Virology & Mycology

Virology & Mycology
Open Access

ISSN: 2161-0517

Review Article - (2015) Volume 4, Issue 2

Human Papilloma Virus (HPV) Infection and Non-Cervical Oncogenic Disease States

Cristina Merkhofer1 and Joel Maslow2*
1Department of Medicine, Division of Infectious Diseases,Morristown Medical Center, Morristown, USA
2Department of Medicine, Simon 4, 100 Madison Ave, Morristown, USA
*Corresponding Author: Joel Maslow, Department of Medicine, Simon 4, 100 Madison Ave, Morristown, New Jersey 07960, USA, Tel: 971-5136, Fax: 290-7565 Email:

Abstract

Human papillomavirus (HPV) infection is causative for cervical cancer and has been implicated in cancers at other sites. We review the English language literature in regard to the epidemiology of HPV infection as a risk for non-cervical cancer with a focus on the molecular evidence to support HPV having an etiologic role at each anatomic site. HPV DNA is detected in and/or serology provides evidence that HPV is associated with 35-50% of penile cancers, 80-95% of anal cancers, and ~35% of oropharyngeal cancers. HPV has also been implicated for cancer of the larynx, esophagus, lung, and urinary bladder with varying levels of linkage. Molecular studies at the respective anatomic sites provide evidence of HPV integration, E6 and E7 oncogene expression coupled with tumor suppressor down-regulation consistent with a role in the pathogenesis of tumor formation. HPV-associated cancer may represent a distinct disease entity relative to non HPV-associated cancers based upon natural history and outcome studies. Thus, the disease burden of HPV associated non-cervical cancers may be significant and justifies a comprehensive investigation of the utility of global prophylactic vaccination strategies.

<

Keywords: Human papilloma virus; Vaccination; Penile cancer; Laryngeal papillomatosis, Anal cancer, Esophageal cancer, Lung cancer, Bladder cancer, P53, P16, E6 Oncogene, E7 Oncogene, Prostate cancer

Introduction

Human papillomavirus (HPV) is the most prevalent sexually-transmitted infection in the United States. Of the over 100 HPV genotypes, approximately 60 are considered pathogenic for the genital tract and are classified by oncogenic potential. High-risk genotypes, including HPV16, 18, and 33, are more commonly associated with cancers, whereas low-risk types such as HPV6 and 11 typically cause genital warts [1,2].

HPV is causative for cervical and vulvar cancer. In contrast, although HPV has been documented in cancers at other anatomic sites [3], there is not universal agreement as to the role of HPV in the pathophysiology of carcinomatous transformation. While evidence is strongly supported for a role of HPV in anal squamous cell cancer and for cancers of the head and neck, HPV as an independent risk factor for cancers at other anatomic sites is less clear. Moreover, some data has suggested that HPV-associated non-cervical tumors may represent distinct clinical entities than non-viral associated tumors [4]. This paper reviews the epidemiology, molecular basis, and clinical outcome for HPV-associated cancer, focusing on sites other than the female genital tract.

Methods

The English language literature was reviewed using Entrez-PubMed with search terms combining HPV, E6, and E7 and each respective cancer. All papers related to each tumor type were reviewed and articles were selected based on a sufficient sample set and methodological controls. When there was content duplication, representative articles were selected that addressed key issues of epidemiology of disease, molecular associations, and genotype specificity

Anal Cancer

Anal cancer presents primarily between the ages of 60 and 70, with an earlier peak among men who have sex with men [5]. While some studies have found a gender bias towards women with a 2-fold increase in incidence than men [6], or predominate in women [7], others studies have found that the rates of anal cancer are relatively equivalent between genders [8]. The incidence of anal cancer ranges from 1.2-1.3 [9] to 2.04-2.06 cases per 100,000 individuals [8]. Squamous cell cancer (SCC) accounts for 80% of cases, with adenocarcinomas, basal cell carcinomas, and melanomas accounting for the remainder [8,10]. Anal SCC develops from precancerous anal intraepithelial neoplasia (AIN), with AIN classification mirroring that for cervical intraepithelial neoplasia (CIN) and based on the extent of epithelial dysplasia and penetration [6].

HPV is detected in greater than 80% of anal SCC [6,10-12], with two studies utilizing high-sensitivity assays demonstrating 90-100% detection [7,13] (Tables 1 and 2). Whereas HPV was absent in control samples of normal epithelial tissue and hemorrhoidal biopsy specimens [12]. Non-SCC tumor types had significantly lower HPV prevalence [10]. Multivariate risk factors for HPV-associated anal cancer include a history of persistent HPV infection, especially persistence of high-risk oncogenic genotypes, infection with multiple HPV genotypes, receptive anal intercourse, presence of anal condylomata, HIV co-infection (with a greater risk for those with a low CD4 count), immunosuppression in general and solid organ transplantation in particular, cigarette smoking, and younger age [5,7,10] (Table 2).

Risk factor Odds Ratio
Penetrative anal intercourse: women 3.87
Penetrative anal intercourse: men 6.8
Any anal sexual contact 2.62
≥ 5 lifetime sexual partners 2.43
Smoking 2.33
Presence of cervical intraepithelial neoplasia 4.2
Immunosuppression: corticosteroids 3.2
Corticosteriod use in men not exclusively heterosexual 5.6
Genital warts 7.4
HIV infection 6.8

Table 1: Independent risk factors for squamous cell cancer of the anus in women and men. Risk factors are modified from Slama et al. [16], Hasegawa et al. [17], and Uronis and Bendell [5]. Additional independent risks have been reported to include anal presence of HPV, especially high-risk HPV types; immunosuppression, especially organ transplantation; and younger age [5]. Receptive anal intercourse was reported as an independent risk for both heterosexual women l [16] and homosexual men [5], data for men who are not exclusively homosexual are similar to that reported for women, whereas data for men who exclusively have sex with men are significantly greater.

  HPV prevalence Histology Primary HPV type Secondary HPV types
Anal 80-100% Squamous cell 16  
Bladder 0-80% Transitional cell 16 6, 11, 18
Esophageal 0-100% Squamous cell 16  
Laryngeal 8-59% Squamous cell 16  
Lung 20-25% Adenocarcinoma 16 6
21-55% Squamous cell 16 18, OHR
Oropharyngeal 20-58% Squamous cell 16 18, OHR
Penile 15-92% Basaloid 16 6, 11, 18, OHR
Prostate 12-75% Adenocarcinoma 16  

Table 2: HPV prevalence, histologic type, and genotype associated with cancer by anatomic site. HPV prevalence is based on composite of epidemiologic studies. Histology type is that primarily associated with HPV positive cancers. For anatomic sites for which HPV 16 represents <90% of HPV genoptypes, secondary HPV genotypes are given; OHR: Other High Risk Genotypes.

For women a systematic review of the literature of anal carcinoma notes a prevalence of high-risk HPV types ranging from 4-85% among HIV negative women and 16-85% among those HIV-positive, although the study did not differentiate histologic type in relation to HPV presence [14]. Veo et al. in a study of 117 women with cervical cancer, cervical sampling detected HPV16 in 66.7% of cases and HPV18 in 7.7% of cases with anal canal samples yielding HPV16 in 59.8% of women, with simultaneous presence of HPV in the cervix and anal canal detected for 53.8% of whom 19.2% had E6 oncogene expression in anal and cervical biopsy samples [15]. Similarly, Slama et al in a study among HIV-negative women detected anal HPV in 42.4% of women with high-grade cervical lesions [16]. While penetrative anal intercourse had the greatest association for HPV detection in anal samples (OR 3.87) on multivariate analysis, any any anal sexual contact including non-penetrating anal contact remained a significant risk (OR 2.62); other independent risk factors included >5 lifetime sexual partners (OR 2.43) and smoking>60 cigarettes per week (OR 2.33) [16]. Among HIV positive women with high CD4 counts and well controlled infection, Hasegawa et al. found that the presence of cervical squamous intraepithelial neoplasia predicted anal intraepithelial neoplasia with an OR of 4.2 [17]. Thus, cervical HPV infection represents a marker for the anal HPV infection in women with a significant proportion of anal samples demonstrating active oncogene expression. Risk factors associated with anal carcinoma are summarized in Table 1 for both men and women.

With respect to genotype, HPV16 is found in 92-100% of HPV-associated anal cancers regardless of histologic type [6,7,10-12], with multiple high risk types commonly detected [6-11], although others have reported that while HPV16 is predominate it is found in a lower percentage of cases [18,19] (Table 2). In contrast, low risk types HPV6 and 11 are associated with AIN I and II [6,10]. A longitudinal study of HPV anal infection among HIV infected men who had sex with men found that while spontaneous lesion regression and HPV clearance was in general uncommon, those with E6-specific CD4+ T-cell responses had a high rate of clearance [20].

