2021;23:
100471.
255. Zhang Y, D'Souza G, Fakhry C, et al. Oral human papillomavirus
associated with differences in oral microbiota beta diversity and
microbiota abundance. J Infect Dis. 2022;226(6):10981108.
256. Orlandi E, Iacovelli NA, Tombolini V, et al. Potential role of
microbiome in oncogenesis, outcome prediction and therapeutic
targeting for head and neck cancer. Oral Oncol. 2019;99:104453.
257. Robayo DAG, Erira HAT, Jaimes FOG, Torres AM, Galindo AIC.
Oropharyngeal squamous cell carcinoma: human papilloma virus
coinfection with Streptococcus anginosus.Braz Dent J. 2019;30:
626633.
258. Tuominen H, Rautava S, Syrjänen S, Collado MC, Rautava J. HPV
infection and bacterial microbiota in the placenta, uterine cervix
and oral mucosa. Sci Rep. 2018;8(1):9787.
259. Onywera H, Williamson AL, Ponomarenko J, Meiring TL. The penile
microbiota in uncircumcised and circumcised men: relationships
with HIV and human papillomavirus infections and cervicovaginal
microbiota. Front Med. 2020;7:383.
How to cite this article: Akbari E, Milani A, Seyedinkhorasani
M, Bolhassani A. HPV coinfections with other pathogens in
cancer development: a comprehensive review. J Med Virol.
2023;95:e29236. doi:10.1002/jmv.29236
AKBARI ET AL.
|
21 of 21
... Also, it has been proven that Chlamydia trachomatis function as a strong cofactor for HPV-related cervical cancer [69,70]. Indeed, CT provides an ideal inflammatory microenvironment for HPV persistence, because this bacterium has the potential to cause cell damage in cervical mucosa and immune escape strategies, contributing to cervical neoplasia development [71,72]. CT has immune evasion mechanisms, in particular, it produces Chlamydial-protease-like activity factor (CPAF), which has an incredible ability to destroy the major histocompatibility complex (MHC), the cellular machinery involved in the adaptative immune response, capable of antigen presentation and subsequent T-cell activation [73]. ...
Article
Full-text available
Some infectious agents have the potential to cause specific modifications in the cellular microenvironment that could be propitious to the carcinogenesis process. Currently, there are specific viruses and bacteria, such as human papillomavirus (HPV) and Helicobacter pylori, that are well established as risk factors for neoplasia. Chlamydia trachomatis (CT) infections are one of the most common bacterial sexually transmitted infections worldwide, and recent European data confirmed a continuous rise across Europe. The infection is often asymptomatic in both sexes, requiring a screening program for early detection. Notwithstanding, not all countries in Europe have it. Chlamydia trachomatis can cause chronic and persistent infections, resulting in inflammation, and there are plausible biological mechanisms that link the genital infection with tumorigenesis. Herein, we aimed to understand the epidemiological and biological plausibility of CT genital infections causing endometrial, ovarian, and cervical tumors. Also, we covered some of the best suitable in vitro techniques that could be used to study this potential association. In addition, we defend the point of view of a personalized medicine strategy to treat those patients through the discovery of some biomarkers that could allow it. This review supports the need for the development of further fundamental studies in this area, in order to investigate and establish the role of chlamydial genital infections in oncogenesis.
... HPV is a significant sexually transmitted infectious pathogen, and persistent HR-HPV infection is a leading cause of cervical cancer. HPV types 16 and 18 are responsible for approximately 70% of all cervical cancer cases (3). The increasing prevalence of co-infections between HPV and other RTIs, coupled with changing modern lifestyles and sexual attitudes, highlights the need for heightened attention in public health and medicine. ...
Article
Full-text available
What is already known about this topic? Previous studies have indicated a possible association between reproductive tract infections (RTIs) and high-risk human papillomavirus (HPV) infection, but the evidence is still inconclusive. What is added by this report? This multicenter study found significantly higher positive rates of HPV, including general HPV, high-risk HPV, and HPV 16/18 infections, among women who tested positive for single or multiple RTIs compared to women who tested negative for RTIs in gynecological outpatient clinics. What are the implications for public health practice? Infection with HPV, especially high-risk types, is linked to RTIs and imbalances in the vaginal microbiota. Implementing standardized protocols for identifying and treating RTIs could support the establishment of a healthy vaginal microenvironment. This, in turn, may offer a novel approach to preventing cervical cancer.
