Subido por valentina garcia

The natural history of human papillomavirus infection

Anuncio
Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
Contents lists available at ScienceDirect
Best Practice & Research Clinical
Obstetrics and Gynaecology
journal homepage: www.elsevier.com/locate/bpobgyn
1
The natural history of human papillomavirus
infection
a, b, *, Maria Brotons a, c,
Silvia de Sanjose
n a, b
Miguel Angel Pavo
Biom
Cancer Epidemiology Research Programme, Institut d’Investigacio
edica de Bellvitge (IDIBELL),
Catalan Institute of Oncology, L'Hospitalet de Llobregat, Spain
n Biom
Centro de Investigacio
edica en Red en Epidemiologia y Salud Pública CIBERESP, Barcelona, Spain
c
n Biom
Centro de Investigacio
edica en Red en Oncologia CIBERONC, Barcelona, Spain
a
b
a b s t r a c t
Keywords:
Natural history
HPV
Risk factors
Cervical cancer
Acquisition
Persistence
Human papillomavirus (HPV) is a small double-stranded DNA virus
that commonly infects humans. The oncogenic characteristics of
HPV derive from the oncoproteins E6 and E7 that act inhibiting
p53 and pRB tumor suppressors. About 5% of all cancers worldwide are attributable mainly to those known as high-risk,
including HPV types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, and
59. Infection with HPV is common after sexual initiation, but the
majority of HPV infections do not cause symptoms or disease and
are cleared within 12e24 months post-infection. Only a small
fraction of those infections that persist or progress to a preneoplastic lesion result in cancer. Persistence of HPV infection is
needed to start the oncogenic process. Clearance of infection is
common in young adults. Viral load and viral type are the main
cofactors for progression from infection to cervical intraepithelial
lesions and cancer. Smoking, hormonal exposure, and HIV are
additional exposures that increase the risk of progression to cancer. The adverse health effects of HPV infections can be largely
controlled through vaccination and screening.
© 2017 Published by Elsevier Ltd.
Biome
dica de Bellvitge (IDIBELL),
* Corresponding author. Cancer Epidemiology Research Programme, Institut d’Investigacio
d'Oncologia, Av. Gran Via de l'Hospitalet 199-203, 08908 L'Hospitalet de Llobregat, Spain.
CIBERESP, Institut Catala
).
E-mail address: [email protected] (S. de Sanjose
http://dx.doi.org/10.1016/j.bpobgyn.2017.08.015
1521-6934/© 2017 Published by Elsevier Ltd.
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
3
Introduction
Human papillomavirus (HPV) has been identified as the cause of approximately 5% of all cancers
worldwide [1]. HPV infection is associated with virtually all cervical cancers and a significant proportion of anogenital (vulvar, vagina, penile, and anal) and oropharyngeal cancers [2]. HPV infection is
also associated with other skin and mucosal lesions such as warts and benign papillomas [3]. The
majority of HPV infections do not cause symptoms or disease and are cleared within 12e24 months
post infection. Only a small fraction of those infections that persist or progress to a preneoplastic lesion
result in cancer. Each infection stage and behavior may be influenced by environmental, host, and viral
factors, the knowledge of which is essential to understand the natural history of the HPV infection and
to develop new tools for improving the management of HPV-positive lesions and the prevention, early
detection, and treatment of HPV-associated cancers.
HPV infects both men and women, although the burden of attributable disease is much larger in
women because of the high susceptibility to HPV infection of cervical cells [1]. Thus, the accumulation
of research data is far larger for this site as it is our understanding of the natural history of cervical HPV.
We recommend specific reviews on HPV in other sites [4,5]. In this chapter, we will review the natural
history of genital HPV infections focusing on the biological characteristics of HPV and factors intervening in its acquisition, clearance, persistence, and progression to human cancer and particularly
cervical cancer. Other chapters will provide detailed information on HPV worldwide burden of related
disease and relevant interventions to prevent the adverse effects of HPV infections.
HPV classification
More than 200 HPV genotypes have been identified in the last century and grouped in different
genera (Alpha-, Nu-/Mu-, Beta- and Gamma-papillomavirus) according to the viral genome structure
and tropism to human epithelial tissues [6e8]. The Alpha genus includes genotypes that have been
described to cause cancer, while Beta- and Gamma-papillomavirus infection courses are generally
asymptomatic, but immunosuppression states (i.e. organ graft, HIV infection, epidermodysplasia verruciformis disorder among others) can trigger these types to produce cutaneous papilloma or increase
the predisposition to skin cancer [9].
Twelve types (16,18, 31, 33, 35, 39, 45, 51, 52, 56, 58, and 59), also known as high-risk types, have been
classified as carcinogenic to humans according to the International Agency for Research on Cancer
(Table 1). Low-risk types including HPV6 or HPV11 generally cause benign diseases such as genital warts
[10], while other types classified as probably or possible carcinogenic are rarely found in large series of
cancers or are associated with additional factors, so their oncogenicity remains to be clarified [11].
HPV genome
Despite the papillomavirus family representing a remarkably heterogeneous group of viruses, they
share the same genome structure and organization [12] (Fig. 1). A circular double-stranded DNA genome
of approximately 8 kb is structured into three main regions: (i) The early region (E) encodes genes that
Table 1
Broad classification of human papillomavirus (HPV) by oncogenic risk and associated diseases according to the International
Agency for Research on Cancer Evaluation.
