Open access peer-reviewed chapter

Serological Biomarkers for the Prediction and Detection of Human Papillomavirus Associated Cancers

Written By

Lourdes Gutierrez-Xicotencatl, Azucena Salazar-Piña, Lilia Chihu- Amparan and Adolfo Pedroza-Saavedra

Submitted: 12 January 2018 Reviewed: 09 February 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.75143

From the Edited Volume

Immunoregulatory Aspects of Immunotherapy

Edited by Seyyed Shamsadin Athari

Chapter metrics overview

1,367 Chapter Downloads

View Full Metrics

Abstract

High-risk human papillomavirus (HPV) types are not only associated to uterine cervical cancer, but also to a fraction of cancers of the vulva, vagina, penis, anus, head and neck. An HPV infection generates a protective humoral immune response against the capsid proteins L1 and L2; however, an immune response against other HPV early proteins is also generated. This latter is not a protective response, but those antibodies can be useful as biomarkers of the status of the infection and/or the stage of the cancer lesion. Until now, there are no conclusive results regarding the use of anti-HPV antibodies as biomarkers in diagnosis. In this review, we hereby summarized the actual panorama of the humoral immune response against different HPV early proteins during the development of the disease as possible biomarkers for the prediction and detection of HPV-associated cancers.

Keywords

  • serological biomarkers
  • human papillomavirus
  • humoral immune response
  • HPV-associated cancers
  • cancer diagnostic

1. Introduction

Prevention of cervical cancer (CC) and other related human papillomavirus (HPV) diseases constitutes a public health priority worldwide [1]. Primary prevention has been achieved through the introduction of the prophylactic HPV vaccines, but the target groups are only adolescent girls and young women (up to 25 years old) [2]. Secondary prevention has been implemented through screening methods to prevent precancerous lesions from progressing to cancer [3]. The CC prevention programs in the world are based on cervical cytology and colposcopy to detect precancerous lesions, which have helped to reduce significantly the incidence of this illness in countries with well-organized programs and good coverage of the target population, but this is not the case in developing countries [4, 5]. The main problems are the lack of qualified personnel, the poor quality of the screening tests, lack of follow-up colposcopy and treatment, and over-saturation of the health system facilities, estimating that less than 20% of the CC cases are detected opportunely in these countries [6]. The HPV has been the target for the new molecular diagnostic technologies to detect high-risk (HR)-HPV DNA in cervical cells, but these tests have not been sufficient to discriminate women with precancerous lesions in progression to cancer, from those that eliminate the infection and the lesion. Thus, the increasing incidence of HPV-related cancers worldwide, the inefficacy of the cancer prevention programs in developing countries, and the lack of efficient HPV diagnostic tests, make this a priority health problem worldwide [7].

For this reason, it is important to develop new screening methods, which should achieve high sensitivity, specificity, and should be inexpensive for developing countries. These new diagnostic methods could be used in triage with the cytology or HPV-screening tests to detect opportunely women at risk to develop CC. In addition, it would be important to develop new technologies and to identify new biomarkers that allow the early detection of other HPV-related cancers. In this sense, antibodies against HPV antigens have become the new biomarkers that can be used to detect persistent HPV infection that in combination with other molecular tests could be useful for early detection of HPV-associated cancers.

Advertisement

2. Differential expression of human papillomavirus proteins during the viral cycle

The HPV is a non-enveloped icosahedral virus of approximately 50 nm in diameter that contains a double-stranded circular DNA genome of around 8 Kb, which is divided into three regions: the long control region (LCR) that regulates the viral DNA transcription and replication; the early region (E1, E2, E4, E5, E6, and E7 genes) that controls the transcription and replication of the viral genome as well as to control the carcinogenesis; the late region (L1 and L2 genes) that contains the genes that expresses the viral capsid proteins [8, 9]. Differential expression of HPV proteins during the viral cycle is important for virus replication and evasion of the host immune response. In new infected cells, the HPV replication starts with the expression of low levels of HPV E6 and E7 viral oncoproteins that generates cellular proliferation and genome instability [10, 11]. First, the major viral oncoprotein E7 binds to the retinoblastoma tumor suppressor protein (pRb), which allows the cell to continue into the cell cycle [12]. Simultaneously, the E6 oncoprotein is expressed, and binds to the cellular ubiquitin ligase E6AP, which in turn results in degradation of the p53 protein, a transcription factor for cell cycle arrest, and in extreme situations, for induction of apoptosis [12]. As the infected basal cells migrate to the upper layers and differentiate, viral DNA replication is favored by the binding of E1 and E2 proteins to the LCR to regulate viral proteins expression [13, 14]. Once the viral DNA replication ends, the E2 protein represses the expression of the E6 and E7 oncogenes to allow the continuation of the viral cycle [15]. In the middle of this process, the E5 oncoprotein is expressed to maintain for a longer time the S-phase of the cell cycle and to delay the differentiation process to allow the complete expression of the viral proteins and the viral DNA replication [16, 17]. In the upper layers, E4 protein interacts with the cytoskeleton collapsing the cytokeratin filaments and enhancing the liberation of viral particles [18, 19], and it is also involved in the viral DNA replication [20]. Finally, the two viral capsid proteins L1 and L2 are expressed in terminally differentiated cells, once the replication of the viral genome has been completed, and ending with the release of the viral particles [21, 22, 23].

On the other hand, during a HPV persistent infection, there is a gradual loss of regulation of the E6 and E7 expression genes, which allows the development of early cervical lesions (CIN 1–3; cervical intraepithelial neoplasia grade 1–3) [24]. However, more than 70% of the CIN lesions are eliminated by the immune system. Progression to CC occurs due to an over-expression of E6/E7 oncoproteins, as a result of integration of the viral genome, which leads to the loss of the regulator E2 gene [15]. This is an important event in the carcinogenesis of CC, as the over-expression of E6 and E7 oncoproteins generates cellular immortalization [25], stop cellular differentiation that generates dysplasia, the cells became anergic for TNF-α (tumor necrosis factor) and TGF-β (transforming growth factor) [25, 26], and chromosomal rearrangements could occur, as has been observed with the c-myc gene [27].

During a normal viral cycle, all the early HPV proteins carry out their functions inside the cells, and the viral antigens are poorly exposed to the immune system of the host. However, persistence of HPV infection allows the production of antibodies. Although the antibodies generated against the early HPV proteins are not of neutralizing type, these are suitable for their study as possible biomarkers, which recently is under investigation.

Advertisement

3. Cancers associated with HPV infection

Among all human cancers, 15% are caused by viral infections. HPV infection is recognized as one of the major causes of infection-related cancers in both men and women. Generally, HPV has been associated with more than 90% of anal and cervical cancers, about 70% of vaginal and vulvar cancers, 70% of oropharyngeal cancers, and more than 60% of penile cancers [28].

The HPV is the most common sexually transmitted virus and the HR-HPV types 16 and 18 are more prevalent in CC (approximately 70%) [28]. This type of cancer has been a major public health problem among adult women in developing countries. The last worldwide report for CC identified more than 440,000 incident cases and over 230,000 deaths due to this disease [1]. HR-HPV infection is necessary but not sufficient to cause this cancer, which develops over a long period of time through precursor lesions at the squamocolumnar junction cells near the transformation zone [29]. These cells shown to be multipotent residual embryonic cells have also been identified at the anorectal junction similar to the cervix [29]. Although, the cellular origin and the HPV-DNA prevalence are similar in the anus and in the cervix, the incidence ratio of cervical/anal cancer is quite different (17:1) [30]. The majority of low-grade squamous intraepithelial lesions (LSIL) do not progress to high-grade lesions (HSILs) or carcinoma, which suggests that the HPV infection alone is not sufficient to generate cancer, as other cofactors such as immune deficiency, host genetic factors, among others are involved [30].

In anal cancer (AC), the HPV infection is detected in 80–90% of the cases, and HPV16 is the predominant type (80%) [31, 32] with a frequency higher than in other anatomical sites [32, 33]. This high frequency of HPV16 may reflect a differential tropism of this type that leads to malignant transformation in the anal mucosa. The prevalence of HPV in AC differs by geographic region, with the highest prevalence in North America and Europe and the lowest in Africa [31]. From the gastrointestinal tract malignancies, the prevalence of AC is around 2–3%, with 27,000 new cases reported worldwide in 2008 [31].

Vulvar cancer (VC) is originated from a precursor in intraepithelial lesions named vulvar intraepithelial neoplasia (VIN) and this type of cancer accounts for >90% of the malignant tumors in the vulva [34]. Recently, there is increasing evidence that suggests two different etiopathogenic pathways for the development of VC, one that is associated to HR-HPV and the second that is HPV independent. The prevalence of HPV-DNA in VIN lesions varied from 52 to 100%, but it is over 90% in VC [32, 34]. Over the last decade, the incidence of HPV-associated VC has increased mainly in young women, probably because of high-risk sexual behavior and a better recognition of these lesions due to HPV-DNA detection [32, 35].

On the other hand, penile cancer (PC) has been considered a relatively rare malignancy in the western world, although recent reports indicate an increase in incidence rates in developing countries (from 0.8 to 1.4/100,000) [36]. The etiology of PC is multifactorial with multiple established risk factors including infection with HPV. Epidemiological studies found that 48% of evaluated PC samples were positive for HPV-DNA and the type 16 or 18 was implicated in approximately 31% of these tumors, with HPV16 being the predominant type [37, 38]. In men, HPV infection can result in a spectrum of genitourinary manifestations that can cause genital warts, penile intraepithelial neoplasia (PIN), and PC. However, most HPV infections are asymptomatic, and up to 70% are cleared within 1 year [38].

The final group of the HPV-associated cancers is the one related to the head and neck cancer (HNC) that is the fifth most common cancer in the world [1]. Every year, there are more than 640,000 cases of this cancer reported and it causes over 350,000 deaths [1]. Squamous cell carcinoma is the most frequent type of neoplasia lesions affecting the head and neck area [39]. The laryngeal cancer (LC) is the most common among head and neck neoplasia and it accounts for about 60% of all cancers in the head and neck area [39]. LC may result from late complications of squamous cell papilloma (SCP), although most of those malignant changes develop without papillomas. Generally, squamous laryngeal cancer development begins with dysplastic changes within the epithelium of mucosa membrane lining the organ, this is followed by an intraepithelial neoplasia and finally the development of the pre-invasive cancer (carcinoma in situ) [39, 40]. However, HPV involvement in LC etiology has not yet been fully evaluated [41].