Article
Full-text available
Sexually transmitted infections (STIs) have detrimental effects on overall health and can cause infertility in males and females. Untreated infections like gonorrhea or chlamydia may lead to pelvic inflammatory disease (PID), causing damage and obstruction of the fallopian tubes. Human papillomavirus (HPV) infections, particularly high-risk strains, may progress to cervical cancer, requiring invasive treatments and may cause fertility loss. Herpes simplex virus (HSV) outbreaks can disrupt sexual contact, affecting conception attempts. Syphilis, is another sexually transmitted disease, if left untreated, can lead to epididymitis and erectile dysfunction in males, indirectly influencing fertility. Early detection and treatment of these STIs are crucial to protect infertility in males and females.
Article
Full-text available
Objectives: The evidence in the literature regarding the relationship between Trichomonas vaginalis (TV) infection and cervical neoplasia is conflicting. The main aim of this study was to evaluate the magnitude of the risk of cervical neoplasia associated with TV infection. Methods: A meta-analysis of observational studies, which provided raw data on the association of TV infection with cervical neoplasia, was performed. For this aim, we searched scientific databases (PubMed/Medline, Scopus, the Web of Sciences, and Embase) from inception to March 15, 2023. A random-effects model was applied by Stata 17.0 to calculate the pooled and adjusted odds ratios (ORs) with 95% confidence intervals (CI), including subgroup, sensitivity, and cumulative analyses to explore sources of heterogeneity. Results: Of the 2584 records initially identified, 35 eligible studies contributed data for 67,856 women with cervical neoplasia, and 933,697 healthy controls from 14 countries were included. The pooled (2.15; 1.61-2.87; I2 = 87.7%) and adjusted (2.17; 1.82-2.60; I2 = 31.27%) ORs indicated a significant positive association between TV infection and the development of cervical neoplasia. There was no significant change in pooled and adjusted ORs by applying sensitivity and cumulative analyses, indicating the robustness of our findings. The pooled OR was significant in most sub-group analyses. There was no publication bias in the included studies. Conclusion: Our findings indicated that women with a TV infection are at significantly greater risk of cervical neoplasia. Future research, particularly longitudinal and experimental studies, should be done to better understand the various aspects of this association.
Article
Full-text available
Objectives To determine the feasibility (as measured by tolerability and safety) and efficacy of topical 5‐fluorouracil (5‐FU) and imiquimod for the treatment of cervical intraepithelial neoplasia (CIN) 2/3. Methods This pilot prospective study was conducted in women aged 18–45 years with p16+ CIN 2/3. Participants underwent an 8‐week alternating regimen of self‐applied 5% 5‐FU on weeks 1, 3, 5, and 7 and physician‐applied imiquimod on weeks 2, 4, 6, and 8. Adverse events (AEs) were collected by symptom diary and clinical exam. Feasibility was measured by tolerability and safety (AEs) of the study intervention. Tolerability was assessed as the number of participants able to apply 50% or more of the treatment doses. The safety outcome was calculated as the number of participants who experienced “specified AEs” defined as possibly, probably, or definitely related grade 2 or worse AE or grade 1 genital AEs (blisters, ulcerations, or pustules) lasting more than 5 days. The efficacy of the intervention was determined by histology and high‐risk human papillomavirus (hrHPV) testing was done after treatment. Results The median age of the 13 participants was 27 ± 2.9 years. Eleven (84.61%) participants applied 50% or more of the treatment. All participants reported grade 1 AEs; 6 (46.15%) reported grade 2 AEs; and 0 reported grade 3/4 AEs. Three (23.08%) participants had specified AEs. Histologic regression to normal or CIN 1 among those completing 50% or more of the treatment doses was observed in 10 (90.91%) participants, and 7 (63.63%) tested negative for hr‐HPV at the end of the study. Conclusions Topical treatment for CIN 2/3 with 5‐FU/imiquimod is feasible, with preliminary evidence of efficacy. Topical therapies need further investigation as adjuncts or alternatives to surgical therapy for CIN 2/3.
