Next Article in Journal
P38 Mediates Tumor Suppression through Reduced Autophagy and Actin Cytoskeleton Changes in NRAS-Mutant Melanoma
Next Article in Special Issue
Correlations between Molecular Alterations, Histopathological Characteristics, and Poor Prognosis in Esophageal Adenocarcinoma
Previous Article in Journal
Intraoperative Autofluorescence Imaging for Parathyroid Gland Identification during Total Laryngectomy with Thyroidectomy
Previous Article in Special Issue
HER2-Positive Gastroesophageal Cancers Are Associated with a Higher Risk of Brain Metastasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Causal Link of Human Papillomavirus in Barrett Esophagus and Adenocarcinoma: Are We There Yet?

by
Shanmugarajah Rajendra
1,2,3,* and
Prateek Sharma
4,5
1
Gastro-Intestinal Viral Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, Sydney, NSW 2170, Australia
2
South Western Sydney Clinical School, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
3
Department of Gastroenterology & Hepatology, Bankstown-Lidcombe Hospital, South-Western Sydney Local Health Network, Bankstown, Sydney, NSW 2200, Australia
4
Division of Gastroenterology and Hepatology, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
5
School of Medicine, University of Kansas, Kansas City, MO 66160, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(3), 873; https://doi.org/10.3390/cancers15030873
Submission received: 27 November 2022 / Revised: 7 January 2023 / Accepted: 26 January 2023 / Published: 31 January 2023
(This article belongs to the Special Issue Oesophageal Adenocarcinoma)

Abstract

:

Simple Summary

Esophageal cancer is not an uncommon malignancy in the world with a relatively high mortality. There has been a dramatic upward trajectory (approximately 400%) in the number of patients diagnosed with the glandular form of esophageal cancer (adenocarcinoma) since the 1970’s. It has been thought that this condition is related to chronic heartburn causing Barrett’s esophagus (a pre-cancerous condition), smoking and obesity. Nevertheless, this excess number of esophageal cancers is not entirely explained by these known risk factors despite improved screening, detection and treatment strategies. The discovery of high-risk type human papillomavirus (with an increased cancer potential) being strongly associated with this glandular form of tumor of the esophagus in a subset of patients (approximately 25%) may partially explain this anomaly. This discovery will aid improved detection rates of those with a greater risk of progressing to malignancy and modified treatment strategies including vaccination in the hope of reduced mortality.

Abstract

Esophageal cancer is a relatively common malignancy worldwide with a high mortality (5-year survival of <15%). Despite screening, surveillance, improved imaging and treatment, the exponential rise in OAC continues. The strongest risk factors for OAC are chronic heartburn and metaplastic transformation of the lower third of the esophagus (Barrett’s esophagus). The risk profile includes Caucasian race, male gender older age, obesity and smoking. Although the tumor risk in BO has been progressively revised downwards, the exponential rise in OAC remains unchecked. This paradox points to an unidentified missing link. Relatively recently, we provided the world’s initial data for a strong association of biologically relevant hr-HPV with BD and OAC. Since then, systematic reviews and meta-analysis have documented HPV DNA prevalence rates in OAC of between 13 to 35%. In this review, we provide some evidence for a probable causal relationship between hr-HPV and OAC. This is challenging given the multifactorial etiology and long latency. Increasingly, high-risk HPV (hr-HPV) is regarded as a risk factor for OAC. This discovery will aid identification of a sub-group of high-risk progressors to esophageal cancer by surveillance and the development of effective preventive strategies including vaccination.

1. Introduction

Fifteen to twenty percent of all human malignancies are caused by bacterial or viral pathogens [1,2]. Helicobacter pylori, high-risk human papillomavirus (hr-HPV), hepatitis B virus and hepatitis C are responsible for more than ninety percent of pathogen related cancers [3,4]. Most of these oncogenic pathogens are modifiable risk factors with either effective treatments and/or prevention strategies in place [1].
Epidemiological and basic science studies have demonstrated that hr-HPV causes cervical, anogenital, and some oropharyngeal cancers [5,6] This virus has been detected in the vast majority of squamous cervical tumor specimens (90–100%) as compared to a minority of controls (20%) [7]. In addition, the HPV viral genome has been identified in a significant majority of cervical dysplastic lesions [8]. HPVs were previously thought to preferentially infect squamous epithelia only. More recently, studies have established a consistent and strong association between hr-HPV and adenocarcinoma of the cervix. Eighty-five percent of this heterogeneous group of malignancies are HPV associated [9,10]. In this respect, HPV exhibits an affinity for glandular tissue as well as squamous epithelium.

2. Literature Search and Study Selection

This is a narrative review and articles were evaluated in Pubmed, MEDLINE, Scopus and Google Scholar between 1960 and 2022 except for one seminal publication by Koch (English translation from German) published in 1881. This article was identified in the course of review of references of the evaluated publications to locate further relevant research.
We searched publications relevant to the title of the review using the following search terms: human papillomavirus, HPV, viruses, viral pathogens, oncogenic viruses, HPV integration, HPV associated cancers, esophageal adenocarcinoma, esophageal cancer, esophageal tumor, esophagus, Barrett’s esophagus, Barrett’s metaplasia, Barrett’s dysplasia and epidemiology.
Publications deemed suitable for inclusion in this review were (i) all in English or translated into English, (ii) full-text articles, (iii) related to the above search terms. This resulted in a total of 7042 articles. Studies pertaining only to squamous cell carcinoma of the esophagus or gastro-esophageal junction tumors or cardia malignancies without separately including OAC were excluded. In addition, duplicate and irrelevant publications were also excluded. Ultimately, a total of 108 articles were included in the review.