Within this group of cancers, oropharyngeal carcinomas (OPC) are the most dependent on HPV. The incidence of HPV-positive OPC has been markedly increasing in North America and Europe, with a higher rate in men than in women in North America [30], and HPV16 has been detected in the majority of these cancer cases [40]. Until now, little is known about the transmission and immunogenicity of HR-HPVs within the oropharynx. There is a strong association with having performed oral sex and the number of lifetime partners [42], suggesting that initial infection of HPV in the oropharynx is related to high-risk sexual activity. HPV nucleic acid examination in rinse and gargle samples showed a prevalence of 4.7% of HR-HPV infection in the general population among the ages 45–65 years old. However, it is still unclear the implications of the viral infection in the development of OPC [42].

Moreover, esophageal cancer (EC) is the leading cause of cancer mortality worldwide, with approximately 500,000 incident cases and more than 400,000 deaths each year [1]. There are two types of EC; the most common is the squamous cell carcinoma (ESCC), which is highly prevalent in Eastern countries and in developing countries. The second type is the adenocarcinoma (EAC), which is associated with Barrett’s esophagus, and its incidence has raised by 5–10% each year, in developed (Western) countries [43].

Advertisement

4. Immune response to the HPV infection

Mucosal HPV infections frequently arise in the anogenital tract and in the head and neck region, and these sites of infection have high threshold of immune tolerance [44].

The infection and replication of HPV is restricted to differentiating epithelial cells, where there is a limiting presentation of viral antigens to the host immune system. As a result, there is a low but detectable humoral immune response in most infected individuals [45]. HR-HPV types 16 and 18 mainly induce persistent infections without frequent serious complications for the host; they are also highly successful in releasing viral particles transmissible to others [46]. This virus takes the host to a point of balance where the infection does not represent a serious drawback, and viral replication is not limited by the host immune response [46], because the virus does not have a blood-borne phase or viremia. The HPV infection does not induce necrosis, cytolysis, or inflammation, and as a result, there is little or no release of pro-inflammatory cytokines in the local environment [47]. The HPV viral cycle occurs in cells that are destined for death by anoikis (detachment), and because of this, there are no danger signals to alert the immune system to generate an efficient response to eliminate the infection [48].

It is well documented that more than 80% of the genital lesions caused by HPV infections are cleared as a result of a successful cell-mediated immune response, during which cells of the innate immune system such as keratinocytes, dendritic cells (DCs), Langerhans cells (LgCs), macrophages, natural killer (NK), and NKT cells, may play an important role in clearing the infection by promoting a pro-inflammatory process [49]. In the female genital track, the natural host of the HPV infection, there are keratinocytes that could act as immune sentinels, as it has been shown in skin [50]. These cells express Toll-like receptors (TLRs, belonging to the pathogen recognition receptors (PRR) family) on the cell surface (TLR-1, -2, -4, -5 and -6) and in the endosomes (TLR-3 and -9). Specifically, TLR-9 is activated by unmethylated double-stranded CpG-rich DNA [51], allowing the secretion of interferons (IFNs) to activate the NK cells [52], which in turn kill the HPV-infected cells [53]. However, if the HPV infection becomes persistent, there is a downregulation of the innate immune response, which facilitates the virus to escape from the immune system. This mechanism could be through the downregulation of the IFN response by the oncoproteins E6 and E7 that interfere with different molecules involved in the signal transduction pathways of these cytokines [54].

Conversely, the LgCs and DCs (antigen-presenting cells) initiate the adaptive immune response to eliminate the HPV infection, through antigen-specific presentation to B and T cells in the lymph node. In this process, there is a generation of a Th1-type microenvironment by secretion of pro-inflammatory cytokines, which helps to activate Tc cells (directed against the early HPV proteins E2, E6 and E7) to kill the infected cells [55]. This immunological response is complemented by the generation of neutralizing antibodies against the L1 capsid protein to further inhibit the spread of the viral infection, but the virus uses several strategies to evade this adaptive immune response [56]. Recently, it was demonstrated that E2, E6, and E7 proteins upregulate the expression of IL-10 and TGF-β by interacting with their promoters, which allows an immunosuppressive environment [56]. Additionally, the E5 oncoprotein regulates the antigen presentation to the Tc cells by retaining the MHC-I in the Golgi apparatus, and preventing the MHC-I complex transportation to the cell membrane [57]. Besides, the antigen presentation to the Th cells is also regulated by E5, since this protein prevents the acidification of the endosomes, where the MHC-II restricted antigen is processed [58]. At the end, the immune system fails to clear the HPV infection as a result of an imbalance between Th1 (pro-inflammatory) and Th2 (anti-inflammatory) cytokines, which allows a persistent infection with a high risk for the development of CC [56, 59].

Finally, the humoral immune response against the HPV infection is driven through the activation of the B-cell receptor by recognition of HPV antigens and stimulation by the CD40 Th cells receptor that allows the differentiation into plasma cells to produce antibodies against HPV proteins [48]. In this way, a differential antibody response against different HPV antigens (E1, E2, E4, E5, E6, E7, L1, and L2) is generated and detected in the sera of HPV-infected women [60], and specifically, anti-E7 antibodies have been identified and associated to CC, and suggested as a possible markers for late stages of this disease [61, 62, 63, 64, 65].

Advertisement

5. Humoral immune response against human papillomavirus antigens

For several decades, the humoral immune response against HPV proteins has been used to study the viral cycle, and more recently as markers of HPV-associated cancers at different anatomical sites [66]. In this regard, some studies showed that seroconversion and the presence of anti-HPV antibodies were associated to the occurrence of precancerous lesions at different anogenital sites such as CC, AC, and from other sites like oral cancer (OC) [66, 67].

The presence of anti-HPV antibodies has been investigated through several epidemiological studies in several populations with different exposures to the virus and found a great variety in the prevalence and kinetics of these antibodies. The variations observed in the antibody response could depend on the population type, anatomical site of infection, viral antigens present, among others, but also the detection method may influence the antibody results observed (Tables 1 and 2). The immune response to the HPV proteome (or lack thereof) may provide some biological clues required to answer some of these questions. The HPV oncoproteins E6 and E7 are early viral proteins that drive neoplastic transformation, are reliable indicators of an HPV-associated neoplasia, and can lead to detectable serum antibodies prior to and at time of diagnosis, as well as post treatment [68]. To evaluate the serum antibodies against HPV antigens, generated during infection, precancerous lesions and cancer, several laboratory technics have been used, such as the ELISA (enzyme linked immunosorbent assay), western blot, radioimmunoassay (RIA) and more recently, Luminex multiplex. These methods use different antigens such as L1 virus like particles (VLPs) from different HPV types; synthetic peptides from L1, L2, E2, E4, and E7, bacterial recombinant proteins, or in vitro-translated viral antigens [7, 69].

Cancer type Method Population Serum antibodies (%) Ref.
E1 E2 E4 E6 E7 L1
Anal ELISA AC cases 70 45 50 [78]
ELISA Hospital controls and AC cases 25–52 [71, 72]
Multiplex GST Anogenital cancer cases 21 13 29 29 33 54 [79]
Penile ELISA VLPs Hospital controls and PC cases 63 [84]
ELISA L1 Hospital controls and PC cases 11 24–38 [66, 71, 72]
Multiplex GST Anogenital cancer cases 8 13 17 8 13 17 [79]
Vulvar ELISA VLPs Hospital controls and VC cases 27 [84]
ELISA L1 Hospital controls and VC cases 43 [72]
Multiplex GST Anogenital cancer cases 5 8 16 2 8 27 [79]
Vaginal ELISA VLPs Hospital controls and cancer cases 27 [84]
ELISA L1 Healthy controls and cancer cases 44 [72]
ELISA Hospital controls and cancer cases 0 26 [66]
Multiplex GST Anogenital cancer cases 0 0 0 8 0 25 [79]
Cervical cancer Multiplex GST Healthy controls and CC 10 12 17 32–37 28–42 19–44 [70, 74, 75, 88]
Western blot Healthy controls and CC 60 75 [62]
ELISA Healthy controls and CC 19–54 13–53 28–68 [63, 70, 76, 77, 81, 82, 83, 86, 87]
RIA Healthy controls and CC 50–51 33–39 56 [76, 80]
Luminex multiplex Healthy controls and CC 11–44 14–61 21–35 [70, 73, 89]
Slot blot Healthy controls and CC 73 80 40 [69]

Table 1.

Antibodies against HPV16 antigens in different types of anogenital cancers.

AC, anal cancer; PC, penile cancer; VC, vulvar cancer; CC, cervical cancer.

Cancer type Method Population Serum antibodies (%) Ref.
E1 E2 E4 E6 E7 L1
Tongue ELISA Healthy controls and TC 25 4–21 [66, 84]
Oral cavity ELISA Hospital controls and OC 12–25 [84]
ELISA GST Hospital controls and OC 9 10 [100]
Multiplex GST Hospital controls and OC 8 6 8 1 6–14 5–23 [101, 102]
Oropharyngeal ELISA GST Healthy controls and OPC 63–74 36–72 24–42 42–90 12–80 6–33 [66, 100, 103, 104, 105]
Luminex GST Healthy controls OPC and Partners 73 80–83 43 50 63 23 [106]
Multiplex GST Healthy and Hospital controls and ADC 16–21 24–25 11–13 30–35 20–25 14–42 [101, 102]
OPC and Non-OPC 37–70 45–77 34–45 61–85 47–72 55–61 [107, 108]
OPC 64 84 36 90 71 70 [91]
Laryngeal ELISA Healthy controls and LC 12 20 [66]
Multiplex GST Hospital controls and ADC 9 5 12 1–2 6–12 3–24 [101, 102]
Esophageal ELISA Hospital controls and EC cases 17 0 8–31 [66, 84, 109, 110, 111]
Multiplex GST Hospital controls and EC cases 6 5 8 0.3–3 5–9 2–23 [101, 102]

Table 2.

Antibodies against HPV16 antigens in different types of head and neck cancers.

TC, tongue cancer; OC, oral cancer; ADC, aero-digestive cancer; OPC, oropharyngeal cancer; LC, laryngeal cancer; EC, esophageal cancer.