Article
Full-text available
Human papillomavirus (HPV) is causally associated with 5% of cancers, including cancers of the cervix, penis, vulva, vagina, anus and oropharynx. The most carcinogenic HPV is HPV-16, which dominates the types causing cancer. There is also sufficient evidence that HPV types 18, 31, 33, 35, 39, 45, 51, 52, 56, 58 and 59 cause cervical cancer. The L1 protein, which, when assembled into virus-like particles, induces HPV-type-specific neutralising antibodies, forms the basis of all commercial HPV vaccines. There are six licensed prophylactic HPV vaccines: three bivalent, two quadrivalent and one nonavalent vaccine. The bivalent vaccines protect from HPV types 16 and 18, which are associated with more than 70% of cervical cancers. Prophylactic vaccination targets children before sexual debut, but there are now catch-up campaigns, which have also been shown to be beneficial in reducing HPV infection and disease. HPV vaccination of adults after treatment for cervical lesions or recurrent respiratory papillomatosis has impacted recurrence. Gender-neutral vaccination will improve herd immunity and prevent infection in men and women. HPV vaccines are immunogenic in people living with HIV, but more research is needed on the long-term impact of vaccination and to determine whether further boosters are required.
Article
Full-text available
Background Trichomonas vaginalis infection is the most prevalent non‐viral sexually transmitted infection (STI) in women and has been suggested as a risk factor for developing cervical cancer. Objective We aimed to investigate the associations between T. vaginalis infection and cervical carcinogenesis. Search Strategy A comprehensive systematic search was conducted in five databases on 21 October 2021. Selection Criteria Studies assessing the relationship between T. vaginalis infection, HPV co‐infections, cervical dysplasia, and cervical cancer were found eligible. Data Collection and Analysis Summary estimates for pooled odds ratios (ORs) and their 95% confidence intervals (CIs) were calculated with a random‐effects model. Statistical heterogeneity was measured with I² and Cochran's Q tests. Main Results The 29 articles included 473 740 women, of whom 8518 were T. vaginalis‐positive. Our results showed that T. vaginalis‐infected women had 1.79 times higher odds of being diagnosed with HPV co‐infection (95% CI 1.27–2.53; I² 95%). We also found that T. vaginalis infection was associated with high‐grade squamous intraepithelial lesion diagnosis (OR 2.34, 95% CI 1.10–4.95; I² 75%) and cervical cancer (OR 5.23, 95% CI 3.03–9.04; I² 3%). Conclusions Our results showed an association between T. vaginalis and cervical carcinogenesis in sexually active women.
Article
Full-text available
Introduction Human papillomavirus (HPV) infection, especially persistent high-risk HPV, is associated with cervical cancer. Female reproductive tract microecological disorders and lower genital tract infections have been increasingly correlated with HPV infection and cervical lesions. Due to their common risk factors and transmission routes, coinfection with other sexually transmitted infections (STIs) has become a concern. Additionally, the clinical significance of Mycoplasma subtypes appear to vary. This study aimed to assess the correlations between common STIs and HPV infection, and to investigate the clinical significance of Mycoplasma subtypes. Methods We recruited 1,175 patients undergoing cervical cancer screening at the Peking University First Hospital gynecological clinic from March 2021 to February 2022 for vaginitis and cervicitis tests. They all received HPV genotyping and detection of STIs, and 749 of them underwent colposcopy and cervical biopsy. Results Aerobic vaginitis/desquamative inflammatory vaginitis and STIs (mainly single STIs) were found significantly more often in the HPV-positive group than in the HPV-negative group. Among patients with a single STI, rates of infection with herpes simplex virus type 2 or UP6 in the HPV-positive group were significantly higher than in the HPV-negative group (ORadj: 1.810, 95%CI: 1.211–2.705, P=0.004; ORadj: 11.032, 95%CI: 1.465–83.056, P=0.020, respectively). Discussion Through detailed Mycoplasma typing, a correlation was found between different Mycoplasma subtypes and HPV infection. These findings suggest that greater attention should be paid to detecting vaginal microecological disorders in those who are HPV-positive. Further, lower genital tract infections, including both vaginal infections and cervical STIs, are significantly more common among women who are HPV-positive and who thus require more thorough testing. Detailed typing and targeted treatment of Mycoplasma should become more routine in clinical practice.