Human papillomavirus
Genotypes
Associated disease
High risk or oncogenic
HPV 16, 18, 31, 33, 35, 39,
45, 51, 52, 56, 58, 59
HPV 6, 11
HPV 68
HPV 5, 8
Cervical, anal, vaginal, vulvar, penile, and oropharyngeal
cancer and associated precursor lesions
Genital warts, recurrent respiratory papillomatosis
Cervical cancer
Squamous cell carcinoma of the skin in patients
affected by epidermodysplasia verruciformis
Uncertain
Low-risk types
Probable carcinogenic
Possible carcinogenic
Possible carcinogenic
HPV 26, 30, 34, 53, 66, 67,
69, 70, 73, 82, 85, and 97
4
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
URR: origin of replication,
transcription factor-binding
sites and gene expression
control
URR
E1: Viral DNA replication and transcription
E7
E6
E2: Viral DNA replication, apoptosis,
transcriptional repressor of E6/E7
E4: viral DNA replication
L1: Major capsid protein
L2: Minor capsid protein
HPV16
L1
E1
dsDNA
7906 bp
L2
E5
E5: cell proliferation, alteration of signaling
pathways (EGFR), immune recognition ( MHC)
and apoptosis
E6: p53 degradation, alteration of cell cycle
regulation, apoptosis resistance chromosomal
instability and cell immortalization (TERT)
E4
E2
E7: pRB degradation, re-entry into S-phase cell
cycle, p16 realize and overexpression,
chromosomal instability
Fig. 1. HPV 16 structure and viral proteins.
are necessary for the viral cycle and with an important role in cell transformation (E1, E2, E4, E5, E6, and
E7). (ii) The late region (L) encodes the L1 and L2 capsid proteins. (iii) The upstream regulatory protein,
referred also as the long control region, is a noncoding region containing the replication origin and
transcription factor-binding sites that contribute to regulate DNA replication by controlling the viral
gene transcription. E6 and E7, together with E1, E2, E4, and E5, expression are essential for viral genome
replication and virion synthesis and release, but they also play a key role in cell transformation [9].
HPV life cycle
HPV life cycle begins with the infection of the basal layer through microtraumas that compromise
the epithelial barrier [13] (Fig. 2). HPV genome is maintained at low copy number in the infected host
basal cells. Upon differentiation of epithelial cells, the virus replicates to a high copy number and
expresses the capsid genes (L1 and L2), resulting in the production of new progeny virions that are
released from the epithelial surface. For persistence, HPV needs to infect basal cells showing stem celllike features that are still able to proliferate [14]. This phenomenon is far less common in HPV low-risk
types. The epithelial transition zones, such as the endo-/ectocervix and anorectal junctions, are regions
more susceptible to carcinogenesis by high-risk HPV types [15]. High-risk types are more prone to
activate cell proliferation in basal and differentiated layers promoting the transition from a productive
infection to an infection, which is unable to complete the viral life cycle but is able to activate several
pathways essential for the epithelial transformation. One plausible explanation of the increased
oncogenic capacity of the high-risk types and particularly the HPV16 type resides in the activity of the
E6 and E7 oncoproteins. Although the E6 and E7 activity is present in high- and low-risk types, their
role in low-risk types is limited to increase the viral fitness and viral production and it is largely
insufficient to trigger the development of preneoplastic lesions and cancer [16].
High-risk HPV types have developed several mechanisms to avoid host immune response, which is
important for viral persistence and progression to HPV-associated neoplastic diseases. One of the first
strategies to avoid detection is to maintain a very low profile [17,18]. The HPV cycle is exclusively
intraepithelial and not lytic, therefore preventing the associated pro-inflammatory signal. As a result,
the recruitment of antigen-presenting cells such as Langerhans cells (LC) and the release of cytokines
that mediate the immune response are absent or very low after the HPV infection. Other mechanisms
of HPV immune evasion include the regulation of the interferon signaling, inhibition of LC by the E6
and E7 activity, inhibition of adherence molecules such as the CDH1, and modulation of intracellular
signaling pathways.
HPV-mediated carcinogenesis
E6 and E7 early proteins conduct their main role in the carcinogenic process through the inhibition
of p53 and pRB tumor suppressors [19,20]. E6 functions include also the activation of the telomerase
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
Fig. 2. Schematic representation of the HPV infection in cervical mucose and its different potential squamous intrepitelial lesions.
5
6
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
activity and deregulation of pathways involved in immune system response, epithelial differentiation,
cell proliferation, and survival signaling. Besides cell cycle deregulation and proliferation, E7 enhances
genomic instability and promotes the accumulation of chromosomal abnormalities. The deregulation
of the cell cycle, the activation of the telomerase activity, and genomic instability create a favorable
environment for the epithelial cell transformation. HPV integration can also drive the carcinogenic
process through the inactivation of the E2 expression, the main inhibitor of E6 and E7, and the
disruption of host genes because of the viral sequence insertion [21].