A large number of epidemiological and clinical studies have been carried out to search for the presence of antibodies against HPV proteins and to identify associations of these serological markers with different types of anogenital cancers. In this sense, antibodies against proteins L1, E4, E6, and E7 from HPV16 are the most explored and are most frequently associated with different HPV-associated cancers (Table 1) [62, 66, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89].

Until now, the anti-L1 antibodies are more commonly associated to anal, penile, vulvar, vaginal, and cervical cancer, but the results are contradictory. Some researchers found similar prevalence of anti-L1 antibodies in controls and in penile, vulvar, and vaginal cancer cases (17-38%) [66, 79], while others have shown higher prevalence of anti-L1 antibodies in AC (~54%) and CC (~68%) than in controls [79, 82]. These variations could be the result of different sensitivities in the tests used, as well as the purity and the origin of the viral antigens [55, 90]. Even though there are anti-L1 antibodies present in different types of cancer, these antibodies do not differentiate the anatomical site of the HPV infection or the lesion site. Still some differences have been identified, as anti-E7 antibodies are good markers for CC and anti-E6 antibodies for OC [62, 91]. In this way, several studies have been carried out and showed that antibodies against E7 have been commonly associated with AC and CC, with prevalence that goes from 45% in the anus [78] and up to 75% in the cervix [62, 69], while in the penis, vulva, and vagina, the antibody prevalence was under 15% [79]. In contrast, although serological antibodies against E6 prevalence were high in CC patients by using different tests (from 37 to 44%) [70, 86], the association of anti-E6 antibodies with this type of cancer has not been very well defined [70, 74, 75, 88].

One of the most studied HPV proteins is E4, and this is probably due to its abundance (20–30% of total protein in condylomas) and to its differential production along the viral cycle [92]. During HPV DNA replication in low-grade lesions, high expression levels of E4 protein are observed, while in high-grade lesions, this protein is almost absent [24, 92, 93]. As a result of these observations, the E4 protein is proposed as a marker of viral replication [92, 93]. However, the methodology to detect E4 protein relies on biopsy samples, which are difficult to obtain. For that reason, the detection of HPV antibodies has become a more sensitive system to indirectly follow-up the expression of viral proteins. Several epidemiological studies have shown higher prevalence of anti-E4 antibodies is observed in women with premalignant lesions than among CC cases or in the general population [62, 75, 94, 95]. Previously in our laboratory, we showed that anti-E4 antibodies were in low prevalence in healthy women (11%), but the prevalence increased in subjects with CIN1-3 lesions (70%), and slightly decreased in CC (60%), which suggests an early recognition of this protein by the immune system [61, 62], and postulated as early markers of the disease (Table 1).

In contrast, little is known about the presence of anti-E4 antibodies in other anogenital HPV-associated cancers. The study of Kreimer and coworkers [79] carried out in AC, VC, and PC patients identified prevalence of anti-E4 antibodies under 30%, but they did not look for these antibodies in early lesions of these anogenital cancers, where this early HPV marker should be the prevalent, as it was observed for CC (Table 1).

Antibodies against E1 and E2 have been analyzed in anogenital cancers and only in AC, the anti-E2 antibodies have been observed with high prevalence (70%), which is through the use of peptides in ELISA [78]. However, a more recent study using a multiplex assay showed that the prevalence of these anti-E2 antibodies was fewer than 15% [79]. In the case of CC, the observed prevalence of anti-E1 and anti-E2 antibodies was under 15% [75] (Table 1), but the prevalence of anti-E2 in different degrees of cervical lesion by ELISA was high in CIN1-2 lesions (64%), and it decreased with the increasing severity of the cervical lesion (CIN3, 31%) [96, 97]. Overall, these data suggest that anti-E2 antibodies could constitute a good biomarker for CIN lesions, but these need further studies.

The presence of anti-E5 antibodies in cervical lesions has been described in only one report by using a microarray assay, but no associations were identified with any stage of the uterine cervical lesions [98]. It is necessary to characterize better anti-E5 antibody response, with a more sensitive and specific assay, as this could be an interesting serological marker, since by looking for the presence of E5 mRNA, this was associated with low-grade anogenital lesions [99].

Studies of anti-HPV antibodies in other HPV-associated cancers are underway, but the most recently studied are those localized in the head and neck sites, where different antibodies against E1, E2, E4, E5, E6, E7, and L1 viral proteins have been studied to try to identify associations with the presence of some type of cancer lesion in the oral cavity, in the tongue, pharynx, larynx, and even esophagus (Table 2) [66, 84, 91, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111].

The study of anti-HPV antibodies in sites such as the mouth and the esophagus began in the late 1990s, but more recently, the study of these antibodies has focused on lesions caused in the oropharyngeal area. In TC, low prevalence of anti-E7 antibodies (25%) and anti-VLPs (4–21%) antibodies were observed [66], while in OC, the prevalence of anti-E7 was under 15%, but it was very variable for anti-VLPs antibodies (from 5 to 25%), differences that could depend on the methodology of antibody detection used (Table 2) [79, 84, 101].

One of the cancers in which the presence of anti-HPV antibodies has been analyzed in more detail is the OPC, in which ELISA and multiplex assays have been used with different HPV antigens. These studies strongly showed that anti-E6 antibodies are highly prevalent in OPC (from 30 to 85%), but this prevalence increased up to 90% when the OPC cases were HPV16 positives [91, 102, 106]. However, this was not the case for anti-E7 antibodies, where the prevalence varied from 12 up to 80% [66, 103]. Also, some studies measured the antibodies presence before and after cancer treatment, and they showed that seropositivity to E6 and E7 significantly decreased after treatment. However, only anti-E6 antibodies showed an increased risk of disease recurrence, making these anti-E6 antibodies good biomarkers for disease prognostic [100, 103, 104, 105].

The other anti-HPV antibodies with high prevalence in OPC have been those against E1 (~74%) and E2 (~77%) viral proteins [66, 100, 103, 104, 105, 107, 108], but this is not the case for other head and neck cancers, where the highest prevalence of antibodies against these viral antigens was under 10% (Table 2) [101]. Although the E4 protein is proposed as a marker of viral replication, in the case of OPC, the prevalence of anti-E4 antibodies was under 45%, and this was using the ELISA-GST system that is a highly sensitive method (Table 2) [104]. These studies suggest that the same serological markers are not present at the different anatomical site where the HPV-associated cancers appear. These results are very promising in the search for serological biomarkers, which in combination, they generate profiles that could differentiate the anatomical sites where the HPV-associated cancer is present, as it has been the case for the profile anti-E1/E2 + anti-E6 that has been associated to OPC [91], and for CC, the suggested antibody profile is anti-E4 + anti-E7 [69].

There are few studies that have analyzed anti-HPV antibodies in LC and showed low prevalence of the antibodies that varied from 2% for anti-E6 antibodies to a maximum of 24% for anti-L1 antibodies (Table 2) [66, 68, 101, 102]. Similar results have been reported for EC, and the prevalence of anti-HPV antibodies fluctuated from 31% for anti-L1 antibodies and were under 10% for the rest of the anti-HPV antibodies [66, 68, 84, 109, 111]. At this moment, the identification of serological markers for LC and EC are inconclusive, and more studies need to be carried out with more sensitive methodologies such as the slot-blot system, but also a restriction to HPV positive cancer cases should be considered, as a low proportion of these two types of cancers are associated to HPV.

In addition, most of the studies of anti-HPV antibodies in different HPV-associated cancers have been carried out in the late stages of these cancers. It would be of great interest to study early stages of the HPV-associated cancers as some of these anti-HPV antibodies seem to be important in early diagnostic and during follow-up as possible prognostic markers. In the case of CC, there are several studies of anti-HPV antibodies carried out with precancerous uterine cervical lesions, where it has been suggested that anti-E4 antibodies are important markers for CIN1-2 [61, 62], while the profile anti-E4 + anti-E7 is a good marker for CC [69]. In the case of other HPV-associated cancers, there are only few studies that measured anti-HPV antibodies in early stages of the disease, as it is the case of anus and oral cavity lesions, where anti-VLPs antibodies presented the highest prevalence (43 and 30%, respectively) [111, 112, 113, 114, 115]. However, more studies are necessary to characterize the humoral immune response in the different HPV-associated cancers and their related precancerous lesions.

It is important to mention that differences in methodology and concerns about HPV misclassification, aside from the heterogeneous responses in the antibody patterns seen in the various studies, and in the different HPV-associated cancers require further evaluation. Besides, there are other confounding variables such as HPV type, viral load, viral exposure history, host immune system factors, and clinical risk factors. Therefore, prospective evaluations of anti-HPV serum antibodies should be controlled for as many of these factors. In addition, it is likely that an antibody signature consisting of a panel rather than a single antibody may provide the highest yield to be able to differentiate anatomical site, as well as early detection of these HPV-associated cancers.

Advertisement

6. Diagnostic and prognostic tests for HPV-associated cancers

Essentially all cervical cancers, most anal and oropharyngeal cancers, and some vaginal, vulvar, and penile cancers are caused by HR-HPVs, but until now, there are no general guidelines for screening of all these HPV-associated cancers. Recently, the availability of new tests and ongoing research are changing the approach to screening and diagnosis of these types of cancers. However, most of the studies have been carried out in CC.

6.1. Cervical cancer

For CC, there are specific guidelines that have been modified in the last years, which include the introduction of new testing technologies, which have improved the early diagnostic of this disease. The cytology is the primary screening system for precancerous lesions and can be done using Papanicolaou-stained smears (Pap), although this test has a low sensitivity (50–80%) [116, 117], and a high percentage of false positives, due to the fact that inflammatory cytology is considered abnormal [7, 117]. To confirm the presence of a uterine cervical lesion, a colposcopy test is required, which has a high sensitivity (80–95%) and a low specificity (23–63%), because the test becomes positive in the presence of an inflammatory process, and it is not useful to detect early uterine cervical lesions [118].

The introduction of molecular tests to detect DNA from HR-HPVs has shown to be highly sensitive and makes them good screening systems. Recently, there are different HPV molecular technics that are FDA (Food and Drug Administration, USA) approved to use in conjunction with cytology in CC screening programs. Among these tests are the Hybrid Capture® 2 (HC2) (Qiagen, GmbH, Hilden Ger) and Cervista® HR-HPV Test (Hologic, Inc., MA, USA) that amplify the positive hybridization signal and allow the detection of multiple HR-HPV types in one step [119, 120]. The Cervista® HR-HPV and Cervista® HPV types 16/18 tests have shown to be complementary with a 100% of sensitivity for detection of CIN3+ and of 98% for CIN2+ in women with diagnostic of ASC-US (atypical squamous of undetermined significance) by cytology and HR-HPV positive [121], system that has been approved by FDA.