Article
Full-text available
Persistent human papillomavirus (HPV) infection is closely associated with cervical carcinoma. Co-infection in the endocervical environment with other microorganisms, such as Chlamydia trachomatis, may increase the risk of HPV infection and neoplastic progression. While in some individuals, Chlamydia trachomatis infection is resolved with the activation of Th1/IFN-γ-mediated immune response, others develop a chronic infection marked by Th2-mediated immune response, resulting in intracellular persistence of the bacterium and increasing the risk of HPV infection. This work aimed to quantify cytokines of the Th1/Th2/Th17 profile in exfoliated cervix cells (ECC) and peripheral blood (PB) of patients positive for Chlamydia trachomatis DNA, patients positive for Papillomavirus DNA, and healthy patients. Cytokine levels were quantified by flow cytometry in ECC and PB samples from patients positive for C. trachomatis DNA (n = 18), patients positive for HPV DNA (n = 30), and healthy patients (n = 17) treated at the Hospital de Amor, Campo Grande-MS. After analysis, a higher concentration of IL-17, IL-6, and IL-4 (p <0.05) in ECC; INF-γ and IL-10 (p <0.05) in PB was found in samples from patients positive for C. trachomatis DNA compared to samples from healthy patients. When comparing samples from patients positive for HPV DNA, there was a higher concentration of cytokines IL-17, IL-10, IL-6, and IL-4 (p <0.05) in ECC and IL-4 and IL-2 (p <0.05) in PB of patients positive for C. trachomatis DNA. These results suggest that induction of Th2- and Th17 mediated immune response occurs in patients positive for C. trachomatis DNA, indicating chronic infection. Our results also demonstrate a high concentration of pro-inflammatory cytokines in ECC of patients positive for C. trachomatis DNA.
Article
Full-text available
Chlamydia trachomatis and human papillomavirus (HPV) are the most common pathogens found in sexually transmitted infections (STIs), and both are known to increase the risk of cervical cancer (CC) and infertility. HPV is extremely common worldwide, and scientists use it to distinguish between low-risk and high-risk genotypes. In addition, HPV transmission can occur via simple contact in the genital area. From 50 to 80% of sexually active individuals become infected with both C. trachomatis and HPV viruses during their lifetime, and up to 50% become infected with an HPV oncogenic genotype. The natural history of this coinfection is strongly conditioned by the balance between the host microbiome and immune condition and the infecting agent. Though the infection often regresses, it tends to persist throughout adult life asymptomatically and silently. The partnership between HPV and C. trachomatis is basically due to their similarities: common transmission routes, reciprocal advantages, and the same risk factors. C. trachomatis is a Gram-negative bacteria, similar to HPV, and an intracellular bacterium, which shows a unique biphasic development that helps the latter continue its steady progression into the host throughout the entire life. Indeed, depending on the individual’s immune condition, the C. trachomatis infection tends to migrate toward the upper genital tract and spread to the uterus, and the fallopian tubes open up a pathway to HPV invasion. In addition, most HPV and C. trachomatis infections related to the female genital tract are facilitated by the decay of the first line of defense in the vaginal environment, which is constituted by a healthy vaginal microbiome that is characterized by a net equilibrium of all its components. Thus, the aim of this paper was to highlight the complexity and fragility of the vaginal microenvironment and accentuate the fundamental role of all elements and systems involved, including the Lactobacillus strains (Lactobacillus gasseri, Lactobacillus jensenii, Lactobacillus crispatus) and the immune–endocrine system, in preserving it from oncogenic mutation. Therefore, age, diet, and genetic predisposition together with an unspecific, persistent low-grade inflammatory state were found to be implicated in a high frequency and severity grade of disease, potentially resulting in pre-cancerous and cancerous cervical lesions.