Molecular studies have found sequence variation of the HPV E7 oncogene, which controls binding to the retinoblastoma (Rb) tumor suppressor, in anal SCC [12]. Also HPV E6 variants have been shown to correlate with high-grade disease through down-regulation of the p53 tumor suppressor [21]. Immunofluorescence studies have shown strong nuclear expression of the cyclin-dependent kinase inhibitor 2A (CDKN2A or p16) with concomitant loss of Rb and p53 tumor suppressor activity among HPV-positive anal tumors [11]. In HPV-positive tumors, expression of CDKN2A directly correlated with E7 mRNA expression [22].

Numerous groups have investigated the correlation between molecular markers of HPV infection and disease outcome for anal SCC (Table 3).

Tumor type Molecular correlates of disease Outcome in relation to HPV status
Improved outcome Worse outcome No outcome difference
Anal p16INK4a expression [23] [24] [22]
  p16INK4a levels      
  MCM7 expression      
Bladder Clinical association   [52]  
  p53 mutations   [59] [60]
Esophageal Tumor grade [130,134,136] [135,140] [141]
  Telomere length   [147]  
Lung p53 expression [107,108] [96,99]  
Oropharyngeal E7 expression [84,85]    
Penile Survival for HPV presence [32] [27]  
  p53 expression   [49] [50,51]
  p16INK4a expression   [50,51]  
Prostate E7 expression   [68] [71]

Table 3: HPV infection and the risk for cancer progression in relation to molecular markers. For each tumor type, studies showing the respective outcome, either life expectancy or disease progression, for HPV positive tumors versus HPV-negative tumors are provided. Molecular correlates of when studied are cited; otherwise differences are based on clinical observations. For esophageal cancer, each of the molecular markers that have been studied with respect to outcome in relation to HPV infection are considered independently.

Expression of p16 was a marker of increased survival and disease-free progression in one study [23], versus a risk for poor prognosis and occurrence of distant metastases in a second study [24], but was indifferent to outcome in a third study [22]. The association between p16 and disease outcome is further clouded as one group found that high levels of expression correlated with a poor outcome relative to those with absent or low levels of expression [25] whereas a second study found the opposite result that a high level of p16 expression was protective on univariate analysis but had no prognostic significance on multivariate analysis [26]. Expression of minichromosome maintenance complex component 7 (MCM7), whose expression is regulated by the transcriptional regulator E2F, correlated with relapse-free survival following standard treatments [22].

Penile Cancer

Squamous cell cancer (SCC) of the penis is rare, with an incidence in the United States of 0.7-0.8 cases per 100,000 [9], and highest among men in their sixties [27]. Penile cancer has a five-year survival probability of 75-93% in the absence of metastases and provided that surgical resection has been performed [28].Formal epidemiologic studies of HPV infection as a risk for penile cancer are lacking. The prevalence of HPV DNA in penile cancers varies widely between 15 and 55% [27,29-34], but has been detected in 92% of cases of carcinoma-in-situ [30]. HPV prevalence differs by histologic type with basaloid, warty and mixed tumor types having higher rates of HPV detection (45-100% of cases) versus keratinizing and verrucous tumor types (35% or less) [27,29,31,33-35] (Table 3). In fact, a recent comprehensive review of HPV and its role in penile cancer confirmed this histologic differentiation and noted two distinct etiologic pathways of disease: one related to HPV infection and the other with phimosis as the major risk factor for penile cancer [36] (Table 4). Technical factors may affect the detection of HPV, as stored tissue blocks appeared to have lower yields inversely correlating with storage time [27,31].

Anatomic site Risks
Lung Female non-smokers: greater risk for HPV infection Male smokers: greater risk for non-HPV related cancer
Oropharyngeal Anatomic difference versus HPV status Oropharynx, especially base of tongue & tonsils are typically HPV associated Oral cavity, lips, salivary glands are typically non-HPV associated Epidemiologic risks versus HPV status HPV related: sexual factors (number lifetime partner, oral sexual practice) Non-HPV related: alcohol use, smoking
Penis Basaloid histology type: primary risk is HPV infection Keratinizing and verrucous histology type: primary risk is phimosis

Table 4: Tumors with distinct risks based on HPV status. For the tumors listed above, HPV-positive tumors appear to represent distinct clinical entities with separate primary risk factors as delineated. Additionally, for esophageal and lung cancer there is a marked geographic variation in cancer rates with Asians having greater association with HPV infection.

Incident infection and clearance of HPV of the penis has been examined relative to circumcision status in two longitudinal studies with follow-up for 15-17 months [37,38]. Overall, incident infections of HPV did not vary although non-significant differences were noted in specific genotypes in one study [37]. And, while the rate of clearance did not differ between circumcised and uncircumcised men, the time to clearance was significantly shorter among circumcised men: 154 versus 91 days [38]. HIV status was examined with respect to HPV infection types and persistence in one study that examined 144 HPV positive men of whom half were HIV positive with samples taken at baseline and 6 months [39]. Overall, rates of HPV persistence were similar and high for both HIV positive (69.5%) and HPV negative men (66.9%). Notably, HIV infected men were more likely to have persistence with high-risk genotypes and to have multiple HPV genotypes detected.

HPV16 is the most common genotype found in penile cancer, detected in 52-80% of tumors [27,30-34]. In contrast to cervical cancer, HPV18 was no more prevalent than other high-risk genotypes, and there was a higher frequency of low risk genotype HPV6 [27,31,32,34,40] (Table 4). There was no apparent correlation between genotype and histologic subtype [29,33]. However one study from Brazil found that HPV11 was more common in pre-cancerous lesions (80%) with HPV16 and HPV31 and next most common (25% of lesions each) although in cancers, HPV16 still predominated (63% of tumors) followed by HPV11 (34% of tumors) [41].

Genomic profiling of penile carcinomas documented HPV integration at 9 of the 19 chromosomal sites associated with cervical cancer oncogenesis, with predilection for sites associated with chromosomal fragility [42]. Studies have additionally noted HPV E6 oncogene expression associated with p53 ubiquitination and breakdown in tumor tissue [43]. The presence of HPV DNA inversely correlated with p53 staining but directly correlated with mTOR expression, a gene within the AKT pathway of tumorigenesis, which was found to be upregulated in HPV-negative tumors [44]. However, the finding of p53 regulatory differences in HPV related tumors is not universal as others found only that the retinoblastoma protein p16INK4A was associated with HPV positive tumors [45]. Evidence that tumor pathways such as p16INK4a, c-ras and myc are involved in HPV-related penile cancer is lacking [43] and MYC alterations were found at equal prevalence among HPV-positive and negative tumors [42]. Finally, upregulation of cyclooxygenase (COX-2) expression, implicated in the pathogenesis of cancers at other sites [46,47], has been demonstrated in some specimens [48]. Molecular correlates of disease outcome have been found with p53 expression inversely correlating with regional lymph node metastasis and survival [49] while others have found that p16INK4a allelic loss or promotor hypermethylation and not p53 was associated with negative clinical outcomes [50,51]. (Table 3) Observed survival differences has led to the postulate that HPV-positive and negative tumors represent different clinical entities [34,43]. However, studies have disagreed as to whether HPV affects the prognosis of penile cancer. While some have reported a significant survival advantage [32], others found that HPV presence correlated with local and nodal metastases [27], suggesting a poorer outcome.

Bladder Cancer

A 1997 review found that transitional cell cancer of the bladder was infrequently associated with HPV infection, with HPV DNA detected by PCR in approximately 10% of cases overall, although prevalence ranged as high as 80% in some studies [52].More recently, three separate subsequent meta-analyses examined the relationship between HPV infection and bladder cancer. The paper by Wiwanitkit et al. included 5 studies between 1993 and 1996, Jimenez-Pacheco et al. included 21 studies between 1991 and 2010, and Yang examined 52 studies and restricted their analysis to 19 studies over the time range of 1991 to 2010 [53-55]. Each of the three studies found a significant risk for bladder cancer associated with HPV infection with pooled OR ranging between 2.13and 2.84. HPV16, and to a lesser extent HPV18, were the most common HPV types in cancer tissue [52] . Geographic variation was striking as evidenced by two studies from North Africa with HPV DNA was detected in 22 (52.4%) of 43 of fresh bladder cancers from Moroccan patients whereas HPV DNA was not detected in any of 125 formalin fixed bladder cancers among Tunisian patients [56,57]. One study found that HPV risk for bladder cancer was greater than for non-smokers [58]. HPV6 was frequently associated with condylomata of the bladder, with HPV6 and 11 occasionally detected in bladder cancer.

HPV-associated tumors were more aggressive with poorer outcomes [52]. High-grade cancers were associated with p53 mutations that correlated with the presence of high-risk HPV types 16 and 18 [59], although the correlation between p53 status and outcome for HPV-associated bladder cancer was not universal [60] (Table 3).