On the other hand, HC2 has been tested at the general population for the detection of HPV worldwide. This system detects 13 HR-HPV types (-16, -18, -31, -33, -35, -39, -45, -51, -52, -56, -58, -59, and -68) and 5 of low risk (LR) (-6, -11, -42, -43, and -44), and it is semi-quantitative (detects up to 1 pg HPV-DNA/ mL). The HC2 was FDA approved to detect women who have been referred to colposcopy with an ambiguous cytology of ASC-US and for the screening of women over 30 years old in conjunction with the cytology test [120]. Several epidemiological studies have shown the high predictive value of the HPV test for early detection of CC. This test is highly sensitive (93–98%) and specific (60–85%) to detect high-grade lesions, which makes the HC2 ideal for screening, and in combination with the cytology test increases the possibility to identified correctly women in risk to develop CC [3, 122, 123].

There are other systems to detect HPV-DNA by using reverse hybridization, some of which are INNO-LiPA® (Line Probe Assay, Innogenetics, Ghent, Belgium), CLART® HPV2 (Genómica, Madrid, Esp), Clinical Arrays® HPV (Genómica, Madrid, Esp) and the Linear Array® (Roche Mol Diagnostics, CA, USA), and this last one detects 37 HR- and LR-HPV types [119, 124]. The difference between these tests is the sensitivity as the LiPA detects from 10 to 100 DNA viral copies. These genotyping methods have a high sensitivity (95%) to detect CIN2+ lesions, making these tests suitable for screening the general population [119].

The detection of the HPV-DNA as indicator of the viral presence does not determine the presence of an active infection, and because of this, a complementary diagnostic test is necessary. Evaluation of viral load has been used as a biomarker of persistent infection, because cytological abnormalities are more frequently observed in CIN2-3 and CC with high viral load [125]. The viral load is determined by Real-time PCR and this system is highly sensitive and specific, and genotyping can be carried out in the same assay. Two diagnostic tests have been developed that use this technology, the Abbott Real-Time HR-HPV (Abbott Mol GmbH & Co. KG, Germany) and the COBAS® 4800 HPV (Roche Mol Diagnostics, CA, USA). These systems detect 14 HR-HPV types in one reaction, and use a colorimetric detection system to differentiate the types 16 and 18; however, the sensitivity of COBAS® 4800 to detect CIN2 is higher (97%) than the one from Abbott (95%) [123, 126], and just recently have been FDA approved.

In the case of CC, the integration of HPV to the cellular genome seems to be an important part of the carcinogenesis, as this generates an abortive infection with high-level production of the E6 and E7 oncoproteins due to the loss of the E2 gene expression. This event causes the high expression of mRNA of the E6 and E7 oncogenes in the upper layer of the stratified epithelium in CIN2-3 and CC [127]. Thus, the E6/E7 mRNAs have been suggested as specific markers for precancerous lesions. In this way, several commercial tests have been developed to detect mRNAs from E6/E7 such as PreTect® HPV Proofer (NorChip AS, Norway) and APTIMA® HPV (Gen-Probe CA, USA). Both tests detect E6/E7 mRNAs by Real-time PCR, but PreTect® detects only 5 HPV types while APTIMA® detects 14 HR-HPVs, and this is carried on samples from liquid-based cytology. The APTIMA® assay has a similar sensitivity (98%), but higher specificity for HR-HPVs than HC2 (90% vs. 85%, respectively) to detect CIN2-3, making these molecules potential markers for the detection of high-grade lesions, but this is still under investigation [119, 128].

For more than 15 years, it was established that the treatment of early lesions (CIN1, ASC-US, and LSIL) was through follow-up patients with cytology and colposcopy for up to 2 years. However, this procedure is costly, and several visits to the medical office are required, and saturation of the colposcopy system, and loss of patients during the follow-up have made unable to give adequate treatment to those women at risk for developing CC. Because of these problems, it has been necessary to look for new biomarkers that allow the early identification of uterine cervical lesions that could progress to CC. From these efforts, some biomarkers have been identified and are described below.

The p16INK4a is an inhibitor of the cell cycle by inhibiting the hyperphosphorylation of pRb, through blocking the activity of the cyclin D-CDK4/6 complex. It has been observed that the inactivation of pRb by E7 results in the over-expression and accumulation of p16INK4a in the cells, making this protein a surrogated marker of E7 expression, which is associated to the CC development [129, 130]. The detection of p16INK4a is carried out in liquid-based cytology samples and in tissue biopsies by immunohistochemistry (IHC). Different epidemiological studies have demonstrated that p16INK4a is a good marker to identify HPV positive women with CIN 2-3 [129, 130] and it is a useful marker to clarify 90% of ambiguous histopathological diagnostics [131, 132]. The disadvantage of this biomarker is that in other non-associated HPV cancers also it is over-expressed [133].

The Ki-67 proliferation nuclear antigen (cell proliferation marker) is localized in the parabasal layer in the normal stratify uterine cervical epithelium, but during the development of CIN lesions, the expression of Ki-67 is extended all along the cervical epithelium. This marker is ideal to identify tumor cells, which correlates with the clinical stage of the lesion and the development of cancer. The detection of this Ki-67 marker is carried out in liquid-based cytology samples by IHC, and it is useful to detect CIN 2-3-associated HPV lesions [131]. Epidemiological studies in Europe and USA showed that Ki-67 can be used in combination with p16INK4a and helps to increase the sensitivity to a similar level as for HPV detection, but research is still under way to better characterize and validate these biomarkers [131, 132, 134].

6.2. Anal cancer

The AC has similar features with CC, and natural history studies showed that high-grade anal intraepithelial neoplasia (AIN) is a precursor to invasive anal cancer. Because of these similarities, several features of the CC screening program have been taken for routine screening to detect precancerous anal lesions. In the general population, the Pap test to screening for precancerous anal lesions is carried out by taking a sample from the anal canal and the lesions are classified by using the Bethesda nomenclature. At the moment, anal cytology seems to be useful for screening of high-risk individuals, including HIV-positive patients [135].

In contrast, HPV testing has limited utility for AC screening because of the high prevalence of the infection and multiple HPV types in the anal canal of women and HIV-infected men. Only when the test is restricted to HPV16/18, the specificity of the test increased (77%), but the sensitivity dropped (62%), making this diagnostic system not suitable to detect individuals at risk to develop AC [136].

In AC, the high-resolution anoscopy is the standard procedure to diagnose this type of cancer, although several biomarkers such as Ki-67, p16INK4a, and others have been investigated to improve the accuracy of histologic diagnosis. Up to date, only the p16INK4a IHC test has been very well documented to increase the predictive positive value of the histopathology test to identify correctly AC cases [137].

In the case of other anogenital HPV-associated cancers such as vulvar, vaginal, or penile, the diagnostic is carried out by identifying a lesion by visual examination from the genital site to the perianal area, and confirming by performing a biopsy. Screening tests are not available and there are no recommended screening methods to detect HPV infections in these types of cancers. From the surrogated markers of HPV infection, p16INK4a was the only marker with sufficient data to support its utility in the evaluation of lower anogenital tract lesions [138].

6.3. Head and neck cancers

HNCs are often detected at late stages, as conventional visual and tactile examination is a way to diagnose this type of cancer. Substantial efforts to develop oral lesion detection systems such as those based on autofluorescence or tissue reflectance (e.g., the Dentlight Oral Exam Light kit, Microlux DL, Orascoptic DK, Sapphire Plus, Trimira Identafi 3000, and ViziLite-Blue and VELscope) have been developed. However, the ability of these tests to discriminate between cancer and benign mucosal lesions is limited and because of this, the OC screening guidelines still do not recommend these diagnostic tests for routine screening of asymptomatic adults for HNCs [139]. The presence of HPV DNA in saliva was thought to be promising for early detection, but the test showed to have low sensitivity and specificity. For this reason, more studies are required to define the populations where the HPV test could have a positive impact and to evaluate the clinical value of this test [140].

On the other hand, the variable prevalence reported for HPV in HNCs could be attributed to the anatomical site where the sample is taken, but approximately 25% of all HNCs are HPV-DNA positive, and type 16 is the most prevalent. The variations in viral prevalence among different studies may be due to a combination of lesions of the different head and neck anatomical subsites, sample sizes, sampling techniques (frozen, formalin-fixed or paraffin-embedded sections, scraping or oral rinses), and the methodologies used to detect HPV. Because of this, rigorous criteria should be considered for the separation of samples from the various anatomical subsites of the head and neck, as well as to increase sensitivity, specificity, accuracy, and reproducibility of the HPV tests for this type of cancer [140, 141].

Recently, it was suggested that HPV-DNA status in HNC should be analyzed together with other specific markers of active infection such as E6/E7 mRNAs transcription or p16INK4a expression, thus to better characterize these types of HPV-associated cancers. In this sense, the presence of E6/E7 mRNAs and p16INK4a expression was detected mostly in OPC [142]. Numerous other markers such as Ki-67, over-expression of epidermal growth factor receptor, p53, and others, have been studied in HNCs, but none of them have been consistently reliable [140, 141].

6.4. Serological biomarkers for the detection of HPV-associated cancers

The diagnostic of HPV infections for the detection of HPV-associated cancers has been difficult as there is no general screening test that alone or in combination with others allows the early identification of the disease. Because of this, it is necessary to look for tests that would be highly sensitive, specific, less invasive, and inexpensive, and that could be implemented at the general population.

It has been very well described that during the viral cycle, there is a sequential expression of the HPV proteins, which has been associated to different infection stages such as replication (associated to E4 protein), transformation (associated to E6/E7 proteins), and past infections (associated to L1 protein). Thus, the organism generates an antibody response against the viral antigens, in the same way as they are expressed during the viral cycle, letting the identification of different infection stages. Therefore, the humoral immune response naturally amplified the reaction against HPV antigens so that this becomes a good source of new biomarkers to detect HPV-associated premalignant lesions at risk of developing cancer. As a result, the presence of antibodies against E4 protein has been related to viral replication, whereas anti-E7 antibodies are considered markers of a current HPV-associated malignancy [7, 63, 143]. In this context, the use of serological markers has been constantly studied to identify patients with different types of cancer associated with HPV. At present, most of the studies have focused mainly on CC and HNCs.