3. HPV as a Common Denominator in Esophageal Adenocarcinoma and Cervical Cancer

One of the authors has previously hypothesized (SR) that HPV could be involved in the pathogenesis of esophageal adenocarcinoma just as in cervical cancer. It is based on the premise of similar immunology and genetics and a not dissimilar transformation zone that could be a common predilection site for the virus [11]. Papillomatous lesions have been identified in the esophagus which throws up the possibility of a viral pathogen (biological plausibility) [12]. In fact, up to 10% of esophageal squamous cell papillomas have detectable HPV [13]. Other viruses, namely Herpes simplex virus (HSV), cytomegalovirus (CMV) and Epstein-Barr virus (EBV) have all been detected in the esophagus and, in a few publications, have even been associated with cancer [14]. In the animal world, cattle suffer with upper gastro-intestinal tract papillomas and carcinomas which have been linked to the bovine papillomavirus (BPV 4) and bracken fern (co-factor) (animal model) [15]. BPV, although species-specific, belongs to the Papillomavirus genus. Most BPV infections in cattle regress but when the animals feed on bracken fern the papillomatosis persists with cancer formation both of epithelial and mesenchymal origin [16]. Moreover, the transforming activity of BPV preparations in bovine cells has been demonstrated [17].
We have proceeded to undertake studies to confirm or disprove the above hypothesis and will discuss these in detail as well as investigations undertaken by others, showing both positive and negative associations. More importantly, we shall frame the data in a manner consistent with the title of this review.

4. Establishing Causality

Establishing that a virus causes cancer is a tedious undertaking with a high barrier for acceptance by the scientific community [2]. Attributing causality to microbes in human cancers has to take into account (a) the long latency between infection and cancer development, (b) that only a small number of infected individuals develop malignancies, (c) that persistent chronic infection is a key precursor event, (d) that host and environmental co-factors are a necessary pre-requisite and (e) possible strain-dependent variation in oncogenicity.
The existing model in the development of esophageal glandular carcinogenesis clearly only refers to chronic heartburn/reflux and Barrett’s esophagus (BO). Chronic, frequent and severe GORD symptoms are strongly associated with OAC (odds ratio (OR) of 43.5) [18]. Barrett’s metaplasia is considered a pre-malignant condition that can lead to OAC. The malignancy potential ranges between 0.12 and 0.13% per annum [19,20]. In the past, the cancer risk was thought to be approximately eight times higher at 1.0% per year (one case per 125 patient-years). This estimated risk has been reduced over time to as low as one case per 200 to 300 patient years [21,22].
Herein lies the dichotomy. OAC has been growing at an exponential rate in the Western world, i.e., by 600% since the 1970’s [23], although recently, the rate of rise has stabilized in the US and Sweden [24,25]. This exponential rise of OAC has occurred against a background of a reduction in the cancer risk linked with BO. It has been postulated that to “explain a rise of esophageal adenocarcinoma of this magnitude, the prevalence of a strong risk factor must also rise exponentially” [23]. The finding of a strong association of biologically relevant high-risk human papillomavirus (hr-HPV) with Barrett’s dysplasia (BD) and cancer [26] may potentially explain this anomaly. The data on head and neck tumors, another type of virally linked cancer [27,28,29], provides further credence to this statement. The prevalence of HPV in oropharyngeal cancers (OPC) increased more than four-fold in 20 years from 16% in the 1980’s to 73% in the 2000s (incidence: 0.8 cases/100,000 subjects to 2.6/100,000) [27]. Is there a common denominator here in regard to hr-HPV and OAC and oropharyngeal malignancies?
The usual criteria for establishing causality are the consistency of association at the epidemiological and/or molecular level and the ability of the pathogen to cause tumors in animal models or its transforming ability in cell culture [30].
Assigning causality in medical research usually involves adhering to Koch’s postulates. These state that a pathogen be found in all hosts with disease but not in those without but with the exception of asymptomatic carriers. Furthermore, the microbe must be able to be identified and shown to cause disease in a healthy host [31].
The Hill criteria examine (a) the robustness of association, (b) consistency, (c) temporality, (d) biological gradient, (e) biological plausibility, (f) biological coherence, (g) specificity, (h) experimental evidence and (i) analogy [32]. Lipkin has amalgamated both Koch’s postulates and Hill’s criteria and graded causality as possible, probable, or confirmed. A “possible relationship” has been defined as evidence of exposure to a microorganism, the detection of genetic sequences/proteins, visualization by imaging, or demonstration of an adaptive immune response to a pathogen. A “probable relationship” is one where there is a template for a comparable disease caused by a similar pathogen in a related host (analogy), and the biological gradient is greatest at the site of pathology. This may include presence of pathogen genetic material or protein particles in or adjacent to diseased host cells, or IgM antibodies against the microbe indicating recent exposure. A “confirmed relationship” requires adherence to Koch’s postulates or establishing that the disease can be ameliorated or prevented by pathogen specific drugs, antibodies, or vaccines [33].