Prostate Cancer

The role of HPV in the development prostate cancer is one of least well defined of urogenital sites. The epidemiology was explored in two meta-analyses of which one found no increase in risk for HPV infection and prostate cancer, although there was higher prevalence of HIV-16 in cases suggesting a possible relationship [61] whereas a second study found that HPV-16 in contrast to HPV-18 was significantly associated with a risk for prostate cancer [62] (Table 2). Case control studies from South Africa, Sweden, Saudi Arabia, and the US failed to show evidence of a relationship using a combination of serology and DNA detection in prostate cancer tissue [63-66]. In contrast, other studies have reported varying rates of HPV DNA in prostatic cancer tissue ranging from 12.5% in a study from Tehran Iran [67], an Argentinian study with HPV E6 expression detected in 41.5% of samples (17 of 45) of prostate adenocarcinoma, an Italian study that found that 112 (74.7%) of 150 paraffin embedded tumors demonstrated E7 expression [68], and a Greek study of prostate cancer tissue samples in paraffin blocks with HPV DNA in 16% of tumors versus 3.3% of normal tissue from patients without cancers [69], while cases of prostatic hyperplasia were negative [70]. In contrast to tumors at other sites there was no association with p53 codon polymorphism [69]. E7 expression was negatively correlated with survival [68] (Table 3). Finally, Tachezy et al also found no difference in the seroprevalence rates for the E6 and E7 oncoproteins between cases and controls; and the rate of HPV DNA detection in prostate cancer was the same (2%) as that for normal tissue that they concluded showed no pathophysiologic role in tumirogenesis but indicated that the prostate may serve as a reservoir for HPV infection [71]. Thus, the role of HPV infection in prostate cancer remains unproven.

Oropharyngeal Cancer

In the United States, cancer of the oral cavity and pharynx disproportionately affects males, with an incidence of 15.5-15.9 cases per 100,000 men versus 5.7-6.0 cases per 100,000 women [9]. Less than 50% of patients with advanced oral or oropharyngeal SCC survive past 5 years [72].

An etiologic role for HPV in cancer of the head and neck was posited in the early 1980’s, when Syrjänen noted histologic and morphologic characteristics shared with HPV-associated genital tumors [73,74]. Subsequent serologic and PCR-based studies provided further evidence of association, although HPV was detected in only a fraction of tumor samples. More recent work has shown that while HPV is detected in a minority of cancers (~25%), there is a clear niche preference for the oropharyx [75] versus oral cavity [76] (Table 4). Additionally, HPV-related cancers of the oropharynx had a predilection for the base of the tongue (41.1% HPV positive) and tonsils (58.1% HPV positive), but was absent from cancers of the lip or major salivary glands [76] (Table 2).

Among HPV-positive tumors of the head and neck, HPV16 is detected in approximately 90% of oropharyngeal cancers and approximately 75% of oral cavity tumors [4,75,77]. High-risk genotypes HPV31, 33, and 35 were also frequently reported, with numerous cases infected with multiple HPV genotypes [4,75,77]. Gender differences in the incidence of oropharyngeal cancer have been found to parallel

HPV rates by some but not all investigators. Ryerson et al., reported an incidence for oropharyngeal and oral cavity cancers of 4.5 cases per 100,000 individuals for men and 1.7 for women [78]. In contrast, Smith et al. found no gender difference in the prevalence of HPV positive tumors [76]. Ryerson found that between 1998 and 2003, the rate for cancer of the base of the tongue and tonsils, two cancers with a higher likelihood of HPV-relatedness, increased 150-175% and inferred a causal role for HPV [78]. Ernster et al., using United States cancer registry data from the Denver metropolitan area, found a similar rise in oropharyngeal cancers in men from 1980 to 2004, but no change in cancer rates for women [79]. Moreover, while the rates of HPV-negative tumors were relatively static for both genders, HPV-positive tumors increased 10-fold for men and 4.5 fold for women over the 25-year period [79].

There is a growing consensus that SCC of the head and neck represents distinct disease entities dependent upon risk group and HPV presence or absence (Table 4). Multivariate studies find that smoking, alcohol consumption, HPV seropositivity, sexual practices, and a greater number of lifetime oral and vaginal sex partners represent significant risks for cancer of the oral cavity and oropharynx [75,76,80-83]. While alcohol and tobacco exposure represent risks for HPV-negative cancers, sexual risk factors were linked to HPV-positive tumors [4,75,76,80] whereas HPV-positive cancers show no linkage to tobacco or alcohol use on multivariate analysis [75,81]. The 5-year survival for HPV-positive head and neck SCC is 71% versus 49% for HPV-negative tumors [84], with others reporting similar results [85], although, one group reported the survival benefit to be restricted to males [84]. Seroreactivity to the E6 and E7 oncogenes is associated with a 12-fold greater adjusted odds ratio for cancer risk relative to seronegative individuals, whereas seroreactivity to the L1capsid protein was not predictive [81]. Microarray genomic analysis demonstrated that HPV-negative tumors carried alterations in four distinct host cellular chromosomal regions not found in HPV-positive tumors [86]. In contrast, HPV-positive tumors express a variety of cell cycle genes expressed during meiosis, which correlate with E6 and E7 [87]. In cervical cancer, HPV16 E7 oncogene inactivates the pRB retinoblastoma tumor suppressor protein, allowing for an increase in the cyclin-dependent kinase inhibitor p16INK4a [88,89] (Table 3). This finding was replicated in HPV-positive oropharyngeal cancer specimens, and was associated with greater survival and lower recurrence rates versus HPV-negative tumors [85].

Laryngeal Papillomatosis, and Laryngeal Carcinoma

Laryngeal or respiratory papillomatosis (RP) is a benign tumor of the upper respiratory tract with juvenile and adult forms [90]. HPV has been detected in 76.2% of adult cases, typically associated with low-risk genotypes, particularly HPV6 and 11 [90]. While airway obstruction poses the greatest risk, a small number of cases may undergo malignant transformation into SCC [90].

Unlike adult laryngeal papillomatosis, the link between HPV infection and laryngeal carcinoma is less clear. The incidence of laryngeal cancer in the United States was 7.1-7.4 cases per 100,000 men, and 1.5-1.6 cases per 100,000 women [9]. HPV detection in laryngeal carcinomas ranges from 8.0-58.8% [90], with HPV16 as the most common genotype [90] .

Studies have demonstrated a potential genetic linkage to disease progression for RP, as HLA-DRB1*0102 was seen at higher frequency among those with cancer than HLA-DRB1*0301, DQB1*0201 and DQB1*0202 [91]. The differential expression of host genes supports a dominant Th2-like adaptive immune response [92]. Moreover, such patients typically manifest lower interferon-gamma (IFNγ) responses against the E6 oncoprotein of HPV11 [91]. Malignant transformation has been postulated to occur through upregulation of Rac1 by epidermal growth factor that mediates COX-2 expression [93] through phosphatidylinositol-3 kinase [94].

Lung Cancer

HPV had been postulated in the etiology of cancers of the respiratory tract since the mid-1960’s [95]. Causality was inferred in earlier studies that showed a linkage between serology and lung cancer and the finding of HPV in tumors by immunofluorescence for 21.7% of cases [95]. There has been a notable geographic variation with Asian studies finding that 33.3-45% of non-small cell lung cancers were HPV DNA positive [96-98], and a similar prevalence was detected in a Chilean study [99] (Table 4). In contrast, among Western populations studies from Norway, Finland, Italy, and the United States found no correlation with HPV [100-103]. In high-risk regions, HPV DNA is detected in almost 55% of squamous cell lung cancers, versus 20-25% of adenocarcinomas [97,98] (Table 2) and more prevalent in males than females [97,98]. A comprehensive, systematic review of the literature examined the role of HPV in never smokers with lung cancer [104] (Table 4). Authors were contacted to provide information regarding smoking status if not provided in the primary references. As above there were significant geographic differences with the rate highest in Asia and lowest in Western countries, and even regional differences within the same country. None of the studies resolved the question of second-hand smoke as a confounder. HPV16 is the predominant HPV genotype detected, with other high-risk types comprising the bulk of the remainder of pulmonary SCC, whereas HPV6 is predominant in a small number of adenocarcinomas [99] (Table 2). Moreover, tumor cells actively shed virus, although with viral loads many logs lower than for cervical cancer cells [98,99,105].

HPV chromosomal integration has been demonstrated among HPV-positive lung cancers [98,99,105], along with cellular expression of HPV oncogenic proteins [96,97]. HPV infected lung cancer cells demonstrate inhibition of the tumor suppressor p53 and increased IL-6 production, the latter which appears to be induced via expression of the antiapoptotic protein Mcl-1, a finding documented for paraffin embedded tissue samples and then confirmed for the A549 adenocarcinoma cell line [106]. The presence of HPV DNA inversely correlated with p53 expression [96,99], although other studies demonstrated a direct correlation for SCCA of the lung [107,108] (Table 3). Pro-inflammatory cytokine production of IL-8 and IL-17 is induced in E6 transfected adenocarcinoma cell line H1299 inducing metalloproteinases MMP-2 and MMP-9 upregulating angiogenesis [109]. A549 cells and the human non-small cell lung cancer cell line NCI-H460 transfected with the HPV E6 and E7 oncogenes also induce angiogenesis in mouse xenograft models through upregulation of HIF-1 which in turn promotes vascular endothelial growth factor (VEGF) production [110]. In fact, HPV infected tissue samples were more likely to have endothelial growth factor receptor (EGFR) mutations versus uninfected tissue samples from China [111] a finding also noted in a study of Japanese patient samples [112]. The latter study found non-significant trends in EGFR mutation rates for female non-smokers which was postulated to promote lung cancer for that demographic [112].