To study these HPV serological biomarkers, different techniques and reagents have been developed; as for instance, recombinant fusion proteins have been used as antigens in Western blot; synthetic peptides with important immunogenic epitopes for B cell are used in ELISA tests and modifications of this technique have been used to increase the specificity and sensitivity of the assay (Tables 1 and 2). Other systems developed to detect HPV antibodies involve the in vitro protein transcription and translation, and this is used for radio-immunoassays and also for a novel slot-blot system [7, 61, 62, 69, 143]. The use of these tests to measure anti-HPV antibodies in different populations have shown the utility of these as biological markers of different types of lesions not only at the uterine cervix, but also at other anatomical sites.

In the uterine cervix, antibodies against HPV16 E6 and E7 were detected late during the development of CC with a new streptavidin-biotin capture ELISA, and were pointed as bad prognosis markers [81]. In a retrospective study, by using an ELISA-GST, the presence of anti-E6 and anti-E7 IgG antibodies were identified between 0.5 and 5 years before CC diagnosis, suggesting the usefulness of these antibodies as disease predictors [144]. More recently, in a study conducted by Salazar-Piña and coworkers, by using a novel slot-blot system, they showed that anti-E4 + anti-E7 antibodies discriminate CC from CIN 2-3 lesions with high sensitivity (80%) and a low false negative rate (20%), which corroborate the usefulness of these antibodies as markers for early detection of CC. In this study, they also observed that anti-E4 antibodies alone could be useful as HPV exposure markers at early stages of the disease [69].

Similar results were also observed with OPC, where a bead-based multiplex serology method (multiplex-GST) was used to detect different anti-HPV antibodies and showed a strong association between HPV16 E6 seropositivity and the disease, which suggests that these antibodies can be predictive markers of the disease as they were present more than 10 years before the cancer diagnosis [101, 102]. The sensitivity and specificity of this multiplex-GST system for anti-E6 antibodies was close to 100%, and because of that these antibodies have been proposed as a tool for diagnosis and prognosis of HPV-OPC [91, 145]. This multiplex-GST system also showed a high sensitivity (82%) and specificity (100%) for anti-E2 antibodies in the diagnosis of HPV-OPC. However, low sensitivity and specificity were observed for anti-E7, anti-E1, anti-E4, and anti-L1 antibodies [145].

All this together suggests that anti-HPV antibodies are promising diagnostic, prognostic, and potentially screening markers of HPV-associated cancers, as the presence of anti-HPV serum antibodies can vary according to the anatomical site where the cancer is generated by the HPV infection, however, more studies of anti-HPV antibodies are needed to validate them as serological markers for HPV-associated cancers.

Advertisement

7. Conclusions

This review of serological biomarkers is not intended to be an exhausted one, but rather to bring together the most important findings in the field and to point out the usefulness of these biomarkers in the diagnostic and prognostic of the different HPV-associated cancers. Numerous methods are being developed to detect HPV and related biomarkers that alone or in combination can be used to improve the positive predictive value of current screening methods, and to be able to identify precancerous lesions with a high risk of progression to cancer.

At present, serological anti-HPV antibodies are promising diagnostic, prognostic, and potentially screening markers of HPV-associated cancers. It is likely that a combination of anti-HPV antibodies will generate profiles that could discriminate precancerous lesions in progression to cancer, and also to differentiate the presence of HPV-associated cancers at different anatomical sites. For instance, it was shown that anti-E4 antibodies are associated to CIN1-2 lesions and that the profile anti-E4 + anti-E7 antibodies are useful for early detection of CC, while the presence of anti-E1/E2 + anti-E6 antibodies are prognostic of OPC. Besides, the immunoglobulin isotype also seems to play an important role in differentiating the site where the HPV infection develops, as it was shown in CC. It is clear that the presence of anti-HPV serum antibodies can vary according to the anatomical site where the cancer is generated by the HPV infection. These results are very promising, however, more studies with larger populations, different anatomical sites, evaluation in precancerous lesions, and the development of new and more sensitive methodologies are required to better characterize the humoral immune response against HPV and to validate these anti-HPV antibodies as serological markers of different HPV-associated cancers.

Advertisement

Acknowledgments

Some of the results reported in this review were carried out at the Department of Epidemiological Interactions, supported by the National Institute of Public Health and funded through grants from CONACYT (No. 290022), PROMEP-PIFI (UAEMOR-CA-26), and PRODEP (511-6/17-7762) UAEMOR-PTC-405.

Advertisement

Conflict of interest

The authors declare no conflict of interest. The founding sponsors had no role in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish.