5. Consistency across Studies and Biological Gradient (Probable Relationship)

A systematic review reported a pooled HPV prevalence rate of 35% (95%CI, 13.2–65.7%) from 5 studies which totaled 174 patients (predominantly Caucasians) with OAC [34]. Another review of 19 studies found the prevalence of HPV in OAC was 13% (95% CI: 2–29%) (Table 1). The investigators opined that the low detection rate may have been due to small sample sizes and unreliable assays [35]. The shortcomings of the meta-analysis included lack of data to analyze genotypes, primer types and geographical variations, and paucity of studies which compared HPV prevalence in subjects with BO and OAC or those with normal esophageal epithelium. Some of these studies used unsatisfactory classification of tissue histology. Many used a less reliable non-nested PCR method, failed to obtain biopsies from the transformation zone (squamo-columnar junction rather than the gastro-esophageal junction) and most either lacked proper controls or were devoid of them altogether [36,37,38,39]. The low viral load in esophageal epithelium further exacerbates the problem [26,40]. Very recently, the largest meta-analysis to date on the association between the virus and both sub-types of esophageal cancer, i.e squamous and adenocarcinoma, involving 13,401 subjects (33 studies of which 19 were case-control, 11 cohort/observational and 3 cross-sectional) revealed that hr-HPV 16 and 18 are strong co-factors in the etiopathogenesis of esophageal cancer [41].

6. Strength of Association and Biological Gradient (Probable Relationship)

In 2013, we published for the very first time 2 papers in the American Journal of Gastroenterology demonstrating a strong link between transcriptionally active high risk genotypes 16 (predominantly) and 18 with Barrett’s dysplasia and OAC [26,40]. HPV DNA detection was undertaken by nested polymerase chain rection (PCR) and viral oncogene activity (indicative of viral transcription) estimated by E6/7 oncogene messenger ribonucleic acid (mRNA) expression (gold standard) and p16INK4A immunohistochemistry in esophageal tissue of patients with dysplastic and non-dysplastic (metaplastic) Barrett’s, and OAC, as well as controls. Both junctional and lesional biopsies were obtained for analysis.
We found that 81/261 (31%) patients had detectable HPV DNA in the esophagus. In both controls and BO, the virus was more likely to be located at the transformation (transitional) zone. When compared with controls (18.0%), HPV positivity was significantly more common in BD (68.6%) and OAC (66.7%), but not in BO (22.1%) [26].
Of the viral positive patients, approximately 93% were high-risk (HR) HPV, i.e., types 16 and 18. In addition, p16INK4A expression was greater in BD (44.1%) and OAC (44.4%) and significantly reduced in BO patients (10.6%).
In the 66 viral positive subjects, tested for E6/E7 oncoproteins, none of the controls (n = 16) or those with Barrett’s metaplasia (n = 13) were positive. In contrast, 9/22 (40.9%) BD and 9/15 (60%) OAC patients had detectable E6/E7mRNA (p < 0.001). When all 3 markers were positive, (HPV DNA, p16INK4A, and E6/E7 mRNA), a strong association with disease severity was noted as compared with none detected (fulfills criteria for probable relationship). This strength of association is as strong as chronic GORD for OAC [18].
Viral infections in the esophagus have a predilection for the transformation zone. In both dysplastic/OAC subjected to endoscopic mucosal resection, high-risk genotypes 16 and 18 were concentrated at the transition zone (SCJ) compared to the lesion (biological gradient, plausibility, and specificity) [11,42,43,44]. Transforming HPV infections in varying epithelial sites (ano-genital including cervix and oropharynx) cause cancers mainly at the transition zone (analogy). In the cervix, this is the anatomical area where metaplastic squamous cells are detected in otherwise columnar-lined endocervical glands [45,46,47]. Intriguingly, studies have revealed that Barrett’s metaplastic tissue can originate from a distinct category of embryonic cells located at the junction of the transition zone. Moreover, in a genetically engineered animal (mouse) model, Barrett’s metaplasia and dysplasia was generated from the SCJ (biological mechanism and experiment) [48,49].

7. Analogy and Biological Gradient (Probable Relationship)

The higher the viral load the greater the risk of developing cervical cancer and its precursor lesion, cervical intraepithelial neoplasia (CIN) which is the archetypal HPV driven disease. Nevertheless, this is not necessarily the case in other HPV associated lesions, where viral load does not correspond to disease severity [50,51,52]. Incorporation of the viral DNA into the host genome is an important step in malignant conversion. Generally, frequency of hr-HPV DNA integration corresponds to increasing severity of cervical lesions [53,54]. Integration leads to disruption of the HPV E2 gene, and thus unchecked over-expression of viral E6 and E7 oncogenes [55].
Work undertaken in our laboratory has revealed that a greater abundance of virus as per viral load measurement and greater integration of the pathogen in human esophageal tissue involving the HPV genotypes 16 or 18 was significantly associated with disease severity. This severity increased in ascending order as per the Barrett’s metaplasia-dysplasia-adenocarcinoma sequence [40]. Patients in the benign end of the spectrum were more likely to be infected with the HPV-18 genotype. This was in contrast to the cohort with more severe disease (BD and OAC subjects) in whom the predominant HPV genotype was 16 [40]. HPV DNA in situ-hybridization (ISH) and E6/E7 mRNA expression was confined to columnar lined esophageal lesions only. It was quite intriguing to find that the squamous epithelium was totally spared of any positive detection signals (Figure 1). This clearly indicates that HPV has a tropism for esophageal glandular tissue. An analogous model is the affinity of HPV18 for columnar cells of the endocervical canal [56]. In the latter disease, the columnar cells are not suited for HPV reproduction as they are unfavorable to the establishment of a differentiation gradient, contrary to squamous epithelia. What is baffling is that despite the absence of a productive viral infection, hr-HPV can still cause cancer of the endocervical canal [57].