The carcinogenic process, initiated with the E6 and E7 activation, needs to be complemented by the
accumulation of additional alterations in the host gene to lead to the invasive cancer phenotype. The
integrated genomic analysis conducted by The Cancer Genome Atlas Consortium identified genes that
were significantly mutated in cervical and head and neck HPV-associated tumors [22,23]. Interestingly,
a high percentage of the identified mutations show a pattern compatible with the activity of APOBEC,
an innate immune system that can bind and edit the viral DNA, restricting the viral infection. APOBEC3B can be activated, in turn, by E6 and E7 oncoproteins [24,25]. Thus, APOBEC could be an
important source of mutagenesis in HPV-associated cancers, as has been reported in multiple human
cancers [26].
Viral genetic variation, beyond the HPV genotype, could explain in part differences in clearance,
persistence, and risk to develop cancer among infections positive for the same type. Papillomaviruses
are classified into types depending on L1 sequencing, defining a new type when its genome differs by at
least 10% from that of all other classified types [27]. HPV isolates of the same type that differ by less
than 10% in the genome sequence are classified in variant lineages and sublineages. These variants have
shown differences to be associated with the risk of cancer. For example, women with a HPV16 non-A
variant (B, C, and D) were consistently at an elevated risk of cervical cancer compared to women with
other variants. Moreover, D2/D4 sublineages were more common in glandular cervical lesions, while
A1/A2 variants were identified in the large majority (75.4%) of the squamous cell carcinomas of the
cervix [27,28].
Growing evidence suggests that epigenetic alterations directly or indirectly related with the E6 and
E7 activity are common events during the early steps of epithelial malignization and have been
described as potential biomarkers for cervical cancer [29,30].
HPV prevalence, acquisition, and clearance in the population
Fig. 3 summarizes cervical HPV DNA prevalence by age observed after pooling existing data
worldwide [31]. The figure refers to prevalence, which results from the combined effect of incidence
(acquisition) and duration of infection (clearance/persistence). The majority of anogenital HPV infections are acquired through sexual contact, and acquisition is strongly determined by the accumulation of sexual partners and their respective sexual behavior [32]. A new infection can be detected
soon after the first sexual contact with an infected partner, and most of them will be detectable within
1 year of exposure [33,34]. As a result of this high transmission, HPV infections are very common in
young women depicting the peak in prevalence observed in the figure, which generally takes place
around 20e25 years of age. An abrupt decline follows as a result of frequent clearance and lower
exposure to new partners. This pattern is consistent in populations all over the world and confirms the
sexual transmission as the main mode of transmission. Variations to the left/right of the peak age will
relate to the average age of the sexual initiation in a given population. The intensity of the peak will be
modulated by the average number of sexual partners interchange in men and women. The shape of the
curve could also be affected by screening practices. Infections leading to cervical intraepithelial lesions
may be detected through screening and thus its natural evolution be modified by treatment. After this
period, prevalence of infection remains relatively stable at the range of 5e10%. In some countries, a
second peak is observed after menopause. Reasons for this late increase are not well understood
[16,31,35].
One of the first prospective studies to explore HPV acquisition was conducted in a cohort of 1610
women aged 15e85 years who were HPV negative at baseline in Colombia. Women were followed
every 6 months for about 5 years [36,37]. HPV acquisition was present in cohorts of all ages but
with a varying intensity, being highest in younger cohorts with a cumulative incidence of 0.42 in
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
7
Fig. 3. General pattern of the HPV DNA prevalence in the exofialted cervical cells in healthy women by age and main determinants of
the shape of the curve.
women aged 15e19 years in contrast to a cumulative incidence of 0.12 in women aged >44 years
(Fig. 4). Acquisition of new partners and having had more than one sexual partner were strong
determinants of new infections. On the contrary, having a long-standing first sexual relation
decreased the risk of new infection. Consistently [38], in a cohort of UK students, the authors
identified a significant 1.99 risk increment for new HPV infections (95% CI 1.46e2.72) among those
women who had multiple sexual partners compared to monogamous women. Two concomitant
studies involving women internet daters in Canada showed that reporting new and/or multiple
partners in the past 6 months was positively associated with increased incident HPV [39,40].
A recent analysis of the placebo group of women participating in a vaccine clinical trial resulted
in an overall detection of new HPV infections of 20.61 (95% CI 18.47e22.99) per 1000 personmonths during a 6-year follow-up. The incidence further increased by 88% among women with
more than one sexual partner as compared to those having only one partner. HPV16 was the most
common new infection with an incidence rate per 1000 person-months of 4.16 (95% CI 3.44e5.02)
[41].
Following women over time has demonstrated that up to half of the HPV infections clear within
6 months and that the great majority (>90%) clear within a few years after acquisition [42,43].
Innate and adaptive immune responses are interplayers in the resolution of such common infections [16]. A Th1 pro-inflammatory response in the genital tract has been suggested as a potential mechanism for clearance [44], but cell-mediated immune response may be the most
common mechanism. Shannon et al. [45] studied in a set of 65 African/Caribbean women the
vaginal immunological environment by HPV clearance and persistence. Participants who cleared
HPV had a significantly higher absolute number of endocervical LC (5018 cells/cytobrush) than
either HPV-negative women (635 cells/cytobrush, p ¼ 0.015) or those with persistent HPV infection
(180 cells/cytobrush, p ¼ 0.023). However, no association between prevalent HPV infection and
genital inflammatory cytokines nor with endocervical CD4þ T cell numbers or presence of highly
susceptible CD4þ T cell subsets such as those expressing CCR5 or CD69 were identified in this study.