References

  1. 1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer (Journal International du Cancer). 2015;136(5):E359-E386
  2. 2. Mariani L, Venuti A. HPV vaccine: An overview of immune response, clinical protection, and new approaches for the future. Journal of Translational Medicine. 2010;8:105
  3. 3. Arbyn M, Castellsague X, de Sanjose S, Bruni L, Saraiya M, Bray F, et al. Worldwide burden of cervical cancer in 2008. Annals of Oncology: Official Journal of the European Society for Medical Oncology (ESMO). 2011;22(12):2675-2686
  4. 4. Jin XW, Sikon A, Yen-Lieberman B. Cervical cancer screening: Less testing, smarter testing. Cleveland Clinic Journal of Medicine. 2011;78(11):737-747
  5. 5. Bobdey S, Sathwara J, Jain A, Balasubramaniam G. Burden of cervical cancer and role of screening in India. Indian Journal of Medical and Paediatric Oncology. 2016;37(4):278-285
  6. 6. Flores YN, Bishai DM, Lorincz A, Shah KV, Lazcano-Ponce E, Hernandez M, et al. HPV testing for cervical cancer screening appears more cost-effective than Papanicolau cytology in Mexico. Cancer Causes & Control: CCC. 2011;22(2):261-272
  7. 7. Gutierrez-Xicotencatl L, Plett-Torres T, Madrid-Gonzalez CL, Madrid-Marina V. Molecular diagnosis of human papillomavirus in the development of cervical cancer. Salud Pública de México. 2009;51(Suppl 3):S479-S488
  8. 8. Yugawa T, Kiyono T. Molecular mechanisms of cervical carcinogenesis by high-risk human papillomaviruses: Novel functions of E6 and E7 oncoproteins. Reviews in Medical Virology. 2009;19(2):97-113
  9. 9. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, et al. The biology and life-cycle of human papillomaviruses. Vaccine. 2012;30(Suppl 5):F55-F70
  10. 10. Vande Pol SB, Klingelhutz AJ. Papillomavirus E6 oncoproteins. Virology. 2013;445(1-2):115-137
  11. 11. Roman A, Munger K. The papillomavirus E7 proteins. Virology. 2013;445(1-2):138-168
  12. 12. Narisawa-Saito M, Kiyono T. Basic mechanisms of high-risk human papillomavirus-induced carcinogenesis: Roles of E6 and E7 proteins. Cancer Science. 2007;98(10):1505-1511
  13. 13. Longworth MS, Laimins LA. Pathogenesis of human papillomaviruses in differentiating epithelia. Microbiology and Molecular Biology Reviews. 2004;68(2):362-372
  14. 14. Peh WL, Middleton K, Christensen N, Nicholls P, Egawa K, Sotlar K, et al. Life cycle heterogeneity in animal models of human papillomavirus-associated disease. Journal of Virology. 2002;76(20):10401-10416
  15. 15. Xue Y, Lim D, Zhi L, He P, Abastado JP, Thierry F. Loss of HPV16 E2 protein expression without disruption of the E2 ORF correlates with carcinogenic progression. The Open Virology Journal. 2012;6:163-172
  16. 16. Pedroza-Saavedra A, Lam EW, Esquivel-Guadarrama F, Gutierrez-Xicotencatl L. The human papillomavirus type 16 E5 oncoprotein synergizes with EGF-receptor signaling to enhance cell cycle progression and the down-regulation of p27(Kip1). Virology. 2010;400(1):44-52
  17. 17. Maufort JP, Shai A, Pitot HC, Lambert PF. A role for HPV16 E5 in cervical carcinogenesis. Cancer Research. 2010;70(7):2924-2931
  18. 18. Khan J, Davy CE, McIntosh PB, Jackson DJ, Hinz S, Wang Q, et al. Role of calpain in the formation of human papillomavirus type 16 E1–E4 amyloid fibers and reorganization of the keratin network. Journal of Virology. 2011;85(19):9984-9997
  19. 19. Wilson R, Fehrmann F, Laimins LA. Role of the E1–E4 protein in the differentiation-dependent life cycle of human papillomavirus type 31. Journal of Virology. 2005;79(11):6732-6740
  20. 20. Egawa N, Wang Q, Griffin HM, Murakami I, Jackson D, Mahmood R, et al. HPV16 and 18 genome amplification show different E4-dependence, with 16E4 enhancing E1 nuclear accumulation and replicative efficiency via its cell cycle arrest and kinase activation functions. PLoS Pathogens. 2017;13(3):e1006282
  21. 21. Buck CB, Thompson CD, Pang YY, Lowy DR, Schiller JT. Maturation of papillomavirus capsids. Journal of Virology. 2005;79(5):2839-2846
  22. 22. Tommasino M. The human papillomavirus family and its role in carcinogenesis. Seminars in Cancer Biology. 2014;26:13-21
  23. 23. Graham SV. The human papillomavirus replication cycle, and its links to cancer progression: A comprehensive review. Clinical Science (London, England). 2017;131(17):2201-2221
  24. 24. Middleton K, Peh W, Southern S, Griffin H, Sotlar K, Nakahara T, et al. Organization of human papillomavirus productive cycle during neoplastic progression provides a basis for selection of diagnostic markers. Journal of Virology. 2003;77(19):10186-10201
  25. 25. zur Hausen H. Papillomaviruses and cancer: From basic studies to clinical application. Nature Reviews. Cancer. 2002;2(5):342-350
  26. 26. Syrjanen SM, Syrjanen KJ. New concepts on the role of human papillomavirus in cell cycle regulation. Annals of Medicine. 1999;31(3):175-187
  27. 27. Ocadiz R, Sauceda R, Cruz M, Graef AM, Gariglio P. High correlation between molecular alterations of the c-myc oncogene and carcinoma of the uterine cervix. Cancer Research. 1987;47(15):4173-4177
  28. 28. Forman D, de Martel C, Lacey CJ, Soerjomataram I, et al. Global burden of human papillomavirus and related diseases. Vaccine. 2012;30(Suppl 5):F12-23
  29. 29. Yang EJ, Quick MC, Hanamornroongruang S, Lai K, Doyle LA, McKeon FD, et al. Microanatomy of the cervical and anorectal squamocolumnar junctions: A proposed model for anatomical differences in HPV-related cancer risk. Modern Pathology. 2015;28(7):994-1000
  30. 30. Chaturvedi AK. Beyond cervical cancer: Burden of other HPV-related cancers among men and women. The Journal of Adolescent Health. 2010;46(Suppl 4):S20-S26
  31. 31. Alemany L, Saunier M, Alvarado-Cabrero I, Quiros B, Salmeron J, Shin HR, et al. Human papillomavirus DNA prevalence and type distribution in anal carcinomas worldwide. International Journal of Cancer (Journal International du Cancer). 2015;136(1):98-107
  32. 32. De Vuyst H, Clifford GM, Nascimento MC, Madeleine MM, Franceschi S. Prevalence and type distribution of human papillomavirus in carcinoma and intraepithelial neoplasia of the vulva, vagina and anus: A meta-analysis. International Journal of Cancer (Journal International du Cancer). 2009;124(7):1626-1636
  33. 33. Steinau M, Unger ER, Hernandez BY, Goodman MT, Copeland G, Hopenhayn C, et al. Human papillomavirus prevalence in invasive anal cancers in the United States before vaccine introduction. Journal of Lower Genital Tract Disease. 2013;17(4):397-403
  34. 34. van Seters M, van Beurden M, de Craen AJ. Is the assumed natural history of vulvar intraepithelial neoplasia III based on enough evidence? A systematic review of 3322 published patients. Gynecologic Oncology. 2005;97(2):645-651
  35. 35. Del Pino M, Rodriguez-Carunchio L, Ordi J. Pathways of vulvar intraepithelial neoplasia and squamous cell carcinoma. Histopathology. 2013;62(1):161-175
  36. 36. Kidd LC, Chaing S, Chipollini J, Giuliano AR, Spiess PE, Sharma P. Relationship between human papillomavirus and penile cancer-implications for prevention and treatment. Translational Andrology and Urology. 2017;6(5):791-802
  37. 37. Wiener JS, Effert PJ, Humphrey PA, Yu L, Liu ET, Walther PJ. Prevalence of human papillomavirus types 16 and 18 in squamous-cell carcinoma of the penis: A retrospective analysis of primary and metastatic lesions by differential polymerase chain reaction. International Journal of Cancer (Journal International du Cancer). 1992;50(5):694-701
  38. 38. Diorio GJ, Giuliano AR. The role of human papilloma virus in penile carcinogenesis and preneoplastic lesions: A potential target for vaccination and treatment strategies. The Urologic Clinics of North America. 2016;43(4):419-425
  39. 39. Rodrigo JP, Shah JP, Silver CE, Medina JE, Takes RP, Robbins KT, et al. Management of the clinically negative neck in early-stage head and neck cancers after transoral resection. Head & Neck. 2011;33(8):1210-1219
  40. 40. Syrjanen S. Human papillomavirus (HPV) in head and neck cancer. Journal of Clinical Virology. 2005;32(Suppl 1):S59-S66
  41. 41. Gungor A, Cincik H, Baloglu H, Cekin E, Dogru S, Dursun E. Human papilloma virus prevalence in laryngeal squamous cell carcinoma. The Journal of Laryngology and Otology. 2007;121(8):772-774
  42. 42. D’Souza G, Cullen K, Bowie J, Thorpe R, Fakhry C. Differences in oral sexual behaviors by gender, age, and race explain observed differences in prevalence of oral human papillomavirus infection. PLoS One. 2014;9(1):e86023
  43. 43. Edgren G, Adami HO, Weiderpass E, Nyren O. A global assessment of the oesophageal adenocarcinoma epidemic. Gut. 2013;62(10):1406-1414
  44. 44. Prigge ES, von Knebel Doeberitz M, Reuschenbach M. Clinical relevance and implications of HPV-induced neoplasia in different anatomical locations. Mutation Research, Reviews in Mutation Research. 2017;772:51-66
  45. 45. Morrison BJ, Labo N, Miley WJ, Whitby D. Serodiagnosis for tumor viruses. Seminars in Oncology. 2015;42(2):191-206
  46. 46. Stanley M. Immunobiology of HPV and HPV vaccines. Gynecologic Oncology. 2008;109(Suppl 2):S15-S21
  47. 47. Karim R, Meyers C, Backendorf C, Ludigs K, Offringa R, van Ommen GJ, et al. Human papillomavirus deregulates the response of a cellular network comprising of chemotactic and proinflammatory genes. PLoS One. 2011;6(3):e17848
  48. 48. Stanley M. HPV—Immune response to infection and vaccination. Infectious Agents and Cancer. 2010;5:19
  49. 49. Sasagawa T, Takagi H, Makinoda S. Immune responses against human papillomavirus (HPV) infection and evasion of host defense in cervical cancer. Journal of Infection and Chemotherapy. 2012;18(6):807-815
  50. 50. Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ. Skin immune sentinels in health and disease. Nature Reviews. Immunology. 2009;9(10):679-691
  51. 51. Bauer S, Kirschning CJ, Hacker H, Redecke V, Hausmann S, Akira S, et al. Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(16):9237-9242
  52. 52. Grandvaux N, tenOever BR, Servant MJ, Hiscott J. The interferon antiviral response: From viral invasion to evasion. Current Opinion in Infectious Diseases. 2002;15(3):259-267
  53. 53. Koch J, Steinle A, Watzl C, Mandelboim O. Activating natural cytotoxicity receptors of natural killer cells in cancer and infection. Trends in Immunology. 2013;34(4):182-191
  54. 54. Rincon-Orozco B, Halec G, Rosenberger S, Muschik D, Nindl I, Bachmann A, et al. Epigenetic silencing of interferon-kappa in human papillomavirus type 16-positive cells. Cancer Research. 2009;69(22):8718-8725
  55. 55. Woo YL, van den Hende M, Sterling JC, Coleman N, Crawford RA, Kwappenberg KM, et al. A prospective study on the natural course of low-grade squamous intraepithelial lesions and the presence of HPV16 E2-, E6- and E7-specific T-cell responses. International Journal of Cancer (Journal International du Cancer). 2010;126(1):133-141