8. Analogy and Experimentation [Probable Relationship]

In the cervix, continual hr-HPV infection is associated with CIN and frank carcinoma [58,59]. Prolonged uninterrupted hr-HPV infection is a predictor of failed treatment (loop electrosurgical excision (LEEP)) of CIN [60]. In an analogous fashion, we investigated whether HPV infection and p53 over-expression (as determined by IHC and next generation sequencing) was abolished in virally infected patients after endoscopic treatment for BD and early esophageal adenocarcinoma (intramucosal). In this prospective study which involved 40 patients, it was found that persistent biologically active hr-HPV infection (types 16 and 18) and p53 over-expression were mutually exclusively associated with failed endoscopic ablation of dysplastic BO/OAC [61]. HPV positive BD/intra-mucosal OAC had largely absent p53 mutations and next generation sequencing confirmed wild type TP53 in these lesions. This is a hallmark of HPV driven cancers [62,63].
The failure to detect p53 in BD may be due to silencing mutations of the said gene which results in reduced or absent expression [64] It may also be the case that this negative staining for this tumor suppressor gene is due to the E6 degradation of p53 [65]. It is tempting to postulate that there maybe 2 or more carcinogenic pathways (viral and non-viral) involved in esophageal carcinogenesis, much akin to head and neck tumors [65,66,67], one which is HPV driven, i.e., mostly absent p53 mutations and the other, p53 mediated. Validation of our findings by others is paramount prior to contemplating translation from bench to bedside.

9. Comparative Genomic Analysis Reveals Distinct Differences between HPV-Positive and HPV Negative OAC (Probable Relationship)

HPV+ tumors represent a unique subgroup of cancer with distinct prognosis and genetic aberrations. HPVs exert their oncogenic role via the viral oncoproteins E6 and E7. These oncoproteins target tumor suppressor signaling pathways which are critical for modulation of cellular growth. E6 oncoprotein induces degradation of p53 [68], whereas E7 binds and breaks down the pRB retinoblastoma tumor suppressor protein [69]. Due to feedback loops from pRb loss, up-regulation of p16INK4a occurs in viral positive tumors which serves as a proxy marker for the presence of the active HPV oncoprotein, E7. Compared with HPV-negative HNSCC (which is caused by life-style habits, e.g., smoking and excess alcohol intake), HPV-positive cancers have less genome-wide DNA copy number alterations, reduced genome-wide hypomethylation, quite infrequent or absent TP53 mutations, and reduced expression of EGFR [70,71]. In another study involving targeted next-generation sequencing of HPV+ and HPV− head and neck squamous cell carcinoma (HNSCC), TP53 mutations were detected in all (100%) of HPV negative cases. In contrast, only 1/20 (5%) had TP53 mutations in the HPV+ group [68].
A pilot study involving whole exome sequencing of 4 HPV associated OAC and 8 non-HPV OAC revealed that viral-positive OAC subjects had half the number of somatic (non-silent) mutations compared with virus-negative esophageal cancer patients [72]. TP53 mutations were non-existent in the HPV-positive OAC group whereas half of the HPV-negative OAC patients exhibited TP53 aberrations. There were less than a third of cancer driver genes in the HPV+ group as compared with the viral negative cohort. The integrated form of HPV16 was identified in a few discordant pairs [72]. Exomes make a tiny fraction of the human genome, and it is speculated that this viral integration is a rather unpredictable process [73]. Hence, larger sized studies examining the whole genome are necessary to further confirm these results. For example, protein expression studies which may explain function of these mutated gene are warranted for validation purposes. They may also shed light on esophageal carcinogenesis. Unfortunately, no comparable studies by other investigators have been reported on genomic analysis of HPV+ versus HPV-OAC.
A prospective cross-sectional study investigating the prevalence of biologically active HPV virus and related protein markers in patients representing the Barrett’s metaplastic, dysplastic and adenocarcinoma sequence has shed some light on the molecular mechanisms involved in a subgroup of virally associated esophageal lesions. In this investigation, 56/218 patients were HPV DNA positive (HPV16 (n = 42), genotype18 (n = 13), genotype 6 (n = 1)). Biologically active HPV (DNA+/RNA+) was only found in the dysplastic and adenocarcinoma group (n = 21). The majority of HPV DNA+/RNA+ BD/OAC exhibited over-expression of p16, with reduced or absent staining of pRb and p53. This finding was significantly different to controls. Reduced or absent expression of p53 had the strongest association with DNA+/RNA+ esophageal lesions (OR = 23.5, p = 0.0029). Sequencing confirmed the overwhelming majority of p53low specimens exhibited wild-type status. pRblow/p53low provided the optimal strength of association for DNA+/RNA+ BD/OAC. Hence, this study concluded that active HPV involvement in the pathogenesis of a subset of BD/OAC is characterized by wild-type p53 and aberrations of the retinoblastoma protein pathway [74].