Esophageal Cancer

In 1982, Syrjänen reported on a case series of invasive esophageal SCC (ESCC) manifesting morphologic changes similar to cervical cancer, suggesting a etiologic role for HPV [113] further supported by detection by immunofluorescence of HPV antigens within esophageal papilloma keratinocyte nuclei [114]. A large number of studies, including a group of recent meta-analyses have concluded an etiologic role although there are significant geographic differences.

A review of the literature in 2002 noted a wide variation in HPV prevalence ranging from 0% to 100% [115] (Table 2). Overall, 22.9% of samples assessed by in-situ hybridization were HPV-positive versus 15.2% tested by PCR [115]. Countries with the highest prevalence of positive cases were from South Africa and Southeast Asia [115]. A case control study from China of 300 cases of ESCC reported that HPV represented a significant risk for disease with an OR of 6.4 and even higher (OR of 10.3) when the analysis is restricted to the presence of high-risk HPV genotypes [116]. Agreement between studies, even from the same regions was not universal as Chinese study found that 56% of cancers harbored HPV [117] whereas a contemporary Chinese study from the same region reported no association [118]. In Africa, South Africa has reported a high rate of HPV in esophageal cancer as a group from Cape Town detected HPV DNA in 46 of 114 tumor samples (46%) versus only 3 of 41 (7%) of normal tissue samples [119,120]. In Europe, a group from Uppsala, Sweden detected HPV DNA by PCR in 16 or 100 (16%) of esophageal tumors [119,120]. Other conditions such as tylosis, Barrett’s esophagus, and basaloid SCC of the esophagus do not appear to harbor HPV [121-123].

A number of recent meta-analyses concluded that HPV does represent a risk for esophageal cancer [124-129] with pooled odds ratios (OR) between 3 and 3.7 although one study calculated a pooled OR of 6.36 for the association of HPV16 to esophageal cancer in China [129]. Differences are considered to relate to methodological differences between studies as well as study design [125].

In contrast, there have been a number of studies that reported a lack of correlation between HPV and esophageal cancer. Methodologic issues were raised as one study detected HPV in both tumor and surrounding normal tissue raising questions about the etiologic role of HPV [130] and a Chinese study that carefully avoided DNA contamination during sample preparation found HPV in only 0.4% tumor samples [118]. And Brazilian study found that while 13% of esophageal tumors were HPV positive, most with HPV16, correlation with molecular markers was negative and concluded that there was no association [131].

There is a growing consensus of a multifactorial pathogenesis for esophageal cancer with environmental, exposure, and infectious etiologies driving tumorigenesis [119,132]. A sero-prevalence Chinese study found that HPV infection increased the risk for ESCC among those with a history of ethanol intake and smoking but did not increase the risk for non-smokers and non-drinkers [133].

Overall, HPV16 has been the most prevalent genotype detected including some investigators who have found only HPV16 [119,120] (Table 2). Other high-risk types, particularly HPV18, have been frequently detected albeit at lower prevalence rates [115,117,134,135]. However, some studies found a substantial number of cases with HPV6, 11, and other low-risk types [117,119,120,130,136]. Although, one group reported that HPV16 and 18 predominated in tumors and precancerous lesions, whereas HPV6 and 11 were most common in normal tissue [137]. In contrast, for esophageal papillomas, HPV6 and 11 predominated while HPV16 was relatively infrequent [138].

Despite the epidemiologic controversies, molecular studies appear to support an etiologic role for HPV in the pathogenesis of esophageal cancer. Greater than 90% of HPV-positive esophageal SCC carry integrated rather than episomal viral forms [135,139], which are associated with excretion of low levels of intact virus [139]. Investigators have reported an inverse correlation between HPV presence and tumor grade [130,134,136] and a Chinese study across different ethnic groups reported a non-significant trend towards higher HPV copy number and tumor grade [140]. Although one case-control study in China found no association between HPV presence and disease progression [141].

Esophageal tumors have increased p16 staining and decreased p53 expression, whereas HPV-negative tumors maintain normal levels of both regulators [142]. Others have found that while p53 expression did not correlate with HPV infection [120,135,142], its homolog p73 was significantly associated [120]. Yang found that esophageal cancer typically harbored mutations at p53 codon 72 [143] and that patients with a single nucleotide polymorphism at rs1042522 conferring either an Arg/Arg or Arg/Pro mutation [144] demonstrated a significant increase in the risk for esophageal cancer but only for those with a history of tobacco and ethanol use. Moreover, However, some reported an association between p53 expression and HPV presence only for HPV16 and not HPV18-associated tumors [134] (Table 3). The role of p16INK4A was similarly at odds between studies. While one study reported that p16 represented a prognostic indicator for overall survival and disease progression, others found no association [135,145]. Other correlates of pathogenesis include the transcription factors AP-1 and Fra-1 that were typically present in HPV-negative tumors but absent in HPV-positive tumors [146]. Finally, telomere length which has been inversely associated with tumorigenesis, was found to be progressively shortened with increasing atypia and further in HPV-associated esophageal cancer tissue that correlated with increasing decline of DNA methylation [147] (Table 3). Moreover, short telomere length represented a further independent risk for HPV related esophageal cancer among those carrying p53 Arg/Arg polymorphisms [148].

Discussion

While HPV is considered causative for cervical cancer, its role at other anatomic sites is less well understood and more controversial. As reviewed here, there is evidence to support a role for HPV in cancers of the anus, oral cavity, tonsils, esophagus, lung, and possibly the bladder. The literature is limited by a relative lack of rigorous case-control or cohort epidemiologic studies at many of the anatomic sites, with the bulk of evidence relying on pathogenic studies that demonstrate the presence or absence of HPV DNA in archived specimens. Detection of DNA from preserved specimens is however subject to contamination during sample preparation unless rigorous procedures are employed.

Cervical cancer represents a single disease state with a clear progression from cellular atypia to invasive cancer. However for cancers of the anus, head and neck, and penis, HPV-associated tumors may be restricted to certain histologic subtypes and ecologic niches. Moreover, studies imply a multifactorial etiology for cancers at many sites, with HPV infection being one of many factors. Tumor grade, survival, and risk factors differ in relation to HPV-presence suggesting that HPV-positive and negative tumors may represent distinct clinical entities.Molecular support for an etiologic role for HPV is evident in the finding of viral integration and E6 and E7 expression in tumor tissue, coupled with differential expression of cell cycle regulators such as p53 and p16. Moreover, the latter has been linked to tumor grade and survival differences, and helps to explain the growing consensus that at many anatomic sites, HPV-caused cancer is a separate clinical condition from non-virally associated disease. The potential number of cancers attributable to HPV would infer a significant case burden. Moreover, HPV-associated cancers of the head and neck cancer, esophagus, and penis are male predominant.

Whether the commercially available bivalent, quadrivalent, and nonovalent HPV prophylactic vaccines can reduce the rates of non-cervical cancer is unknown. Cost-benefit studies that have modeled cervical cancer rates have predicted benefit [149,150]. Studies have also modeled the effect of bivalent, quadrivalent, and nonovalent vaccines for anal and head and neck cancers – all demonstrating a beneficial role of vaccination [18,151-154], although the high effectiveness assumed by these studies for non-cervical cancers has yet to be borne out and still awaits study.

Acknowledgements

The authors gratefully acknowledge the tireless assistance of the library staff at Morristown Medical Center.