  56. 56. Stanley MA, Sterling JC. Host responses to infection with human papillomavirus. Current Problems in Dermatology. 2014;45:58-74
  57. 57. Campo MS, Graham SV, Cortese MS, Ashrafi GH, Araibi EH, Dornan ES, et al. HPV-16 E5 down-regulates expression of surface HLA class I and reduces recognition by CD8 T cells. Virology. 2010;407(1):137-142
  58. 58. Zhang B, Li P, Wang E, Brahmi Z, Dunn KW, Blum JS, et al. The E5 protein of human papillomavirus type 16 perturbs MHC class II antigen maturation in human foreskin keratinocytes treated with interferon-gamma. Virology. 2003;310(1):100-108
  59. 59. de Freitas AC, de Oliveira THA, Barros MR Jr, Venuti A. hrHPV E5 oncoprotein: Immune evasion and related immunotherapies. Journal of Experimental & Clinical Cancer Research. 2017;36(1):71
  60. 60. Stanley MA. Immune responses to human papilloma viruses. The Indian Journal of Medical Research. 2009;130(3):266-276
  61. 61. Vazquez-Corzo S, Trejo-Becerril C, Cruz-Valdez A, Hernandez-Nevarez P, Esquivel-Guadarrama R, Gutierrez-Xicotencatl Mde L. Association between presence of anti-Ras and anti-VPH16 E4/E7 antibodies and cervical intraepithelial lesions. Salud Pública de México. 2003;45(5):335-345
  62. 62. Pedroza-Saavedra A, Cruz A, Esquivel F, De La Torre F, Berumen J, Gariglio P, et al. High prevalence of serum antibodies to Ras and type 16 E4 proteins of human papillomavirus in patients with precancerous lesions of the uterine cervix. Archives of Virology. 2000;145(3):603-623
  63. 63. Meschede W, Zumbach K, Braspenning J, Scheffner M, Benitez-Bribiesca L, Luande J, et al. Antibodies against early proteins of human papillomaviruses as diagnostic markers for invasive cervical cancer. Journal of Clinical Microbiology. 1998;36(2):475-480
  64. 64. Sasagawa T, Inoue M, Tanizawa O, Yutsudo M, Hakura A. Identification of antibodies against human papillomavirus type 16 E6 and E7 proteins in sera of patients with cervical neoplasias. Japanese Journal of Cancer Research: GANN. 1992;83(7):705-713
  65. 65. Nguyen HH, Broker TR, Chow LT, Alvarez RD, Vu HL, Andrasi J, et al. Immune responses to human papillomavirus in genital tract of women with cervical cancer. Gynecologic Oncology. 2005;96(2):452-461
  66. 66. Van Doornum GJ, Korse CM, Buning-Kager JC, Bonfrer JM, Horenblas S, Taal BG, et al. Reactivity to human papillomavirus type 16 L1 virus-like particles in sera from patients with genital cancer and patients with carcinomas at five different extragenital sites. British Journal of Cancer. 2003;88(7):1095-1100
  67. 67. Lopez RV, Levi JE, Eluf-Neto J, Koifman RJ, Koifman S, Curado MP, et al. Human papillomavirus (HPV) 16 and the prognosis of head and neck cancer in a geographical region with a low prevalence of HPV infection. Cancer Causes & Control: CCC. 2014;25(4):461-471
  68. 68. Sacco AG, Cohen EE. Serum antibodies open the door to prediction and prognostication in human papillomavirus-related head and neck cancer. Cancer. 2017;123(22):4310-4313
  69. 69. Salazar-Pina DA, Pedroza-Saavedra A, Cruz-Valdez A, Ortiz-Panozo E, Maldonado-Gama M, Chihu-Amparan L, et al. Validation of serological antibody profiles against human papillomavirus type 16 antigens as markers for early detection of cervical cancer. Medicine (Baltimore). 2016;95(6):e2769
  70. 70. Achour M, Zeghal D, Kochbati L, Kahla S, Zouari F, Maalej M, et al. Antibody response for L1, E6 and E7 HPV 16 and HPV 18 antigens in Tunisian women with cervical cancer and controls. Journal of Immunoassay & Immunochemistry. 2008;29(3):266-280
  71. 71. Bjørge T, Dillner J, Anttila T, Engeland A, Hakulinen T, et al. Prospective seroepidemiological study of role of human papillomavirus in non-cervical anogenital cancers. BMJ. 1997;315(7109):646-649
  72. 72. Carter JJ, Madeleine MM, Shera K, Schwartz SM, Cushing-Haugen KL, Wipf GC, et al. Human papillomavirus 16 and 18 L1 serology compared across anogenital cancer sites. Cancer Research. 2001;61(5):1934-1940
  73. 73. Castellsague X, Pawlita M, Roura E, Margall N, Waterboer T, Bosch FX, et al. Prospective seroepidemiologic study on the role of human papillomavirus and other infections in cervical carcinogenesis: Evidence from the EPIC cohort. International Journal of Cancer (Journal International du Cancer). 2014;135(2):440-452
  74. 74. Castro FA, Dominguez A, Puschel K, Van De Wyngard V, Snijders PJ, Franceschi S, et al. Serological prevalence and persistence of high-risk human papillomavirus infection among women in Santiago, Chile. BMC Infectious Diseases. 2014;14:361
  75. 75. Combes JD, Pawlita M, Waterboer T, Hammouda D, Rajkumar T, Vanhems P, et al. Antibodies against high-risk human papillomavirus proteins as markers for invasive cervical cancer. International Journal of Cancer (Journal International du Cancer). 2014;135(10):2453-2461
  76. 76. Fisher SG, Benitez-Bribiesca L, Nindl I, Stockfleth E, Muller M, Wolf H, et al. The association of human papillomavirus type 16 E6 and E7 antibodies with stage of cervical cancer. Gynecologic Oncology. 1996;61(1):73-78
  77. 77. Heim K, Widschwendter A, Pirschner G, Wieland U, Awerkiew S, Christensen ND, et al. Antibodies to human papillomavirus 16 L1 virus-like particles as an independent prognostic marker in cervical cancer. American Journal of Obstetrics and Gynecology. 2002;186(4):705-711
  78. 78. Heino P, Eklund C, Fredriksson-Shanazarian V, Goldman S, Schiller JT, Dillner J. Association of serum immunoglobulin G antibodies against human papillomavirus type 16 capsids with anal epidermoid carcinoma. Journal of the National Cancer Institute. 1995;87(6):437-440
  79. 79. Kreimer AR, Brennan P, Lang Kuhs KA, Waterboer T, Clifford G, Franceschi S, et al. Human papillomavirus antibodies and future risk of anogenital cancer: A nested case-control study in the European prospective investigation into cancer and nutrition study. Journal of Clinical Oncology. 2015;33(8):877-884
  80. 80. Park JS, Park DC, Kim CJ, Ahn HK, Um SJ, Park SN, et al. HPV-16-related proteins as the serologic markers in cervical neoplasia. Gynecologic Oncology. 1998;69(1):47-55
  81. 81. Ravaggi A, Romani C, Pasinetti B, Tassi RA, Bignotti E, Bandiera E, et al. Correlation between serological immune response analyzed by a new ELISA for HPV-16/18 E7 oncoprotein and clinical characteristics of cervical cancer patients. Archives of Virology. 2006;151(10):1899-1916
  82. 82. Rocha-Zavaleta L, Ambrosio JP, Mora-Garcia Mde L, Cruz-Talonia F, Hernandez-Montes J, Weiss-Steider B, et al. Detection of antibodies against a human papillomavirus (HPV) type 16 peptide that differentiate high-risk from low-risk HPV-associated low-grade squamous intraepithelial lesions. The Journal of General Virology. 2004;85(Pt. 9):2643-2650
  83. 83. Silins I, Avall-Lundqvist E, Tadesse A, Jansen KU, Stendahl U, Lenner P, et al. Evaluation of antibodies to human papillomavirus as prognostic markers in cervical cancer patients. Gynecologic Oncology. 2002;85(2):333-338
  84. 84. Strickler HD, Schiffman MH, Shah KV, Rabkin CS, Schiller JT, Wacholder S, et al. A survey of human papillomavirus 16 antibodies in patients with epithelial cancers. European Journal of Cancer Prevention. 1998;7(4):305-313
  85. 85. Sun Y, Hildesheim A, Brinton LA, Nasca PC, Trimble CL, Kurman RJ, et al. Human papillomavirus-specific serologic response in vulvar neoplasia. Gynecologic Oncology. 1996;63(2):200-203
  86. 86. Sun Y, Eluf-Neto J, Bosch FX, Munoz N, Walboomers JM, Meijer CJ, et al. Serum antibodies to human papillomavirus 16 proteins in women from Brazil with invasive cervical carcinoma. Cancer Epidemiology, Biomarkers & Prevention. 1999;8(10):935-940
  87. 87. Tjiong MY, Zumbach K, Schegget JT, van der Vange N, Out TA, Pawlita M, et al. Antibodies against human papillomavirus type 16 and 18 E6 and E7 proteins in cervicovaginal washings and serum of patients with cervical neoplasia. Viral Immunology. 2001;14(4):415-424
  88. 88. Wilson LE, Pawlita M, Castle PE, Waterboer T, Sahasrabuddhe V, Gravitt PE, et al. Natural immune responses against eight oncogenic human papillomaviruses in the ASCUS-LSIL triage study. International Journal of Cancer (Journal International du Cancer). 2013;133(9):2172-2181
  89. 89. Reuschenbach M, Waterboer T, Wallin KL, Einenkel J, Dillner J, Hamsikova E, et al. Characterization of humoral immune responses against p16, p53, HPV16 E6 and HPV16 E7 in patients with HPV-associated cancers. International Journal of Cancer (Journal International du Cancer). 2008;123(11):2626-2631
  90. 90. Daling JR, Madeleine MM, Schwartz SM, Shera KA, Carter JJ, McKnight B, et al. A population-based study of squamous cell vaginal cancer: HPV and cofactors. Gynecologic Oncology. 2002;84(2):263-270
  91. 91. Lang Kuhs KA, Kreimer AR, Trivedi S, Holzinger D, Pawlita M, Pfeiffer RM, et al. Human papillomavirus 16 E6 antibodies are sensitive for human papillomavirus-driven oropharyngeal cancer and are associated with recurrence. Cancer. 2017;123(22):4382-4390
  92. 92. Doorbar J. The E4 protein; structure, function and patterns of expression. Virology. 2013;445(1-2):80-98
  93. 93. Griffin H, Wu Z, Marnane R, Dewar V, Molijn A, Quint W, et al. E4 antibodies facilitate detection and type-assignment of active HPV infection in cervical disease. PLoS One. 2012;7(12):e49974
  94. 94. Suchankova A, Krchnak V, Vagner J, Hamsikova E, Krcmar M, Ritterova L, et al. Epitope mapping of the human papillomavirus type 16 E4 protein by means of synthetic peptides. The Journal of General Virology. 1992;73(Pt. 2):429-432
  95. 95. Jochmus I, Bouwes Bavinck JN, Gissmann L. Detection of antibodies to the E4 or E7 proteins of human papillomaviruses (HPV) in human sera by western blot analysis: Type-specific reaction of anti-HPV 16 antibodies. Molecular and Cellular Probes. 1992;6(4):319-325
  96. 96. Marais D, Rose RC, Williamson AL. Age distribution of antibodies to human papillomavirus in children, women with cervical intraepithelial neoplasia and blood donors from South Africa. Journal of Medical Virology. 1997;51(2):126-131
  97. 97. Rocha-Zavaleta L, Jordan D, Pepper S, Corbitt G, Clarke F, Maitland NJ, et al. Differences in serological IgA responses to recombinant baculovirus-derived human papillomavirus E2 protein in the natural history of cervical neoplasia. British Journal of Cancer. 1997;75(8):1144-1150
  98. 98. Luevano M, Bernard HU, Barrera-Saldana HA, Trevino V, Garcia-Carranca A, Villa LL, et al. High-throughput profiling of the humoral immune responses against thirteen human papillomavirus types by proteome microarrays. Virology. 2010;405(1):31-40
  99. 99. Lorenzon L, Mazzetta F, Venuti A, Frega A, Torrisi MR, French D. In vivo HPV 16 E5 mRNA: Expression pattern in patients with squamous intra-epithelial lesions of the cervix. Journal of Clinical Virology. 2011;52(2):79-83