8
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
Fig. 4. Cumulative incidence of new HPV infections in 5 year follow up by age group in Colombia. Adapted from Bosch et al. Vaccine
2008.
Consistent with others, they observed a differential pattern of the vaginal microbiome among those
clearing the infection with more abundant Lactobacillus spp. [46,47]. Others have identified bacterial vaginosis associated with persistence of HPV infections among pregnant women [48]. More
data are needed to have a full understanding of how HPV infections are modulated by the vaginal
environment.
HPV antibodies acquired through natural infection are observed in a limited number of infected
people. In the US, among unvaccinated women aged 20 years, only 20% had detectable antibodies to
HPV16 [49]. A systematic review of the acquired natural immunity against subsequent genital HPV
infection identified a range of seroprevalence estimates against HPV16 of 6.2%e45.5% with a relative
modest protection for HPV16 reinfection of 0.72 (0.62e0.82) and thus unlikely to play a major role in
clearance [50]. Data from vaccine trials have shown a protective but moderate impact of natural immune response against cervical disease [51,52]. Consistent with this moderate effect, a recent followup study of 1848 women attending cervical cancer screening in Slovenia exploring the natural antibody
response against HPV16 showed that baseline anti-HPV16 antibodies were not associated with
clearance of HPV16 infection at follow-up (odds ratio for clearance ¼ 0.8; 0.3e1.9) [53]. Overall, the
natural immune response is clearly insufficient to control for new infections and is far lower when
compared to the high levels of seroresponse and the high efficacy observed against persistent infections after HPV vaccination [16].
In some circumstances, after follow-up of a positive HPV test, HPV may be undetected simulating a
cleared infection. It is now accepted that HPV infection may be latent and controlled by a cellular
immune surveillance environment in which viruses are kept in very low copy numbers escaping its
detection. Reactivation may occur under moderate (i.e., menopausal state, aging) to relevant immunosuppressive situations (i.e., organ graft) [54] with or without hormonal influence). This reactivation
could explain the second peak in HPV prevalence observed in some population in postmenopausal
women.
HPV persistence and progression to cervical intraepithelial lesions and cancer
Persistent carcinogenic HPV infections clearly predict the risk of cervical cancer in women [55,56].
Persistence is not homogeneously measured but is a very relevant concept as many populations un~ oz et al. defined
dergoing screening using HPV tests will rely on measures of persistent infection. Mun
persistence as those infections that last more than the median duration but this concept is relevant for
natural history studies. Others define persistence as those having two positive HPV DNA consecutive
tests with undetermined time interval [37,57]. Time interval between two measures affects the
persistence estimate as many infections will be cleared by year 2. Marks et al. compared 12 versus 24
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
9
months in defining a persistent infection and showed substantial increases in the specificity of the
combined two HPV tests using 24 months for the detection of cervical lesions [57]. In many screening
programs, a 12-month test repetition is recommended among those who are HPV-positive at first
screening with a negative cervical cytology. Although 12 months may be a too short interval, it may
provide some “safety window” as first infection may have a long-standing prevalence. For newly acquired infections, a longer interval may be more efficient.
Irrespective of the time window, major determinants of HPV persistence are HPV type and viral load
at first detection. It remains unclear whether age is a key element for persistence. However, in the
~ oz et al. previously described, median duration of HPV incident infection was
prospective study by Mun
higher for high-risk HPV types than for low-risk types and for HPV16. Women younger than 30 years
had a longer mean duration for HPV16 infections of 16.6 months, while women >30 years had a
significantly shorter duration of 9.5 months. Others suggest that persistence increases with increasing
age [58].
The presence of more than concomitant HPV infections, a phenomenon very common in younger
populations, is reported to not influence the duration of infection [59].
The low proportion of women who are not able to clear the infection are those at risk of developing
cervical cancer. Soon after the HPV infection is established, cellular changes can be observed in the
cervical exfoliated cells. Persistent infections can result in different grades of squamous intraepithelial
lesions that ultimately can lead to high-grade lesions and cancer in an average of 5e14 years if undetected and untreated [60] (Fig. 2).
Within the types involved in the carcinogenic process, persistent infection with HPV16 is most
commonly associated with a faster progression to cervical lesions and invasive cervical cancer
[55,56]. It is noticeable that progression in HPV16 infection is not affected by strong-acting DNA
variations of the virus [61]. HPV16 is involved in over 60% of all cervical cancers but also in other
HPV-related cancers as shown in Fig. 5 [62e66]. Other types including HPV18, 45, 31, and 33 are
also commonly detected in cancer specimens but their lower contribution suggests a different
natural history as the one of HPV16. As a consequence of these observations, detection of HPV16 in
women aged 30þ is considered in many settings an indication of immediate colposcopy for
screening of cervical cancer [67], while detection of other high-risk types may be followed by a
second triage test.