References

  1. de Villiers EM, Fauquet C, Broker TR, Bernard HU, zur Hausen H (2004) Classification of papillomaviruses. Virology 324: 17-27.
  2. Muñoz N, Bosch FX, de Sanjosé S, Herrero R, Castellsagué X, et al. (2003) Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348: 518-527.
  3. Shukla S, Bharti AC, Mahata S, Hussain S, Kumar R, et al. (2009) Infection of human papillomaviruses in cancers of different human organ sites. Indian J Med Res 130: 222-233.
  4. Gillison ML, D'Souza G, Westra W, Sugar E, Xiao W, et al. (2008) Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers. J Natl Cancer Inst 100: 407-420.
  5. Uronis HE, Bendell JC (2007) Anal cancer: an overview. Oncologist 12: 524-534.
  6. Varnai AD, Bollmann M, Griefingholt H, Speich N, Schmitt C, et al. (2006) HPV in anal squamous cell carcinoma and anal intraepithelial neoplasia (AIN). Impact of HPV analysis of anal lesions on diagnosis and prognosis. Int J Colorectal Dis 21: 135-142.
  7. Ouhoummane N, Steben M, Coutlée F, Vuong T, Forest P, et al. (2013) Squamous anal cancer: patient characteristics and HPV type distribution. Cancer Epidemiol 37: 807-812.
  8. Johnson LG, Madeleine MM, Newcomer LM, Schwartz SM, Daling JR (2004) Anal cancer incidence and survival: the surveillance, epidemiology, and end results experience, 1973-2000. Cancer 101: 281-288.
  9. U.S. Cancer Statistics Working Group (2004) United States Cancer Statistics: 1999-2004 Incidence and Mortality Web-based Report. In: Group USCSW (ed). U.S. Department of Health and Human Services, Centers for Disease Control and Prevention and National Cancer Institute, Atlanta
  10. Tachezy R, Jirasek T, Salakova M, Ludvikova V, Kubecova M, et al. (2007) Human papillomavirus infection and tumours of the anal canal: correlation of histology, PCR detection in paraffin sections and serology. APMIS 115: 195-203.
  11. Lu DW, E-Mofty SK, Wang HL (2003) Expression of p16, Rb, and p53 proteins in squamous cell carcinomas of the anorectal region harboring human papillomavirus DNA. Mod Pathol 16: 692-699.
  12. Youk EG, Ku JL, Park JG (2001) Detection and typing of human papillomavirus in anal epidermoid carcinomas: sequence variation in the E7 gene of human papillomavirus Type 16. Dis Colon Rectum 44: 236-242.
  13. Baricevic I, He X, Chakrabarty B, Oliver AW, Bailey C, et al. (2015) High-sensitivity human papilloma virus genotyping reveals near universal positivity in anal squamous cell carcinoma: different implications for vaccine prevention and prognosis. Eur J Cancer 51: 776-785.
  14. Stier EA, Sebring MC, Mendez AE, Ba FS, Trimble DD, et al. (2015) Prevalence of anal human papillomavirus infection and anal HPV-related disorders in women: a systematic review. Am J Obstet Gynecol .
  15. Veo CA, Saad SS, Fregnani JH, Scapulatempo-Neto C, Tsunoda AT, et al. (2015) Clinical characteristics of women diagnosed with carcinoma who tested positive for cervical and anal high-risk human papillomavirus DNA and E6 RNA. Tumour Biol.
  16. Slama J, Sehnal B, Dusek L, Zima T, Cibula D (2015) Impact of risk factors on prevalence of anal HPV infection in women with simultaneous cervical lesion. Neoplasma 62: 308-314.
  17. Heard I, Etienney I, Potard V, Poizot-Martin I, Moore C, et al. (2015) High Prevalence of Anal Human Papillomavirus-Associated Cancer Precursors in a Contemporary Cohort of Asymptomatic HIV-Infected Women. Clin Infect Dis.
  18. King EM, Gilson R, Beddows S, Soldan K, Panwar K, et al. (2015) Human papillomavirus DNA in men who have sex with men: type-specific prevalence, risk factors and implications for vaccination strategies. Br J Cancer 112: 1585-1593.
  19. Tamalet C, Obry-Roguet V, Ressiot E, Bregigeon S, Del Grande J, et al. (2014) Distribution of human papillomavirus genotypes, assessment of HPV 16 and 18 viral load and anal related lesions in HIV positive patients: a cross-sectional analysis. J Med Virol 86:419-425.
  20. Tong WW, Shepherd K, Garland S, Meagher A, Templeton DJ, et al. (2015) Human papillomavirus 16-specific T-cell responses and spontaneous regression of anal high-grade squamous intraepithelial lesions. J Infect Dis 211: 405-415.
  21. Da Costa MM, Hogeboom CJ, Holly EA, Palefsky JM (2002) Increased risk of high-grade anal neoplasia associated with a human papillomavirus type 16 E6 sequence variant. J Infect Dis 185: 1229-1237.
  22. Bruland O, Fluge O, Immervoll H, Balteskard L, Myklebust M, et al. (2008) Gene expression reveals two distinct groups of anal carcinomas with clinical implications. Br J Cancer 98: 1264-1273.
  23. Koerber SA, Schoneweg C, Slynko A, Krug D, Haefner MF, et al. (2014) Influence of human papillomavirus and p16(INK4a) on treatment outcome of patients with anal cancer. Radiother Oncol 113: 331-336.
  24. Urgoiti GBR, Gustafson K, Klimowicz AC, Petrillo SK, Magliocco AM, et al. (2014) The prognostic value of HPV status and p16 expression in patients with carcinoma of the anal canal. PLoS ONE 9:e108790.
  25. Gilbert DC, Williams A, Allan K, Stokoe J, Jackson T, et al. (2013) p16INK4A, p53, EGFR expression and KRAS mutation status in squamous cell cancers of the anus: correlation with outcomes following chemo-radiotherapy. Radiother Oncol 109:146-151.
  26. Rödel F, Wieland U, Fraunholz I, Kitz J, Rave-Fränk M, et al. (2015) Human papillomavirus DNA load and p16INK4a expression predict for local control in patients with anal squamous cell carcinoma treated with chemoradiotherapy. Int J Cancer 136: 278-288.
  27. Bezerra AL, Lopes A, Landman G, Alencar GN, Torloni H, et al. (2001) Clinicopathologic features and human papillomavirus dna prevalence of warty and squamous cell carcinoma of the penis. Am J Surg Pathol 25: 673-678.
  28. Novara G, Galfano A, De Marco V, Artibani W, Ficarra V (2007) Prognostic factors in squamous cell carcinoma of the penis. Nat Clin Pract Urol 4: 140-146.
  29. Chan KW, Lam KY, Chan AC, Lau P, Srivastava G (1994) Prevalence of human papillomavirus types 16 and 18 in penile carcinoma: a study of 41 cases using PCR. J Clin Pathol 47: 823-826.
  30. Cupp MR, Malek RS, Goellner JR, Smith TF, Espy MJ (1995) The detection of human papillomavirus deoxyribonucleic acid in intraepithelial, in situ, verrucous and invasive carcinoma of the penis. J Urol 154: 1024-1029.
  31. Gregoire L, Cubilla AL, Reuter VE, Haas GP, Lancaster WD (1995) Preferential association of human papillomavirus with high-grade histological variants of penile-invasive squamous cell carcinoma. J Natl Cancer Inst 87:1705-1709.
  32. Lont AP, Kroon BK, Horenblas S, Gallee MP, Berkhof J, et al. (2006) Presence of high-risk human papillomavirus DNA in penile carcinoma predicts favorable outcome in survival. Int J Cancer 119: 1078-1081.
  33. Picconi MA, Eiján AM, Distéfano AL, Pueyo S, Alonio LV, et al. (2000) Human papillomavirus (HPV) DNA in penile carcinomas in Argentina: analysis of primary tumors and lymph nodes. J Med Virol 61: 65-69.
  34. Rubin MA, Kleter B, Zhou M, Ayala G, Cubilla AL, et al. (2001) Detection and typing of human papillomavirus DNA in penile carcinoma: evidence for multiple independent pathways of penile carcinogenesis. Am J Pathol 159: 1211-1218.
  35. Ferrándiz-Pulido C, Masferrer E, de Torres I, Lloveras B, Hernandez-Losa J, et al. (2013) Identification and genotyping of human papillomavirus in a Spanish cohort of penile squamous cell carcinomas: correlation with pathologic subtypes, p16(INK4a) expression, and prognosis. J Am Acad Dermatol 68: 73-82.
  36. Chaux A, Cubilla AL (2012) The role of human papillomavirus infection in the pathogenesis of penile squamous cell carcinomas. Semin Diagn Pathol 29: 67-71.
  37. Albero G, Castellsagué X, Lin HY, Fulp W, Villa LL, et al. (2014) Male circumcision and the incidence and clearance of genital human papillomavirus (HPV) infection in men: the HPV Infection in men (HIM) cohort study. BMC Infect Dis 14: 75.