10. Experimentation (Evidence for Possible Relationship)

Modern techniques facilitate concurrent analysis of antibodies against a broad range of viruses including HPV. Antibodies to HPV6/11/16/18/31/33/45/52/58 and E6/E7/E1/E2 as well as L1 antigens were analyzed using multiplex technology in 438 patients representing the GORD spectrum disease. These patients were categorized as hospital/reflux controls and cases were defined as Barrett’s metaplasia (BM), BD and intramucosal EAC. Seropositivity for individual HPV proteins was approximately 10% in both cases and controls. More specifically, there was no difference in antibody seroprevalence to any HPV antigen/antibody combination between cases and controls. Among HPV16 DNA+ BD/OAC subjects, antibodies to HPV 16 E7 oncoprotein were significantly more prevalent (11.5%) as compared to controls (1.5%). In HPV18 DNA+ cases, antibodies to HPV18 E1 protein were present in 50% of cases versus 1.5% of controls (p = 0.0002) [75]. Antibodies to E6 and E7 cancer proteins are considered markers of HPV-driven cancer, namely head and neck and cervical malignancies [76,77,78,79,80]. L1 antibodies denote exposure, or cumulative infection of HPV [81,82]. Antibodies against E1 and E2 for HPV 16 have been shown to be additional cancer biomarkers in oropharyngeal cancer (OPC) [83]. Larger studies encompassing patients with more severe disease have been undertaken with mixed results. Lagergren et al. tested IgG against HPV 16/18 capsids in 173 OAC (Siewert Class I and II), 121 OSCC and 302 controls and their only significant (unexpected) finding was a negative association between HPV18 and OAC. The authors related this anomaly to possible bias [84]. Site-specific correlation, i.e., testing the esophageal lesion for HPV DNA status and correlating it with serological analysis was not undertaken. Their results differed from 3 prior published positive studies [85,86,87]. In the latter 2 studies, HPV16 seropositivity was associated with a 6 times excess risk of esophageal malignancy (mainly squamous carcinoma). Again, the downside to these studies was that site-specific serological correlation was not performed [86,87].

11. HPV and Survival in OAC (Probable Relationship)

HPV confers a favorable prognosis in Barrett’s high-grade dysplasia and EAC suggesting a distinct tumor biology as compared with viral negative esophageal lesions [72,88]. In 142 patients with Barrett’s high-grade dysplasia (HGD) and EAC (37 HPV-positive and 105 HPV-negative), there was a superior disease-free survival (DFS) in the viral-positive group (40.3 vs. 24.1 months; p = 0.003) and an improved overall survival (OS) [43.7 vs. 29.8 months; p = 0.009].
Similarly, an improved disease-free survival (DFS) was demonstrated for HPV, biologically active virus, E6/E7 mRNA and high p16INK4A expression associated Barrett’s HGD and OAC. This was due to a reduction in distant metastasis and probably improved loco-regional control in virally infected patients. These findings mirror the data in head and neck cancers (HNSCC) with improved survival and reduced loco-regional recurrence in HPV+ lesions tumors versus viral negative head and neck malignancies. The exception was findings of no difference in distant metastases due to underpowered studies [89,90,91]. In OSCC, p16+ patients had superior 5-year OS rates and progression-free survival (PFS) as compared to those lacking this biomarker [91]. Likewise, another study investigating p16 status in OSCC patients subjected to neo-adjuvant chemotherapy found that tumors positive for this biomarker had superior complete remission rates than those without. [92]. Another study by Wang et al. concurred with the above findings. They reported that patients with HPV-16 positive advanced OSCC, had a significantly improved survival and response to chemoradiotherapy than those with viral negative esophageal tumors [93].
Conversely, Furihata et al. found that HPV positivity and the presence of p53 conferred a poorer prognosis in OSCC patients than those without the virus or absent p53 [94].
In a related study, da Costa et al. found HPV, p16 and p53 had no bearing on prognosis in OSCC. [95]. Others have also shown no survival benefit between HPV positive versus viral negative OSCC [96,97,98].
In another study, investigating some of the HPV associated biomarkers, namely, retinoblastoma protein (pRb), cyclin D1 (CD1), mini-chromosome maintenance protein (MCM2) and Ki-67 in relation to survival in patients with HGD/OAC found that only low cyclin D1 levels were associated with a favorable prognosis, specifically for OS [99]. Overexpression of cyclin D1 in OAC has been well documented [99,100]. Nevertheless, when pRb, CD1, MCM2, and Ki-67 were stratified by human papillomavirus status there was a survival benefit in esophageal tumours [101]. The above findings suggest the enticing possibility of personalization of therapy predicated on viral status. We caution that these findings have to be corroborated by other investigators in larger cohorts, preferably with more severe disease. Only then could one begin to contemplate treatment de-escalation and the potential benefit of reduced toxicity without adversely affecting survival.

12. HPV and Esophageal Cancer Cell Lines

Hr-HPV genotype 18 has been isolated from esophageal squamous cell carcinoma cell lines and the authors identified the integration site to a part of chromosome 8. This may represent a possible molecular mechanism for viral oncogenesis in esophageal malignancy [102]. Boon et al. analyzed the transcript expression profiles and functions of E6 and E7 in order to decipher the function of HPV18 in esophageal cancer cell lines. They found disruption of E2, and expression of E6, E7, E1 and L1 transcripts just as in HPV driven cervical cancer. A point of differentiation was that E7 preferentially targeted p130 in the two esophageal cancer cell lines, instead of pRb as in cervical cancer [103].