In addition to the viral characteristics, environmental or exogenous factors have long been identified as modifiers of the natural history of HPV infections leading to cervical cancer. Many of the early
caseecontrol studies were conducted in populations with low coverage of screening for cervical cancer.
Most relevant cofactors identified at the time to increase cervical cancer risk were long-term smoking,
multiparity [68], and long-term use of hormonal [69] contraceptives with an average increased risk of
1.5e2-fold in HPV-positive women [70]. Interestingly, a recent analysis of a large cohort of 308,036
Fig. 5. Most common HPV types detected in HPV DNA positive anogenital invasive cancer in the RISHPV studies.
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
10
women recruited in the European Prospective Investigation into Cancer and Nutrition (EPIC) Study,
with an average follow-up of 9 years, confirmed that increasing number of full-term pregnancies was
positively associated with increased risk of cervical intraepithelial neoplasia grade 3/carcinoma in situ
(CIN3/CIS) and that duration of oral contraceptives use was associated with a significantly increased
risk of both CIN3/CIS and cervical cancer with hazard ratios of 1.6 and 1.8, respectively, for 15 years
versus never used [71]. Identification of persistent HPV infection in smokers should alert women of the
increased risk and reinforce the message to quit smoking. In HPV-positive long-term oral contraceptive
users, a closer surveillance for cervical disease may be advisable.
Coinfection with sexually transmitted infections with Chlamydia trachomatis has been inconsistently associated with the risk of progression to cancer. A prospective study exploring the association
between C. trachomatis DNA and C. trachomatis immunoglobulin G status among HPV-positive women
and the risk of preneoplastic lesions did not find any association. The authors suggested that previous
positive associations may be explained by a confounding effect of HPV acquisition [72]. In a recent
meta-analysis [73], Zhu H et al. reported a significant increase of cervical cancer incidence in both
retrospective and prospective studies associated to C. trachomatis. The increased risk was observed
even after adjustment for HPV detection. The authors suggest that an independent effect could be
attributed to C. trachomatis. Consistency in prospective studies is still needed to confirm such a role of
C. trachomatis as coinfection may be affected by the sexual pathway that both infections share.
HIV has always been associated as a major co-factor with HPV to induce cervical cancer. The
mechanism has been largely associated to the immunosuppression conferred by active HIV infection
and not by a direct effect of HIV. A large number of HIV-infected women are now following antiretroviral therapy (ART), and therefore, it is expected that a good compliance with treatment will be
followed by a reduction of cervical lesions and cervical cancer [74]. A recent analysis compiling the
existing literature shows that women complying with ART in a prolonged time lower their risk of
acquisition of high-risk HPV types and lower the incidence of intraepithelial lesions and their progression [75]. In some settings, like in Eastern and South Africa, the high prevalence of HIV infection in
women alerts of the need to fully control ART compliance to prevent additional damage due to HPV
infection and its consequences. Further details on the impact of HIV in HPV-related disease are shown
in a following chapter 8.
Conclusion
Many enigmas, however, remain to fully understand the natural history of HPV and related diseases.
Some HPV types and particularly HPV16 have an enormous capacity to induce cancer in humans
through clearly identified oncogenes, E6 and E7. The cervix uteri is the tissue that is most vulnerable to
these HPV infections if they persist. In addition to viral load and viral type, factors such as smoking,
hormonal use, and coinfections with HIV may contribute to the progression of HPV infections into
human cancer. Prevention by vaccination and/or screening and improved treatment are key to reduce
the burden of HPV-related disease.
Financial support
This work was partially supported by grants from the Instituto de Salud Carlos III-ISCIII (Spanish
Government) co-funded by FEDER funds/European Regional Development Fund (ERDF) e A WAY TO
n cooperativa en salud (RETICS)
BUILD Europe (References: PI16/01254; Redes tem
aticas de investigacio
n me
dica en Red: CIBERESP CB06/02/0073 and CIBERONC:
RD12/0036/0056, Centro de Investigacio
n de Ayudas Universitarias y de Investigacio
n (AGAUR) de la GenCB16/12/00401), Agencia de Gestio
eralitat de Catalunya (2014SGR756) and Recercaixa 2015 (MD088652). The work conducted by M.
Brotons was supported by a grant from the Spanish Ministry of Economy and Competitiveness e Carlos
III Institute of Health (Río Hortega CM15/00061).
Conflict of interest
SdS holds an Institutional Research Grant from Merck.
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
11
Practice points
Prevalent HPV infection may represent a long-standing infection.
HPV infection is mainly acquired by young adults after sexual initiation but may occur
throughout life.
HPV may be latent and reappear under circumstances such as increased age or
immunosuppression.
Prolonged smoking and use of oral contraceptives may increase persistence of HPV infection.
Research agenda
The impact of hormones in HPV persistence and progression.
The impact of microbiome in HPV persistence.
The identification of genetic and host factors for HPV-related carcinogenesis.
Appendix A. Supplementary data
Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.bpobgyn.2017.
08.015.
References
[1] de Martel C, Ferlay J, Franceschi S, et al. Global burden of cancers attributable to infections in 2008: a review and synthetic
analysis. Lancet Oncol 2012;13(6):607e15.
[2] Bosch FX, Broker TR, Forman D, et al. Comprehensive control of human papillomavirus infections and related diseases.