  100. 100. Rubenstein LM, Smith EM, Pawlita M, Haugen TH, Hamsikova E, Turek LP. Human papillomavirus serologic follow-up response and relationship to survival in head and neck cancer: A case-comparison study. Infectious Agents and Cancer. 2011;6:9
  101. 101. Anantharaman D, Gheit T, Waterboer T, Abedi-Ardekani B, Carreira C, McKay-Chopin S, et al. Human papillomavirus infections and upper aero-digestive tract cancers: The ARCAGE study. Journal of the National Cancer Institute. 2013;105(8):536-545
  102. 102. Kreimer AR, Johansson M, Waterboer T, Kaaks R, Chang-Claude J, Drogen D, et al. Evaluation of human papillomavirus antibodies and risk of subsequent head and neck cancer. Journal of Clinical Oncology. 2013;31(21):2708-2715
  103. 103. Anderson KS, Gerber JE, D’Souza G, Pai SI, Cheng JN, Alam R, et al. Biologic predictors of serologic responses to HPV in oropharyngeal cancer: The HOTSPOT study. Oral Oncology. 2015;51(8):751-758
  104. 104. Fakhry C, Qualliotine JR, Zhang Z, Agrawal N, Gaykalova DA, Bishop JA, et al. Serum antibodies to HPV16 early proteins warrant investigation as potential biomarkers for risk stratification and recurrence of HPV-associated oropharyngeal cancer. Cancer Prevention Research (Philadelphia, Pa.). 2016;9(2):135-141
  105. 105. Dahlstrom KR, Anderson KS, Field MS, Chowell D, Ning J, Li N, et al. Diagnostic accuracy of serum antibodies to human papillomavirus type 16 early antigens in the detection of human papillomavirus-related oropharyngeal cancer. Cancer. 2017;123(24):4886-4894
  106. 106. Anderson KS, Wong J, D’Souza G, Riemer AB, Lorch J, Haddad R, et al. Serum antibodies to the HPV16 proteome as biomarkers for head and neck cancer. British Journal of Cancer. 2011;104(12):1896-1905
  107. 107. Lang Kuhs KA, Pawlita M, Gibson SP, Schmitt NC, Trivedi S, Argiris A, et al. Characterization of human papillomavirus antibodies in individuals with head and neck cancer. Cancer Epidemiology. 2016;42:46-52
  108. 108. Zhang Y, Waterboer T, Haddad RI, Miles BA, Wentz A, Gross ND, et al. Human papillomavirus (HPV) 16 antibodies at diagnosis of HPV-related oropharyngeal cancer and antibody trajectories after treatment. Oral Oncology. 2017;67:77-82
  109. 109. Han C, Qiao G, Hubbert NL, Li L, Sun C, Wang Y, et al. Serologic association between human papillomavirus type 16 infection and esophageal cancer in Shaanxi Province, China. Journal of the National Cancer Institute. 1996;88(20):1467-1471
  110. 110. Bjorge T, Hakulinen T, Engeland A, Jellum E, Koskela P, Lehtinen M, et al. A prospective, seroepidemiological study of the role of human papillomavirus in esophageal cancer in Norway. Cancer Research. 1997;57(18):3989-3992
  111. 111. Lagergren J, Wang Z, Bergstrom R, Dillner J, Nyren O. Human papillomavirus infection and esophageal cancer: A nationwide seroepidemiologic case-control study in Sweden. Journal of the National Cancer Institute. 1999;91(2):156-162
  112. 112. van Rijn VM, Mooij SH, Mollers M, Snijders PJ, Speksnijder AG, King AJ, et al. Anal, penile, and oral high-risk HPV infections and HPV seropositivity in HIV-positive and HIV-negative men who have sex with men. PLoS One. 2014;9(3):e92208
  113. 113. Lu B, Viscidi RP, Wu Y, Nyitray AG, Villa LL, Lazcano-Ponce E, et al. Seroprevalence of human papillomavirus (HPV) type 6 and 16 vary by anatomic site of HPV infection in men. Cancer Epidemiology, Biomarkers & Prevention. 2012;21(9):1542-1546
  114. 114. Vriend HJ, Bogaards JA, van der Klis FR, Scherpenisse M, Boot HJ, King AJ, et al. Patterns of human papillomavirus DNA and antibody positivity in young males and females, suggesting a site-specific natural course of infection. PLoS One. 2013;8(4):e60696
  115. 115. Beachler DC, Viscidi R, Sugar EA, Minkoff H, Strickler HD, Cranston RD, et al. A longitudinal study of human papillomavirus 16 L1, e6, and e7 seropositivity and oral human papillomavirus 16 infection. Sexually Transmitted Diseases. 2015;42(2):93-97
  116. 116. Hegde D, Shetty H, Shetty PK, Rai S. Diagnostic value of acetic acid comparing with conventional pap smear in the detection of colposcopic biopsy-proved CIN. Journal of Cancer Research and Therapeutics. 2011;7(4):454-458
  117. 117. Albert S, Oguntayo O, Samaila M. Comparative study of visual inspection of the cervix using acetic acid (VIA) and papanicolaou (pap) smears for cervical cancer screening. Ecancermedicalscience. 2012;6:262
  118. 118. Underwood M, Arbyn M, Parry-Smith W, De Bellis-Ayres S, Todd R, Redman CW, et al. Accuracy of colposcopy-directed punch biopsies: A systematic review and meta-analysis. BJOG: An International Journal of Obstetrics & Gynaecology. 2012;119(11):1293-1301
  119. 119. Abreu AL, Souza RP, Gimenes F, Consolaro ME. A review of methods for detect human papillomavirus infection. Virology Journal. 2012;9:262
  120. 120. Poljak M, Cuzick J, Kocjan BJ, Iftner T, Dillner J, Arbyn M. Nucleic acid tests for the detection of alpha human papillomaviruses. Vaccine. 2012;30(Suppl 5):F100-F106
  121. 121. Johnson LR, Starkey CR, Palmer J, Taylor J, Stout S, Holt S, et al. A comparison of two methods to determine the presence of high-risk HPV cervical infections. American Journal of Clinical Pathology. 2008;130(3):401-408
  122. 122. Bruni L, Diaz M, Castellsague X, Ferrer E, Bosch FX, de Sanjose S. Cervical human papillomavirus prevalence in 5 continents: Meta-analysis of 1 million women with normal cytological findings. The Journal of Infectious Diseases. 2010;202(12):1789-1799
  123. 123. Cuzick J, Thomas Cox J, Zhang G, Einstein MH, Stoler M, Trupin S, et al. Human papillomavirus testing for triage of women with low-grade squamous intraepithelial lesions. International Journal of Cancer (Journal International du Cancer). 2013;132(4):959-966
  124. 124. Gravitt PE, Peyton CL, Alessi TQ, Wheeler CM, Coutlee F, Hildesheim A, et al. Improved amplification of genital human papillomaviruses. Journal of Clinical Microbiology. 2000;38(1):357-361
  125. 125. Huang Y, Huang MN, Li N, Li XG, Li N, Wu LY. Association between human papillomavirus DNA load and development of cervical intraepithelial neoplasia and cervical cancer. International Journal of Gynecological Cancer. 2008;18(4):755-760
  126. 126. Szarewski A, Mesher D, Cadman L, Austin J, Ashdown-Barr L, Ho L, et al. Comparison of seven tests for high-grade cervical intraepithelial neoplasia in women with abnormal smears: The predictors 2 study. Journal of Clinical Microbiology. 2012;50(6):1867-1873
  127. 127. Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. Human papillomavirus molecular biology and disease association. Reviews in Medical Virology. 2015;25(Suppl 1):2-23
  128. 128. Stoler MH, Wright TC Jr, Cuzick J, Dockter J, Reid JL, Getman D, et al. APTIMA HPV assay performance in women with atypical squamous cells of undetermined significance cytology results. American Journal of Obstetrics and Gynecology. 2013;208(2):144 e1-144 e8
  129. 129. O’Neill CJ, McCluggage WG. p16 expression in the female genital tract and its value in diagnosis. Advances in Anatomic Pathology. 2006;13(1):8-15
  130. 130. Carozzi F, Gillio-Tos A, Confortini M, Del Mistro A, Sani C, De Marco L, et al. Risk of high-grade cervical intraepithelial neoplasia during follow-up in HPV-positive women according to baseline p16-INK4A results: A prospective analysis of a nested substudy of the NTCC randomised controlled trial. The Lancet Oncology. 2013;14(2):168-176
  131. 131. Petry KU, Schmidt D, Scherbring S, Luyten A, Reinecke-Luthge A, Bergeron C, et al. Triaging pap cytology negative, HPV positive cervical cancer screening results with p16/Ki-67 dual-stained cytology. Gynecologic Oncology. 2011;121(3):505-509
  132. 132. Schmidt D, Bergeron C, Denton KJ, Ridder R, European CCSG. p16/ki-67 dual-stain cytology in the triage of ASCUS and LSIL papanicolaou cytology: Results from the European equivocal or mildly abnormal Papanicolaou cytology study. Cancer Cytopathology. 2011;119(3):158-166
  133. 133. Lv X, Ye G, Zhang X, Huang T. p16 methylation was associated with the development, age, hepatic viruses infection of hepatocellular carcinoma, and p16 expression had a poor survival: A systematic meta-analysis (PRISMA). Medicine (Baltimore). 2017;96(38):e8106
  134. 134. Singh M, Mockler D, Akalin A, Burke S, Shroyer A, Shroyer KR. Immunocytochemical colocalization of P16(INK4a) and Ki-67 predicts CIN2/3 and AIS/adenocarcinoma. Cancer Cytopathology. 2012;120(1):26-34
  135. 135. Scott H, Khoury J, Moore BA, Weissman S. Routine anal cytology screening for anal squamous intraepithelial lesions in an urban HIV clinic. Sexually Transmitted Diseases. 2008;35(2):197-202
  136. 136. Park J, Neuman HB, Weiser MR, Wong WD. Randomized clinical trials in rectal and anal cancers. Surgical Oncology Clinics of North America. 2010;19(1):205-223
  137. 137. Darragh TM, Winkler B. Screening for anal neoplasia: Anal cytology—Sampling, processing and reporting. Sexual Health. 2012;9(6):556-561
  138. 138. Yang EJ, Kong CS, Longacre TA. Vulvar and anal intraepithelial neoplasia: Terminology, diagnosis, and ancillary studies. Advances in Anatomic Pathology. 2017;24(3):136-150
  139. 139. Rethman MP, Carpenter W, Cohen EE, Epstein J, Evans CA, Flaitz CM, et al. Evidence-based clinical recommendations regarding screening for oral squamous cell carcinomas. Texas Dental Journal. 2012;129(5):491-507
  140. 140. Burd EM. Human papillomavirus laboratory testing: The changing paradigm. Clinical Microbiology Reviews. 2016;29(2):291-319
  141. 141. Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. Journal of Clinical Oncology. 2011;29(32):4294-4301
  142. 142. Bishop JA, Ogawa T, Chang X, Illei PB, Gabrielson E, Pai SI, et al. HPV analysis in distinguishing second primary tumors from lung metastases in patients with head and neck squamous cell carcinoma. The American Journal of Surgical Pathology. 2012;36(1):142-148
  143. 143. Gutierrez-Xicotencatl L, Salazar-Pina DA, Pedroza-Saavedra A, Chihu-Amparan L, Rodriguez-Ocampo AN, Maldonado-Gama M, et al. Humoral immune response against human papillomavirus as source of biomarkers for the prediction and detection of cervical cancer. Viral Immunology. 2016;29(2):83-94
  144. 144. Lehtinen M, Pawlita M, Zumbach K, Lie K, Hakama M, Jellum E, et al. Evaluation of antibody response to human papillomavirus early proteins in women in whom cervical cancer developed 1 to 20 years later. American Journal of Obstetrics and Gynecology. 2003;188(1):49-55
  145. 145. Broglie MA, Jochum W, Michel A, Waterboer T, Foerbs D, Schoenegg R, et al. Evaluation of type-specific antibodies to high risk-human papillomavirus (HPV) proteins in patients with oropharyngeal cancer. Oral Oncology. 2017;70:43-50

Written By

Lourdes Gutierrez-Xicotencatl, Azucena Salazar-Piña, Lilia Chihu- Amparan and Adolfo Pedroza-Saavedra

Submitted: 12 January 2018 Reviewed: 09 February 2018 Published: 05 November 2018