13. Conclusions

Further replication of the studies alluded to above is paramount in establishing a causal relationship between HPV and a subset of Barrett’s dysplasia and OAC. Thus far, the International Agency on Research on Cancer (IARC) concluded in 2012 that there was inadequate epidemiological evidence for an association between esophageal cancer (squamous cell carcinoma) and HPV based on case series and case-control studies [3]. Since then, at least 3 systematic reviews have been published demonstrating a positive association between HPV and OAC together with a few studies addressing molecular and genomic signatures characteristic of these virally driven BD and OAC [26,34,35,40]. Unfortunately, the Advisory Group recommendations on priorities for the IARC Monographs during 2020–2024 place HPV as a low priority for evaluation, though virologists worldwide will be making submissions for upgrading this assessment [104].
In the interim, we believe there is accumulating evidence for a probable relationship for hr-HPV in the etiology of a sub-group of OAC based on the data provided above. In fact, hr-HPV continues to gain traction as a risk factor for esophageal adenocarcinoma on the basis of more recent investigations, meta-analyses and reviews. [41,105,106,107,108]. The largest meta-analysis conducted so far on both squamous and adenocarcinoma of the esophagus involving over 13,000 subjects has concluded that HPV 16 and 18 are both probable strong risk factors in the pathogenesis of the disease [41]. Critical factors in the understanding of HPV oncogenesis in esophageal adenocarcinoma remain unresolved. Both genomic and proteomic studies will provide mechanistic insights for OAC with and without a viral etiology. Furthermore, the confirmation of a distinct molecular signature via sequencing and proteomic analysis characteristic of HPV positive esophageal malignancy would greatly aid risk stratification and improved/modified treatment protocols for patients, with the potential of reduced toxicity. The identification of a sub-group of high-risk progressors to esophageal cancer by surveillance and the development of effective preventive strategies including vaccination would have a great impact on public health. The potential histopathological reversal (regression) of a subset of patients with Barrett’s dysplasia via HPV therapeutic vaccination is the holy grail we aspire to in the near future.
Part of this data was presented at an invited lecture of an identical title by SR at the Asia Pacific Digestive Week, Kuala Lumpur, 19–22 August 2021.

Author Contributions

Conceptualization: S.R.; Methodology: S.R. and P.S.; Data curation: S.R. and P.S.; Writing—Original Draft Preparation: S.R. and P.S.; Writing—Review and Editing: S.R. and P.S.; Visualization: S.R. and P.S.; Supervision: S.R. and P.S.; Project Administration: S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

S.R. has consulted for Endogene. P.S. has provided consultancy services to Bausch, Boston Scientific Corporation, CDx Labs, Covidien LP, Exact Sciences, Fujifilm Medical Systems USA, Inc., Lucid, Lumendi and Medtronic. He has received grants from Cosmo Pharmaceuticals, Covidien, Docbot, ERBE USA Inc., Fujifilm Holdings America Corporation, Ironwood Pharmaceuticals, Inc., Medtronic USA, Inc., Olympus, Salix and US Endoscopy.