Vaccine 2013;31(Suppl. 8):I1e31.
[3] Doorbar J, Egawa N, Griffin H, et al. Human papillomavirus molecular biology and disease association. Rev Med Virol 2015;
25(Suppl. 1):2e23.
[4] Taylor S, Bunge E, Bakker M, et al. The incidence, clearance and persistence of non-cervical human papillomavirus infections: a systematic review of the literature. BMC Infect Dis 2016;16:293.
[5] Giuliano AR, Nyitray AG, Kreimer AR, et al. EUROGIN 2014 roadmap: differences in human papillomavirus infection natural
history, transmission and human papillomavirus-related cancer incidence by gender and anatomic site of infection. Int J
Cancer 2015;136(12):2752e60.
[6] de Villiers EM, Fauquet C, Broker TR, et al. Classification of papillomaviruses. Virology 2004 Jun 20;324(1):17e27.
[7] Bernard HU, Burk RD, Chen Z, et al. Classification of papillomaviruses (PVs) based on 189 PV types and proposal of
taxonomic amendments. Virology 2010;401(1):70e9.
[8] de Villiers EM. Cross-roads in the classification of papillomaviruses. Virology 2013;445(1e2):2e10.
[9] Doorbar J, Quint W, Banks L, et al. The biology and life-cycle of human papillomaviruses. Vaccine 2012;30(Suppl. 5):
F55e70.
[10] Egawa N, Doorbar J. The low-risk papillomaviruses. Virus Res 2017;231:119e27.
[11] IARC. Working group on the evaluation of carcinogenic risks to humans, vol. 90; 2007. p. 1e636.
lez-S
[12] Bravo IG, Fe
anchez M. Papillomaviruses: viral evolution, cancer and evolutionary medicine. Evol Med Public Health
2015;2015(1):32e51.
[13] Stubenrauch F, Laimins LA. Human papillomavirus life cycle: active and latent phases. Semin Cancer Biol 1999;9(6):
379e86.
[14] Egawa N, Egawa K, Griffin H, et al. Human papillomaviruses; epithelial tropisms, and the development of neoplasia. Viruses 2015;7(7):3863e90.
[15] Yang EJ, Quick MC, Hanamornroongruang S, et al. Microanatomy of the cervical and anorectal squamocolumnar junctions:
a proposed model for anatomical differences in HPV-related cancer risk. Mod Pathol 2015;28(7):994e1000.
[16] Schiffman M, Doorbar J, Wentzensen N, et al. Carcinogenic human papillomavirus infection. Nat Rev Dis Prim 2016;2:
16086.
[17] Stanley M, Pinto LA, Trimble C. Human papillomavirus vaccineseimmune responses. Vaccine 2012;30(Suppl. 5):F83e7.
[18] Kanodia S, Fahey LM, Kast WM. Mechanisms used by human papillomaviruses to escape the host immune response. Curr
Cancer Drug Targets 2007;7(1):79e89.
[19] Psyrri A, DiMaio D. Human papillomavirus in cervical and head-and-neck cancer. Nat Clin Pract Oncol 2008;5(1):24e31.
[20] McLaughlin-Drubin ME, Munger K. Viruses associated with human cancer. Biochim Biophys Acta 2008 Mar;1782(3):
127e50.
12
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
[21] McBride AA, Warburton A. The role of integration in oncogenic progression of HPV-associated cancers. PLoS Pathog 2017;
13(4), e1006211.
[22] Cancer Genome Atlas Research Network. Integrated genomic and molecular characterization of cervical cancer. Nature
2017;543(7645):378e84.
[23] Cancer Genome Atlas Research Network. Comprehensive genomic characterization of head and neck squamous cell
carcinomas. Nature 2015;517(7536):576e82.
[24] Vieira VC, Leonard B, White EA, et al. Human papillomavirus E6 triggers upregulation of the antiviral and cancer genomic
DNA deaminase APOBEC3B. MBio 2014 Dec 23;5(6).
[25] Wang Z, Wakae K, Kitamura K, et al. APOBEC3 deaminases induce hypermutation in human papillomavirus 16 DNA upon
beta interferon stimulation. J Virol 2014 Jan;88(2):1308e17.
[26] Roberts SA, Lawrence MS, Klimczak LJ, et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human
cancers. Nat Genet 2013 Sep;45(9):970e6.
[27] Burk RD, Harari A, Chen Z. Human papillomavirus genome variants. Virology 2013;445(1e2):232e43.
[28] Mirabello L, Yeager M, Cullen M, et al. HPV16 sublineage associations with histology-specific cancer risk using HPV wholegenome sequences in 3200 women. J Natl Cancer Inst 2016 Apr 29;108(9).
[29] Clarke MA, Wentzensen N, Mirabello L, et al. Human papillomavirus DNA methylation as a potential biomarker for cervical
cancer. Cancer Epidemiol Biomarkers Prev 2012 Dec;21(12):2125e37.
[30] Verlaat W, Snijders PJF, Novianti PW, et al. Genome-wide DNA methylation profiling reveals methylation markers associated with 3q gain for detection of cervical precancer and cancer. Clin Cancer Res 2017;23(14):3813e22.