Conference Paper
Introduction/Background STIs can impact HPV infection and persistence, potentially predisposing HPV-related cervical cancer development. This study examines HPV genotype prevalence and co-occurrence with other STIs to inform targeted prevention and treatment strategies for reducing cervical cancer incidence Methodology 129 female patients aged 18–57 were enrolled based on the presence of anogenital warts, individuals with a history of risky sexual behaviors, having a partner with HPV infection, or voluntarily seeking HPV screening. Patients with a history of any STIs, prior HPV vaccination, systemic illnesses, or undergoing cancer treatment were excluded. Results The STI positivity rate among HPV-positive patients was 63.9%, significantly higher than HPV-negative cases. In the genital warts group at admission, Ureaplasma Parvum (UP) was the most common STI (40.0%), followed by Uraeplasma Urealyticum (UU) (28.5%), Mycoplasma Hominis (MH) (17.2%), and Chlamidia Trachomatis (CT) (11.4%). In the asymptomatic group, UP was also the most common STI (41.2%).The prevalence of UP was significantly higher (53.7%) in the HPV-positive group, suggesting a 6.96-fold greater risk of UP infection in individuals with HPV.View this table: • View inline • View popup • Download powerpoint Abstract #963 Table 1 Comparison of STI pathogenes by HPV positivity. Conclusion This study demonstrates a high co-infection rate between HPV and UP, emphasizing the importance of genital infection screening for high-risk HPV-positive women. Disclosures no disclosure
Article
Squamous cell carcinoma of the oral cavity (OSCC) is the most common head and neck malignancy. Importantly, we are experiencing an alarming rise in the incidence of oropharyngeal squamous cell carcinoma (OPSCC) globally. Oncogenic viruses, human papillomavirus (HPV) and Epstein-Barr virus (EBV), are known to be co-associated with OSCC and OPSCC cases. However, the reported incidence of HPV and EBV co-infection in OSCCs and OPSCCs globally is unknown. To address this, we performed a formal meta-analysis and systematic review on published studies that report the detection of both EBV and HPV in OSCCs and OPSCCs. Our analysis revealed 18 relevant studies out of a total of 1820 cases (1181 from the oral cavity and 639 from the oropharynx). Overall, HPV and EBV co-infection was found in 11.9% of OSCC and OPSCC cases combined (95% CI: 8-14.1%). Based on anatomical subsite, dual positivity estimates were 10.5% (95% CI: 6.7- 15.1%) for OSCC and 14.2% (95% CI: 9.1-21.3%) for OPSCC. The highest dual positivity rates described were in European countries: for OSCC 34.7% (95% CI: 25.9 - 44.6%) in Sweden and for OPSCC, 23.4% (95% CI: 16.9 - 31.5%) in Poland. Given these substantive prevalence rates, the value of detecting dual infection in the diagnosis and prognosis of these cancers deserves careful longitudinal studies, as do implications for cancer prevention and therapy. We further proposed molecular mechanisms that could explain how HPV and EBV could co-contribute to the aetiology of OSCCs and OPSCCs. This article is protected by copyright. All rights reserved.
Article
In 2020, approximately 604 127 patients were newly diagnosed with cervical cancer and 341 831 died of the disease worldwide. Unfortunately, 85-90% of new cases and deaths occur in less developed countries. It is well known that persistent human papillomavirus (HPV) infection is the main risk factor for developing the disease. There are more than 200 HPV genotypes identified, but the most important in public health are the high-risk HPV genotypes including HPV 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, and 59 due to their strong association with cervical cancer. Among these, genotypes 16 and 18 are responsible for about 70% of cervical cancer cases worldwide. Implementing systematic cytology-based screening, HPV screening, and HPV vaccination programs have successfully decreased the cervical cancer burden, particularly in developed countries. Although the etiological agent has been identified, we have seen the impact of well-conducted screening programs in developed countries, and we have available vaccines, the fight against this preventable disease has shown poor results globally. In November 2020 the World Health Organization launched its strategy to eliminate cervical cancer from the earth by 2130 (the goal is to achieve a global incidence lower than 4 per 100 000 women/year). The strategy aims to vaccinate 90% of girls before 15 years of age, to screen with a highly sensitive test (HPV-based) 70% of women at 35 and 45 years of age, and to provide proper treatment by trained personnel to 90% of women diagnosed with either cervical dysplasia or invasive cervical cancer. The objective of this review is to update the state of the art on primary and secondary prevention of cervical cancer.