References

  1. De Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018, a worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [Green Version]
  2. White, M.K.; Pagano, J.S.; Khalili, K. Viruses and human cancers: A long road of discovery of molecular paradigms. Clin. Microbiol. Rev. 2014, 27, 463–481. [Google Scholar] [CrossRef] [Green Version]
  3. Biological Agents. A review of human carcinogens. IARC Monogr. Eval. Carcinog. Risks Hum. 2012, 100, 1–441. [Google Scholar]
  4. Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global burden of cancers attributable to infections in 2012, a synthetic analysis. Lancet Glob. Health 2016, 4, e609–e616. [Google Scholar] [CrossRef] [Green Version]
  5. McLaughlin-Drubin, M.E.; Meyers, J.; Munger, K. Cancer associated human papillomaviruses. Curr. Opin. Virol. 2012, 2, 459–466. [Google Scholar] [CrossRef] [Green Version]
  6. Humans IWGotEoCRt. Human papillomaviruses. IARC Monogr. Eval. Carcinog. Risks Hum. 1995, 64, 1–378. [Google Scholar]
  7. Bosch, F.X.; Lorincz, A.; Munoz, N.; Meijer, C.J.; Shah, K.V. The causal relation between human papillomavirus and cervical cancer. J. Clin. Pathol. 2002, 55, 244–265. [Google Scholar] [CrossRef] [Green Version]
  8. Walboomers, J.M.; Jacobs, M.V.; Manos, M.M.; Bosch, F.X.; Kummer, J.A.; Shah, K.V.; Snijders, P.J.; Peto, J.; Meijer, C.J.; Muñoz, N. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J. Pathol. 1999, 189, 12–19. [Google Scholar] [CrossRef]
  9. Castellsague, X.; Diaz, M.; de Sanjose, S.; Muñoz, N.; Herrero, R.; Franceschi, S.; Peeling, R.W.; Ashley, R.; Smith, J.S.; Snijders, P.J.F.; et al. Worldwide human papillomavirus etiology of cervical adenocarcinoma and its cofactors: Implications for screening and prevention. J. Natl. Cancer Inst. 2006, 98, 303–315. [Google Scholar]
  10. Stolnicu, S.; Barsan, I.; Hoang, L.; Patel, P.; Terinte, C.; Pesci, A.; Aviel-Ronen, S.; Kiyokawa, T.; Alvarado-Cabrero, I.; Pike, M.C.; et al. International Endocervical Adenocarcinoma Criteria and Classification (IECC): A new pathogenetic classification for invasive adenocarcinomas of the endocervix. Am. J. Surg. Pathol. 2018, 42, 214–226. [Google Scholar] [CrossRef]
  11. Rajendra, S.; Robertson, I.K. Similar immunogenetics of Barrett’s oesophagus and cervical neoplasia: Is HPV the common denominator? J. Clin. Pathol. 2010, 63, 1–3. [Google Scholar] [CrossRef]
  12. Odze, R.; Antonioli, D.; Shocket, D.; Noble-Topham, S.; Goldman, H.; Upton, M. Esophageal squamous papillomas. A clinicopathologic study of 38 lesions and analysis for human papillomavirus by the polymerase chain reaction. Am. J. Surg. Pathol. 1993, 17, 803–812. [Google Scholar] [CrossRef]
  13. Takeshita, K.; Murata, S.I.; Mitsufuji, S.; Wakabayashi, N.; Kataoka, K.; Tsuchihashi, Y.; Okanoue, T. Clinicopathological characteristics of esophageal squamous papillomas in Japanese patients—With comparison of findings from Western countries. Acta Histochem. Cytochem. 2006, 39, 23–30. [Google Scholar] [CrossRef]
  14. Chang, C.; Worrell, S.G. Viruses and esophageal cancer. Dis. Esophagus 2020, 33, doaa036. [Google Scholar] [CrossRef]
  15. Campo, M.S. Papillomas and cancer in cattle. Cancer Surv. 1987, 6, 39–54. [Google Scholar]
  16. Jarrett, W.F.H.; McNeil, P.E.; Grimshaw, W.T.R.; Selman, I.E.; McIntyre, W.I.M. High incidence area of cattle cancer with a possible interaction between an environmental carcinogen and a papilloma virus. Nature 1978, 274, 215–217. [Google Scholar] [CrossRef]
  17. Black, P.H.; Hartley, J.W.; Rowe, W.P.; Huebner, R.J. Transformation of bovine tissue culture cells by bovine papilloma virus. Nature 1963, 199, 1016–1018. [Google Scholar] [CrossRef]
  18. Lagergren, J.; Bergstrom, R.; Lindgren, A.; Nyren, O. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N. Engl. J. Med. 1999, 340, 825–831. [Google Scholar] [CrossRef]
  19. Bhat, S.; Coleman, H.G.; Yousef, F.; Johnston, B.T.; McManus, D.T.; Gavin, A.T.; Murray, L.J. Risk of malignant progression in Barrett’s esophagus patients: Results from a large population-based study. J. Natl. Cancer Inst. 2011, 103, 1049–1057. [Google Scholar] [CrossRef] [Green Version]
  20. Hvid-Jensen, F.; Pedersen, L.; Drewes, A.M.; Sorensen, H.T.; Funch-Jensen, P. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N. Engl. J. Med. 2011, 365, 1375–1383. [Google Scholar] [CrossRef] [Green Version]
  21. Shaheen, N.J.; Crosby, M.A.; Bozymski, E.M.; Sandler, R.S. Is there publication bias in the reporting of cancer risk in Barrett’s esophagus? Gastroenterology 2000, 119, 333–338. [Google Scholar] [CrossRef]
  22. Yousef, F.; Cardwell, C.; Cantwell, M.M.; Galway, K.; Johnston, B.T.; Murray, L. The incidence of esophageal cancer and high-grade dysplasia in Barrett’s esophagus: A systematic review and meta-analysis. Am. J. Epidemiol. 2008, 168, 237–249. [Google Scholar] [CrossRef] [Green Version]
  23. Pohl, H.; Welch, H.G. The Role of Overdiagnosis and Reclassification in the Marked Increase of Esophageal Adenocarcinoma Incidence. J. Natl. Cancer Inst. 2005, 97, 142–146. [Google Scholar] [CrossRef]
  24. Pohl, H.; Sirovich, B.; Welch, H.G. Esophageal adenocarcinoma incidence: Are we reaching the peak? Cancer Epidemiol. Biomarkers. Prev. 2010, 19, 1468–1470. [Google Scholar] [CrossRef]