X, et al. Cervical human papillomavirus prevalence in 5 continents: meta-analysis of 1 million
[31] Bruni L, Diaz M, Castellsague
women with normal cytological findings. J Infect Dis 2010 Dec 15;202(12):1789e99.
[32] Kjaer SK, Chackerian B, van den Brule AJ, et al. High-risk human papillomavirus is sexually transmitted: evidence from a
follow-up study of virgins starting sexual activity (intercourse). Cancer Epidemiol Biomarkers Prev 2001 Feb;10(2):101e6.
[33] Winer RL, Feng Q, Hughes JP, et al. Risk of female human papillomavirus acquisition associated with first male sex partner.
J Infect Dis 2008 Jan 15;197(2):279e82.
[34] Moscicki AB, Ma Y, Jonte J, et al. The role of sexual behavior and human papillomavirus persistence in predicting repeated
infections with new human papillomavirus types. Cancer Epidemiol Biomarkers Prev 2010 Aug;19(8):2055e65.
[35] Baussano I, Diaz M, Tully S, et al. Effect of age-difference between heterosexual partners on risk of cervical cancer and
human papillomavirus infection. Papillomavirus Res 2017 Jun;3:98e104.
~ oz N, Me
ndez F, Posso H, et al. Incidence, duration, and determinants of cervical human papillomavirus infection in a
[36] Mun
cohort of Colombian women with normal cytological results. J Infect Dis 2004 Dec 15;190(12):2077e87.
~ oz N, Hernandez-Suarez G, Me
ndez F, et al. Persistence of HPV infection and risk of high-grade cervical intraepithelial
[37] Mun
neoplasia in a cohort of Colombian women. Br J Cancer 2009 Apr 7;100(7):1184e90.
[38] Oakeshott P, Aghaizu A, Reid F, et al. Frequency and risk factors for prevalent, incident, and persistent genital carcinogenic
human papillomavirus infection in sexually active women: community based cohort study. BMJ 2012 Jun 22;344, e4168.
[39] Winer RL, Hughes JP, Feng Q, et al. Incident detection of high-risk human papillomavirus infections in a cohort of high-risk
women aged 25-65 years. J Infect Dis 2016 Sep 1;214(5):665e75.
[40] Ma S, Stern JE, Feng Q, et al. Incidence and risk factors for human papillomavirus infections in young female online daters.
J Med Virol 2017 Nov;89(11):2029e36.
[41] Ramanakumar AV, Naud P, Roteli-Martins CM, et al. Incidence and duration of type-specific human papillomavirus
infection in high-risk HPV-naïve women: results from the control arm of a phase II HPV-16/18 vaccine trial. BMJ Open
2016 Aug 26;6(8), e011371.
[42] Rodríguez AC, Schiffman M, Herrero R, et al. Longitudinal study of human papillomavirus persistence and cervical
intraepithelial neoplasia grade 2/3: critical role of duration of infection. J Natl Cancer Inst 2010 Mar 3;102(5):315e24.
[43] Winer RL, Hughes JP, Feng Q, et al. Early natural history of incident, type-specific human papillomavirus infections in
newly sexually active young women. Cancer Epidemiol Biomarkers Prev 2011 Apr;20(4):699e707.
[44] Scott M, Stites DP, Moscicki AB. Th1 cytokine patterns in cervical human papillomavirus infection. Clin Diagn Lab Immunol
1999 Sep;6(5):751e5.
[45] Shannon B, Yi TJ, Perusini S, et al. Association of HPV infection and clearance with cervicovaginal immunology and the
vaginal microbiota. Mucosal Immunol 2017 Sep;10(5):1310e9.
[46] Brotman RM, Shardell MD, Gajer P, et al. Interplay between the temporal dynamics of the vaginal microbiota and human
papillomavirus detection. J Infect Dis 2014 Dec 1;210(11):1723e33.
[47] Dareng EO, Ma B, Famooto AO, et al. Prevalent high-risk HPV infection and vaginal microbiota in Nigerian women. Epidemiol Infect 2016 Jan;144(1):123e37.
€nen K, et al. Association of asymptomatic bacterial vaginosis with persistence of female genital
[48] Kero K, Rautava J, Syrja
human papillomavirus infection. Eur J Clin Microbiol Infect Dis 2017 Jul 5. http://dx.doi.org/10.1007/s10096-017-3048-y
[Epub ahead of print].
[49] Stone KM, Karem KL, Sternberg MR, et al. Seroprevalence of human papillomavirus type 16 infection in the United States.
J Infect Dis 2002 Nov 15;186(10):1396e402.
[50] Beachler DC, Jenkins G, Safaeian M, et al. Natural acquired immunity against subsequent genital human papillomavirus
infection: a systematic review and meta-analysis. J Infect Dis 2016 May 1;213(9):1444e54.
[51] Future II Study Group. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J
Med 2007 May 10;356(19):1915e27.
n J, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against
[52] Paavonen J, Naud P, Salmero
cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised
study in young women. Lancet 2009 Jul 25;374(9686):301e14.
[53] Triglav T, Artemchuk H, Ostrbenk A, et al. Effect of naturally acquired type-specific serum antibodies against human
papillomavirus type 16 infection. J Clin Virol May 2017;90:64e9.