  38. Hernandez BY, Shvetsov YB, Goodman MT, Wilkens LR, Thompson P, et al. (2010) Reduced clearance of penile human papillomavirus infection in uncircumcised men. J Infect Dis 201: 1340-1343.
  39. Silva RJ, Casseb J, Andreoli MA, Villa LL (2011) Persistence and clearance of HPV from the penis of men infected and non-infected with HIV. J Med Virol 83: 127-131.
  40. Dorfman S, Cavazza M, Cardozo J (2006) Penile cancer associated with so-called low-risk human papilloma virus. Report of five cases from rural Venezuela. Trop Doct 36: 232-233.
  41. Lebelo RL, Boulet G, Nkosi CM, Bida MN, Bogers JP, et al. (2014) Diversity of HPV types in cancerous and pre-cancerous penile lesions of South African men: implications for future HPV vaccination strategies. J Med Virol 86: 257-265.
  42. Busso-Lopes AF, Marchi FA, Kuasne H, Scapulatempo-Neto C, Trindade-Filho JC, et al. (2015) Genomic profiling of human penile carcinoma predicts worse prognosis and survival. Cancer Prev Res (Phila) 8:149-156
  43. Kayes O, Ahmed HU, Arya M, Minhas S (2007) Molecular and genetic pathways in penile cancer. Lancet Oncol 8: 420-429.
  44. Ferrandiz-Pulido C, Masferrer E, Toll A, Hernandez-Losa J, Mojal S, et al. (2013) mTOR signaling pathway in penile squamous cell carcinoma: pmTOR and peIF4E over expression correlate with aggressive tumor behavior. J Urol 190: 2288-2295.
  45. Stankiewicz E, Prowse DM, Ktori E, Cuzick J, Ambroisine L, et al. (2011) The retinoblastoma protein/p16 INK4A pathway but not p53 is disrupted by human papillomavirus in penile squamous cell carcinoma. Histopathology 58: 433-439.
  46. Agarwal S, Reddy GV, Reddanna P (2009) Eicosanoids in inflammation and cancer: the role of COX-2. Expert Rev Clin Immunol 5: 145-165.
  47. Wu WK, Sung JJ, Lee CW, Yu J, Cho CH (2010) Cyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett 295: 7-16.
  48. Golijanin D, Tan JY, Kazior A, Cohen EG, Russo P, et al. (2004) Cyclooxygenase-2 and microsomal prostaglandin E synthase-1 are overexpressed in squamous cell carcinoma of the penis. Clin Cancer Res 10: 1024-1031.
  49. Martins AC, Faria SM, Cologna AJ, Suaid HJ, Tucci S Jr (2002) Immunoexpression of p53 protein and proliferating cell nuclear antigen in penile carcinoma. J Urol 167: 89-92.
  50. Poetsch M, Hemmerich M, Kakies C, Kleist B, Wolf E, et al. (2011) Alterations in the tumor suppressor gene p16(INK4A) are associated with aggressive behavior of penile carcinomas. Virchows Arch 458: 221-229.
  51. Senba M, Mori N, Fujita S, Jutavijittum P, Yousukh A, et al. (2010) Relationship among human papillomavirus infection, p16(INK4a), p53 and NF- κkB activation in penile cancer from northern Thailand. Oncol Lett 1: 599-603.
  52. Lopez-Beltran A, Escudero AL (1997) Human papillomavirus and bladder cancer. Biomed Pharmacother 51: 252-257.
  53. Jimenez-Pacheco A, Exposito-Ruiz M, Arrabal-Polo MA, Lopez-Luque AJ (2012) Meta-analysis of studies analyzing the role of human papillomavirus in the development of bladder carcinoma. Korean J Urol 53: 240-247.
  54. Li N, Yang L, Zhang Y, Zhao P, Zheng T, et al. (2011) Human papillomavirus infection and bladder cancer risk: a meta-analysis. J Infect Dis 204: 217-223.
  55. Wiwanitkit V (2005) Urinary bladder carcinoma and human papilloma virus infection, an appraisal of risk. Asian Pac J Cancer Prev 6: 217-218.
  56. Ben Selma W, Ziadi S, Ben Gacem R, Amara K, Ksiaa F, et al. (2010) Investigation of human papillomavirus in bladder cancer in a series of Tunisian patients. Pathol Res Pract 206: 740-743.
  57. Berrada N, Al-Bouzidi A, Ameur A, Abbar M, El-Mzibri M, et al. (2013) Human papillomavirus detection in Moroccan patients with bladder cancer. J Infect Dev Ctries 7: 586-592.
  58. Kim SH, Joung JY, Chung J, Park WS, Lee KH, et al. (2014) Detection of human papillomavirus infection and p16 immunohistochemistry expression in bladder cancer with squamous differentiation. PLoS One Mar 27:e93525.
  59. Moonen PM, Bakkers JM, Kiemeney LA, Schalken JA, Melchers WJ, et al. (2007) Human papilloma virus DNA and p53 mutation analysis on bladder washes in relation to clinical outcome of bladder cancer. Eur Urol 52:464-468.
  60. Alexander RE, Williamson SR, Richey J, Lopez-Beltran A, Montironi R, et al. (2014) The expression patterns of p53 and p16 and an analysis of a possible role of HPV in primary adenocarcinoma of the urinary bladder. PLoS One 9: e95724.
  61. Lin Y, Mao Q, Zheng X, Yang K, Chen H, et al. (2011) Human papillomavirus 16 or 18 infection and prostate cancer risk: a meta-analysis. Ir J Med Sci 180: 497-503.
  62. Bae JM (2015) Human papillomavirus 16 infection as a potential risk factor for prostate cancer: an adaptive meta-analysis. Epidemiol Health 37: e2015005.
  63. Bergh J, Marklund I, Gustavsson C, Wiklund F, Grönberg H, et al. (2007) No link between viral findings in the prostate and subsequent cancer development. Br J Cancer 96: 137-139.
  64. Gazzaz FS, Mosli HA (2009) Lack of detection of human papillomavirus infection by hybridization test in prostatic biopsies. Saudi Med J 30: 633-637.
  65. Korodi Z, Dillner J, Jellum E, Lumme S, Hallmans G, et al. (2005) Human papillomavirus 16, 18, and 33 infections and risk of prostate cancer: a Nordic nested case-control study. Cancer Epidemiol Biomarkers Prev 14: 2952-2955.
  66. Sitas F, Urban M, Stein L, Beral V, Ruff P, et al. (2007) The relationship between anti-HPV-16 IgG seropositivity and cancer of the cervix, anogenital organs, oral cavity and pharynx, oesophagus and prostate in a black South African population. Infect Agent Cancer 2:6.
  67. Aghakhani A, Hamkar R, Parvin M, Ghavami N, Nadri M, et al. (2011) The role of human papillomavirus infection in prostate carcinoma. Scand J Infect Dis 43: 64-69.
  68. Pascale M, Pracella D, Barbazza R, Marongiu B, Roggero E, et al. (2013) Is human papillomavirus associated with prostate cancer survival? Dis Markers 35: 607-613.
  69. Michopoulou V, Derdas SP, Symvoulakis E, Mourmouras N, Nomikos A, et al. (2014) Detection of human papillomavirus (HPV) DNA prevalence and p53 codon 72 (Arg72Pro) polymorphism in prostate cancer in a Greek group of patients. Tumour Biol 35: 12765-12773.
  70. Leiros GJ, Galliano SR, Sember ME, Kahn T, Schwarz E, et al. (2005) Detection of human papillomavirus DNA and p53 codon 72 polymorphism in prostate carcinomas of patients from Argentina. BMC Urol 5: 15.
  71. Tachezy R, Hrbacek J, Heracek J, Salakova M, Smahelova J, et al. (2012) HPV persistence and its oncogenic role in prostate tumors. J Med Virol 84: 1636-1645.
  72. Kumar B, Cordell KG, Lee JS, Prince ME, Tran HH, et al. (2007) Response to therapy and outcomes in oropharyngeal cancer are associated with biomarkers including human papillomavirus, epidermal growth factor receptor, gender, and smoking. Int J Radiat Oncol Biol Phys 69:S109-111.
  73. Syrjänen K, Syrjänen S, Lamberg M, Pyrhönen S, Nuutinen J (1983) Morphological and immunohistochemical evidence suggesting human papillomavirus (HPV) involvement in oral squamous cell carcinogenesis. Int J Oral Surg 12: 418-424.
  74. Syrjänen K, Syrjänen S, Pyrhönen S (1982) Human papilloma virus (HPV) antigens in lesions of laryngeal squamous cell carcinomas. ORL J Otorhinolaryngol Relat Spec 44: 323-334.
  75. D'Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, et al. (2007) Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356: 1944-1956.
  76. Smith EM, Ritchie JM, Summersgill KF, Klussmann JP, Lee JH, et al. (2004) Age, sexual behavior and human papillomavirus infection in oral cavity and oropharyngeal cancers. Int J Cancer 108: 766-772.
  77. Kreimer AR, Clifford GM, Boyle P, Franceschi S (2005) Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomarkers Prev 14: 467-475.
  78. Ryerson AB, Peters ES, Coughlin SS, Chen VW, Gillison ML, et al. (2008) Burden of potentially human papillomavirus-associated cancers of the oropharynx and oral cavity in the US, 1998-2003. Cancer 113: 2901-2909.
  79. Ernster JA, Sciotto CG, O'Brien MM, Finch JL, Robinson LJ, et al. (2007) Rising incidence of oropharyngeal cancer and the role of oncogenic human papilloma virus. Laryngoscope 117: 2115-2128.
  80. Herrero R, Castellsagué X, Pawlita M, Lissowska J, Kee F, et al. (2003) Human papillomavirus and oral cancer: the International Agency for Research on Cancer multicenter study. J Natl Cancer Inst 95: 1772-1783.
  81. Smith EM, Ritchie JM, Pawlita M, Rubenstein LM, Haugen TH, et al. (2007) Human papillomavirus seropositivity and risks of head and neck cancer. Int J Cancer 120: 825-832.
  82. Syrjänen S (2005) Human papillomavirus (HPV) in head and neck cancer. J Clin Virol 32 Suppl 1: S59-66.
  83. Syrjänen S (2007) Human papillomaviruses in head and neck carcinomas. N Engl J Med 356: 1993-1995.
  84. Ritchie JM, Smith EM, Summersgill KF, Hoffman HT, Wang D, et al. (2003) Human papillomavirus infection as a prognostic factor in carcinomas of the oral cavity and oropharynx. Int J Cancer 104: 336-344.
  85. Smith EM, Wang D, Kim Y, Rubenstein LM, Lee JH, et al. (2008) P16INK4a expression, human papillomavirus, and survival in head and neck cancer. Oral Oncol 44: 133-142.
  86. Smeets SJ, Braakhuis BJ, Abbas S, Snijders PJ, Ylstra B, et al. (2006) Genome-wide DNA copy number alterations in head and neck squamous cell carcinomas with or without oncogene-expressing human papillomavirus. Oncogene 25: 2558-2564.
  87. Pyeon D, Newton M, Lambert P, den Boon J, Sengupta S, et al. (2007) Fundamental differences in cell cycle deregulation in human papillomavirus-positive and human papillomavirus-negative head/neck and cervical cancers. Cancer Res 67:4605-4619.
  88. Wang S, Trunk M, Schiffman M, Herrero R, Sherman M, et al. (2004) Validation of p16INK4a as a marker of oncogenic human papillomavirus infection in cervical biopsies from a population-based cohort in Costa Rica. Cancer Epidemiol Biomarkers Prev 13:1355-1360.
  89. Klaes R, Friedrich T, Spitovsky D, Ridder R, Petry U, et al. (2001) Overexpression of p16(INK4a) as a specific marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int J Cancer 92:276-284.
  90. Torrente MC, Rodrigo JP, Haigentz M Jr, Dikkers FG, Rinaldo A, et al. (2011) Human papillomavirus infections in laryngeal cancer. Head Neck 33: 581-586.
  91. Bonagura VR, Vambutas A, DeVoti JA, Rosenthal DW, Steinberg BM, et al. (2004) HLA alleles, IFN-gamma responses to HPV-11 E6, and disease severity in patients with recurrent respiratory papillomatosis. Hum Immunol 65: 773-782.
  92. DeVoti JA, Rosenthal DW, Wu R, Abramson AL, Steinberg BM, et al. (2008) Immune dysregulation and tumor-associated gene changes in recurrent respiratory papillomatosis: a paired microarray analysis. Mol Med 14: 608-617.
  93. Wu R, Coniglio SJ, Chan A, Symons MH, Steinberg BM (2007) Up-regulation of Rac1 by epidermal growth factor mediates COX-2 expression in recurrent respiratory papillomas. Mol Med 13: 143-150.
  94. Wu R, Abramsom A, Shikowitz M, Dannenberg A, Steinberg B (2005) Epidermal growth factor-induced cyclooxygenase-2 expression is mediated through phosphatidylinositol-3 kinase, not mitogen-activated protein-extracellular signal-regulated kinase kinase, in recurrent respiratory papillomatosis. Clin Cancer Res 11:6155-6161.
  95. Syrjänen KJ (2002) HPV infections and lung cancer. J Clin Pathol 55: 885-891.
  96. Cheng YW, Wu MF, Wang J, Yeh KT, Goan YG, et al. (2007) Human papillomavirus 16/18 E6 oncoprotein is expressed in lung cancer and related with p53 inactivation. Cancer Res 67: 10686-10693.
  97. Wang Y, Wang A, Jiang R, Pan H, Huang B, et al. (2008) Human papillomavirus type 16 and 18 infection is associated with lung cancer patients from the central part of China. Oncol Rep 20: 333-339.
  98. Yu Y, Yang A, Hu S, Yan H (2009) Correlation of HPV-16/18 infection of human papillomavirus with lung squamous cell carcinomas in Western China. Oncol Rep 21: 1627-1632.
  99. Aguayo F, Castillo A, Koriyama C, Higashi M, Itoh T, et al. (2007) Human papillomavirus-16 is integrated in lung carcinomas: a study in Chile. Br J Cancer 97: 85-91.
  100. Gatta LB, Balzarini P, Tironi A, Berenzi A, Benetti A, et al. (2012) Human papillomavirus DNA and p16 gene in squamous cell lung carcinoma. Anticancer Res 32: 3085-3089.
  101. Koshiol J, Rotunno M, Gillison ML, Van Doorn LJ, Chaturvedi AK, et al. (2011) Assessment of human papillomavirus in lung tumor tissue. J Natl Cancer Inst 103: 501-507.
  102. Sagerup CM, Nymoen DA, Halvorsen AR, Lund-Iversen M, Helland A, et al. (2014) Human papilloma virus detection and typing in 334 lung cancer patients. Acta Oncol 53: 952-957.
  103. Simen-Kapeu A, Surcel HM, Koskela P, Pukkala E, Lehtinen M (2010) Lack of association between human papillomavirus type 16 and 18 infections and female lung cancer. Cancer Epidemiol Biomarkers Prev 19: 1879-1881.
  104. Hasegawa Y, Ando M, Kubo A, Isa S, Yamamoto S, et al. (2014) Human papilloma virus in non-small cell lung cancer in never smokers: a systematic review of the literature. Lung Cancer 83: 8-13.
  105. Aguayo F, Anwar M, Koriyama C, Castillo A, Sun Q, et al. (2010) Human papillomavirus-16 presence and physical status in lung carcinomas from Asia. Infect Agent Cancer 5: 20.
  106. Cheng YW, Lee H, Shiau MY, Wu TC, Huang TT, et al. (2008) Human papillomavirus type 16/18 up-regulates the expression of interleukin-6 and antiapoptotic Mcl-1 in non-small cell lung cancer. Clin Cancer Res 14: 4705-4712.
  107. Fan X, Yu K, Wu J, Shao J, Zhu L, et al. (2015) Correlation between squamous cell carcinoma of the lung and human papillomavirus infection and the relationship to expression of p53 and p16. Tumour Biol 36: 3043-3049.
  108. Yu Y, Yang A, Hu S, Zhang J, Yan H (2013) Significance of human papillomavirus 16/18 infection in association with p53 mutation in lung carcinomas. Clin Respir J 7: 27-33.
  109. Shiau MY, Fan LC, Yang SC, Tsao CH, Lee H, et al. (2013) Human papillomavirus up-regulates MMP-2 and MMP-9 expression and activity by inducing interleukin-8 in lung adenocarcinomas. PLoS One 8: e54423.
  110. Li G, He L, Zhang E, Shi J, Zhang Q, et al. (2011) Overexpression of human papillomavirus (HPV) type 16 oncoproteins promotes angiogenesis via enhancing HIF-1a and VEGF expression in non-small cell lung cancer cells. Cancer Lett 311: 160-170.
  111. Tung MC, Wu HH, Cheng YW, Wang L, Chen CY, et al. (2013) Association of epidermal growth factor receptor mutations with human papillomavirus 16/18 E6 oncoprotein expression in non-small cell lung cancer. Cancer 119: 3367-3376.
  112. Kato T, Koriyama C, Khan N, Samukawa T, Yanagi M, et al. (2012) EGFR mutations and human papillomavirus in lung cancer. Lung Cancer 78: 144-147.
  113. Syrjänen KJ (1982) Histological changes identical to those of condylomatous lesions found in esophageal squamous cell carcinomas. Arch Geschwulstforsch 52: 283-292.
  114. Syrjänen K, Pyrhönen S, Aukee S, Koskela E (1982) Squamous cell papilloma of the esophagus: a tumour probably caused by human papilloma virus (HPV). Diagn Histopathol 5: 291-296.
  115. Syrjänen KJ (2002) HPV infections and oesophageal cancer. J Clin Pathol 55: 721-728.
  116. Guo F, Liu Y, Wang X, He Z, Weiss NS, et al. (2012) Human papillomavirus infection and esophageal squamous cell carcinoma: a case-control study. Cancer Epidemiol Biomarkers Prev 21: 780-785.
  117. Wang X, Tian X, Liu F, Zhao Y, Sun M, et al. (2010) Detection of HPV DNA in esophageal cancer specimens from different regions and ethnic groups: a descriptive study. BMC Cancer 10: 19.