[54] Maglennon GA, McIntosh PB, Doorbar J. Immunosuppression facilitates the reactivation of latent papillomavirus infections. J Virol 2014 Jan;88(1):710e6.
S. de Sanjose et al. / Best Practice & Research Clinical Obstetrics and Gynaecology 47 (2018) 2e13
13
€m KM, et al. Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up
[55] Ronco G, Dillner J, Elfstro
of four European randomised controlled trials. Lancet 2014 Feb 8;383(9916):524e32.
[56] Khan MJ, Castle PE, Lorincz AT, et al. The elevated 10-year risk of cervical precancer and cancer in women with human
papillomavirus (HPV) type 16 or 18 and the possible utility of type-specific HPV testing in clinical practice. J Natl Cancer
Inst 2005 Jul 20;97(14):1072e9.
[57] Marks MA, Castle PE, Schiffman M, et al. Evaluation of any or type-specific persistence of high-risk human papillomavirus
for detecting cervical precancer. J Clin Microbiol 2012 Feb;50(2):300e6.
[58] Castle PE, Rodríguez AC, Burk RD, et al. Long-term persistence of prevalently detected human papillomavirus infections in
the absence of detectable cervical precancer and cancer. J Infect Dis 2011 Mar 15;203(6):814e22.
[59] Campos NG, Rodriguez AC, Castle PE, et al. Persistence of concurrent infections with multiple human papillomavirus
types: a population-based cohort study. J Infect Dis 2011 Mar 15;203(6):823e7.
[60] Woodman CB, Collins SI, Young LS. The natural history of cervical HPV infection: unresolved issues. Nat Rev Cancer 2007
Jan;7(1):11e22.
[61] Van der Weele P, Meijer CJLM, King AJ. Whole-genome sequencing and variant analysis of hpv16 infections. J Virol 2017
Sep 12;91(19).
[62] de Sanjose S, Quint WG, Alemany L, et al. Retrospective International Survey and HPV Time Trends Study Group. Human
papillomavirus genotype attribution in invasive cervical cancer: a retrospective cross-sectional worldwide study. Lancet
Oncol 2010 Nov;11(11):1048e56.
S, Alemany L, Ordi J, et al. Worldwide human papillomavirus genotype attribution in over 2000 cases of
[63] de Sanjose
intraepithelial and invasive lesions of the vulva. Eur J Cancer 2013 Nov;49(16):3450e61.
[64] Alemany L, Saunier M, Tinoco L, et al. Large contribution of human papillomavirus in vaginal neoplastic lesions: a
worldwide study in 597 samples. Eur J Cancer 2014 Nov;50(16):2846e54.
[65] Alemany L, Saunier M, Alvarado-Cabrero I, et al., HPV VVAP Study Group. Human papillomavirus DNA prevalence and type
distribution in anal carcinomas worldwide. Int J Cancer 2015 Jan 1;136(1):98e107.
[66] Alemany L, Cubilla A, Halec G, et al. Role of human papillomavirus in penile carcinomas worldwide. Eur Urol 2016 May;
69(5):953e61.
[67] Saslow D, Solomon D, Lawson HW, et al. American cancer society, American society for colposcopy and cervical pathology,
and American society for clinical pathology screening guidelines for the prevention and early detection of cervical cancer.
CA Cancer J Clin 2012 May-Jun;62(3):147e72.
[68] International Collaboration of Epidemiological Studies of Cervical Cancer. Cervical carcinoma and reproductive factors:
collaborative reanalysis of individual data on 16,563 women with cervical carcinoma and 33,542 women without cervical
carcinoma from 25 epidemiological studies. Int J Cancer 2006 Sep 1;119(5):1108e24.
[69] International Collaboration of Epidemiological Studies of Cervical Cancer, Appleby P, Beral V, Berrington de Gonz
alez A,
et al. Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with
cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet 2007 Nov 10;
370(9599):1609e21.
X, Bosch FX, Mun
~ oz N. Environmental co-factors in HPV carcinogenesis. Virus Res 2002 Nov;89(2):191e9.
[70] Castellsague
[71] Roura E, Travier N, Waterboer T, et al. The influence of hormonal factors on the risk of developing cervical cancer and precancer: results from the EPIC cohort. PLoS One 2016;11(1):e0147029.
[72] Safaeian M, Quint K, Schiffman M, et al. Chlamydia trachomatis and risk of prevalent and incident cervical premalignancy
in a population-based cohort. J Natl Cancer Inst 2010 Dec 1;102(23):1794e804.
[73] H Zhu, Shen Z, Luo H, et al. Chlamydia trachomatis infection-associated risk of cervical cancer: a meta-analysis. Med (Balt)
2016 Mar;95(13), e3077.
[74] Kelly Helen, Mayaud Philippe, de Sanjose Silvia. Concomitant infection of HIV and HPV: what are the consequences? Curr
Obstet Gynecol Rep 2015;4(4):213e9.
[75] Kelly H, Weiss HA, Benavente Y. Antiretroviral therapy, high-risk human papillomavirus, cervical intraepithelial neoplasia
and invasive cervical cancer: a systematic review and meta-analysis. The Lancet HIV 2017 (accepted).
Descargar