  118. Koshiol J, Wei WQ, Kreimer AR, Chen W, Gravitt P, et al. (2010) No role for human papillomavirus in esophageal squamous cell carcinoma in China. Int J Cancer 127: 93-100.
  119. Dreilich M, Bergqvist M, Moberg M, Brattström D, Gustavsson I, et al. (2006) High-risk human papilloma virus (HPV) and survival in patients with esophageal carcinoma: a pilot study. BMC Cancer 6: 94.
  120. Matsha T, Donninger H, Erasmus RT, Hendricks D, Stepien A, et al. (2007) Expression of p53 and its homolog, p73, in HPV DNA positive oesophageal squamous cell carcinomas. Virology 369: 182-190.
  121. Ashworth MT, McDicken IW, Southern SA, Nash JR (1993) Human papillomavirus in squamous cell carcinoma of the oesophagus associated with tylosis. J Clin Pathol 46: 573-575.
  122. Bellizzi AM, Woodford RL, Moskaluk CA, Jones DR, Kozower BD, et al. (2009) Basaloid squamous cell carcinoma of the esophagus: assessment for high-risk human papillomavirus and related molecular markers. Am J Surg Pathol 33: 1608-1614.
  123. Rai N, Jenkins GJ, McAdam E, Hibbitts SJ, Fiander AN, et al. (2008) Human papillomavirus infection in Barrett's oesophagus in the UK: an infrequent event. J Clin Virol 43: 250-252.
  124. Hardefeldt HA, Cox MR, Eslick GD (2014) Association between human papillomavirus (HPV) and oesophageal squamous cell carcinoma: a meta-analysis. Epidemiol Infect 142: 1119-1137.
  125. Liyanage SS, Segelov E, Garland SM, Tabrizi SN, Seale H, et al. (2013) Role of human papillomaviruses in esophageal squamous cell carcinoma. Asia Pac J Clin Oncol 9: 12-28.
  126. Petrick JL, Wyss AB, Butler AM, Cummings C, Sun X, et al. (2014) Prevalence of human papillomavirus among oesophageal squamous cell carcinoma cases: systemic review and meta-analysis. Br J Cancer 110:2369-2377.
  127. Wang WL, Wang YC, Lee CT, Chang CY, Lo JL, et al. (2015) The impact of human papillomavirus infection on the survival and treatment response of patients with esophageal cancers. J Dig Dis 16: 256-263.
  128. Yong F, Xudong N, Lijie T (2013) Human papillomavirus types 16 and 18 in esophagus squamous cell carcinoma: a meta-analysis. Ann Epidemiol 23: 726-734.
  129. Zhang SK, Guo LW, Chen Q, Zhang M, Liu SZ, et al. (2015) The association between human papillomavirus 16 and esophageal cancer in Chinese population: a meta-analysis. BMC Cancer 15: 1096.
  130. Lu XM, Monnier-Benoit S, Mo LZ, Xu SY, Prétet JL, et al. (2008) Human papillomavirus in esophageal squamous cell carcinoma of the high-risk Kazakh ethnic group in Xinjiang, China. Eur J Surg Oncol 34: 765-770.
  131. Herbster S, Ferraro CT, Koff NK, Rossini A, Kruel CD, et al. (2012) HPV infection in Brazilian patients with esophageal squamous cell carcinoma: interpopulational differences, lack of correlation with surrogate markers and clinicopathological parameters. Cancer Lett 326:52-58.
  132. Lyronis ID, Baritaki S, Bizakis I, Krambovitis E, Spandidos DA (2008) K-ras mutation, HPV infection and smoking or alcohol abuse positively correlate with esophageal squamous carcinoma. Pathol Oncol Res 14: 267-273.
  133. Qi Z, Jiang Q, Yang J, Chen X, Wu H, et al. (2013) Human papillomavirus (HPV) infection and the risk of esophageal squamous cell carcinoma. Dis Esophagus 26: 61-67.
  134. Castillo A, Aguayo F, Koriyama C, Torres M, Carrascal E, et al. (2006) Human papillomavirus in esophageal squamous cell carcinoma in Colombia and Chile. World J Gastroenterol 12: 6188-6192.
  135. Shuyama K, Castillo A, Aguayo F, Sun Q, Khan N, et al. (2007) Human papillomavirus in high- and low-risk areas of oesophageal squamous cell carcinoma in China. Br J Cancer 96: 1554-1559.
  136. Tornesello ML, Monaco R, Nappi O, Buonaguro L, Buonaguro FM (2009) Detection of mucosal and cutaneous human papillomaviruses in oesophagitis, squamous cell carcinoma and adenocarcinoma of the oesophagus. J Clin Virol 45: 28-33.
  137. Shen ZY, Hu SP, Lu LC, Tang CZ, Kuang ZS, et al. (2002) Detection of human papillomavirus in esophageal carcinoma. J Med Virol 68: 412-416.
  138. Bohn OL, Navarro L, Saldivar J, Sanchez-Sosa S (2008) Identification of human papillomavirus in esophageal squamous papillomas. World J Gastroenterol 14: 7107-7111.
  139. Si HX, Tsao SW, Poon CS, Wong YC, Cheung AL (2005) Physical status of HPV-16 in esophageal squamous cell carcinoma. J Clin Virol 32: 19-23.
  140. Liu T, Liu Q, Liang M, Zheng S, Li XL, et al. (2013) Viral load of HPV 16/18 in esophageal squamous cell carcinoma in three ethnic groups living in Xinjiang Autonomous Region, China. Mol Biol Rep 40:2045-2052.
  141. Gao G, Roth M, Wei W, Abnet C, Chen F, et al. (2006) No association between HPV infection and the neoplastic progression of esophageal squamous cell carcinoma: result from a cross-sectional study in a high-risk region of China. Int J Cancer 119:1354-1359.
  142. Qi Z, Huo X, Xu X, Zhang B, Du M, et al. (2006) Relationship between HPV 16/18 and p53, p21WAF1, MDM2, Ki67 and cyclin D1 expression in esophageal squamous cell carcinoma: comparative study by using tissue microarray technology. Exp Oncol 28:235-240.
  143. Yang W, Zhang Y, Tian X, Ning T, Ke Y (2008) p53 Codon 72 polymorphism and the risk of esophageal squamous cell carcinoma. Mol Carcinog 47: 100-104.
  144. Yang J, Liu B, Li W, Xiong H, Qiu H, et al. (2013) Association of p53 and MDM2 polymorphisms with risk of human papillomavirus (HPV)-related esophageal squamous cell carcinoma (ESCC). Cancer Epidemiol 37: 629-633.
  145. Malik SM, Nevin DT, Cohen S, Hunt JL, Palazzo JP (2011) Assessment of immunohistochemistry for p16INK4 and high-risk HPV DNA by in situ hybridization in esophageal squamous cell carcinoma. Int J Surg Pathol 19: 31-34.
  146. Hussain S, Bharti AC, Salam I, Bhat MA, Mir MM, et al. (2009) Transcription factor AP-1 in esophageal squamous cell carcinoma: alterations in activity and expression during human Papillomavirus infection. BMC Cancer 9:329.
  147. Zhang DH, Chen JY, Hong CQ, Yi DQ, Wang F, et al. (2014) High-risk human papillomavirus infection associated with telomere elongation in patients with esophageal squamous cell carcinoma with poor prognosis. Cancer 120:2673-2683.
  148. Yu Q, Yang J, Liu B, Li W, Hu G, et al. (2014) Combined effects of leukocyte telomere length, p53 polymorphism and human papillomavirus infection on esophageal squamous cell carcinoma in a Han Chinese population. Cancer Epidemiol 38:569-575.
  149. Goldie SJ, Kohli M, Grima D, Weinstein MC, Wright TC, et al. (2004) Projected clinical benefits and cost-effectiveness of a human papillomavirus 16/18 vaccine. J Natl Cancer Inst 96: 604-615.
  150. Newall AT, Beutels P, Wood JG, Edmunds WJ, MacIntyre CR (2007) Cost-effectiveness analyses of human papillomavirus vaccination. Lancet Infect Dis 7: 289-296.
  151. Gillison ML (2008) Human papillomavirus-related diseases: oropharynx cancers and potential implications for adolescent HPV vaccination. J Adolesc Health 43: S52-60.
  152. Gillison ML, Chaturvedi AK, Lowy DR (2008) HPV prophylactic vaccines and the potential prevention of noncervical cancers in both men and women. Cancer 113: 3036-3046.
  153. Graham DM, Isaranuwatchai W, Habbous S, de Oliveira C, Liu G, et al. (2015) A cost-effectiveness analysis of human papillomavirus vaccination of boys for the prevention of oropharyngeal cancer. Cancer 121: 1785-1792.
  154. Luttjeboer J1, Westra TA, Wilschut JC, Nijman HW, Daemen T, et al. (2013) Cost-effectiveness of the prophylactic HPV vaccine: an application to the Netherlands taking non-cervical cancers and cross-protection into account. Vaccine 31: 3922-3927.
Citation: Cristina Merkhofer, Joel Maslow (2015) Human Papilloma Virus (HPV) Infection and Non-Cervical Oncogenic Disease States. Virolmycol 4:144.

Copyright: © 2015 Merkhofer C,This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top