  25. Lagergren, J.; Mattsson, F. No further increase in the incidence of esophageal adenocarcinoma in Sweden. Int. J. Cancer 2011, 129, 513–516. [Google Scholar] [CrossRef]
  26. Rajendra, S.; Wang, B.; Snow, E.T.; Sharma, P.; Pavey, D.; Merrett, N.; Ball, M.J.; Brain, T.; Fernando, R.; Robertson, I.K. Transcriptionally active human papillomavirus is strongly associated with Barrett’s dysplasia and esophageal adenocarcinoma. Am. J. Gastroenterol. 2013, 108, 1082–1093. [Google Scholar] [CrossRef]
  27. Chaturvedi, A.K.; Engels, E.A.; Pfeiffer, R.M.; Hernandez, B.Y.; Xiao, W.; Kim, E.; Jiang, B.; Goodman, M.T.; Sibug-Saber, M.; Cozen, W.; et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J. Clin. Oncol. 2011, 29, 4294–4301. [Google Scholar] [CrossRef]
  28. Nasman, A.; Attner, P.; Hammarstedt, L.; Du, J.; Eriksson, M.; Giraud, G.; Ahrlund-Richter, S.; Marklund, L.; Romanitan, M.; Lindquist, D.; et al. Incidence of human papillomavirus (HPV) positive tonsillar carcinoma in Stockholm, Sweden: An epidemic of viral-induced carcinoma? Int. J. Cancer 2009, 125, 362–366. [Google Scholar] [CrossRef]
  29. Hocking, J.S.; Stein, A.; Conway, E.L.; Regan, D.; Grulich, A.; Law, M.; Brotherton, J.M. Head and neck cancer in Australia between 1982 and 2005 show increasing incidence of potentially HPV-associated oropharyngeal cancers. Br. J. Cancer 2011, 104, 886–891. [Google Scholar] [CrossRef] [Green Version]
  30. Pagano, J.S.; Blaser, M.; Buendia, M.A.; Damania, B.; Khalili, K.; Raab-Traub, N.; Roizman, B. Infectious agents and cancer: Criteria for a causal relation. Semin. Cancer Biol. 2004, 14, 453–471. [Google Scholar] [CrossRef]
  31. Koch, R. Zur Untersuchung von Pathogenen Organismen. Mitthdungen Uus Dem Kais. Gesundbeitsamte 1881, 1, 1–48. [Google Scholar]
  32. Hill, A.B. The Environment and Disease: Association or Causation? Proc. R. Soc. Med. 1965, 58, 295–300. [Google Scholar] [CrossRef] [Green Version]
  33. Lipkin, W.I. The changing face of pathogen discovery and surveillance. Nat. Rev. Microbiol. 2013, 11, 133–141. [Google Scholar] [CrossRef] [PubMed]
  34. Li, X.; Gao, C.; Yang, Y.; Zhou, F.; Li, M.; Jin, Q.; Gao, L. Systematic review with meta-analysis: The association between human papillomavirus infection and oesophageal cancer. Aliment. Pharm. 2014, 39, 270–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kunzmann, A.T.; Graham, S.; McShane, C.M.; Doyle, J.; Tommasino, M.; Johnston, B.; Jamison, J.; James, J.A.; McManus, D.; Anderson, L.A. The prevalence of viral agents in esophageal adenocarcinoma and Barrett’s esophagus: A systematic review. Eur. J. Gastroenterol. Hepatol. 2017, 29, 817–825. [Google Scholar] [CrossRef]
  36. El-Serag, H.B.; Hollier, J.M.; Gravitt, P.; Alsarraj, A.; Younes, M. Human papillomavirus and the risk of Barrett’s esophagus. Dis. Esophagus 2013, 26, 517–521. [Google Scholar] [CrossRef] [Green Version]
  37. Rai, N.; Jenkins, G.J.; McAdam, E.; Hibbitts, S.J.; Fiander, A.N.; Powell, N.G. Human papillomavirus infection in Barrett’s oesophagus in the UK: An infrequent event. J. Clin. Virol. 2008, 43, 250–252. [Google Scholar] [CrossRef]
  38. Iyer, A.; Rajendran, V.; Adamson, C.S.; Peng, Z.; Cooper, K.; Evans, M.F. Human papillomavirus is detectable in Barrett’s esophagus and esophageal carcinoma but is unlikely to be of any etiologic significance. J. Clin. Virol. 2011, 50, 205–208. [Google Scholar] [CrossRef]
  39. Antonsson, A.; Knight, L.; Whiteman, D.C. Human papillomavirus not detected in esophageal adenocarcinoma tumor specimens. Cancer Epidemiol. 2016, 41, 96–98. [Google Scholar] [CrossRef]
  40. Wang, B.; Rajendra, S.; Pavey, D.; Sharma, P.; Merrett, N.; Wu, X.; Snow, E.T.; Kumbhari, V.; Ball, M.J.; Robertson, I.K. Viral load and integration status of high-risk human papillomaviruses in the Barrett’s metaplasia-dysplasia-adenocarcinoma sequence. Am. J. Gastroenterol. 2013, 108, 1814–1816. [Google Scholar] [CrossRef]
  41. Hussain, S.; Rani, J.; Tulsyan, S.; Sisodiya, S.; Chikara, A.; Nazir, S.U.; Srivastava, A.; Khan, A.; Dash, N.R.; Anoop Saraya, A.; et al. Influence of HPV infection in esophageal cancer: A systematic review and meta-analysis. Gene Rep. 2022, 28, 101640. [Google Scholar] [CrossRef]
  42. Rajendra, S.; Robertson, I.K. Barrett’s Oesophagus; Acid Hum. Papilloma Virus? J. Clin. Virol. 2009, 44, 176. [Google Scholar] [CrossRef]
  43. Baldwin, A.; Münger, K. Molecular events associated with human papillomavirus-induced human cancers. In Viral Oncology: Basic Science and Clinical Applications; Wiley-Blackwell: Hoboken, NJ, USA, 2010; pp. 23–35. [Google Scholar]
  44. Rajendra, S.; Sharma, P. Transforming human papillomavirus infection and the esophageal transformation zone: Prime time for total excision/ablative therapy? Dis. Esophagus. 2019, 32, doz008. [Google Scholar] [CrossRef]
  45. Parkin, D.M.; Bray, F. Chapter 2, The burden of HPV-related cancers. Vaccine 2006, 24, S11–S25. [Google Scholar] [CrossRef] [PubMed]
  46. Howley, P.; Lowy, D. Papillomaviruses. Fields Virol. 2007, 2, 2299–2354. [Google Scholar]
  47. Ng, W.K.; Cheung, L.K.; Li, A.S.; Cheung, F.M.; Chow, J.C. Transitional cell metaplasia of the uterine cervix is related to human papillomavirus: Molecular analysis in seven patients with cytohistologic correlation. Cancer 2002, 96, 250–258. [Google Scholar] [CrossRef] [PubMed]
  48. Nicholson, A.M.; Graham, T.A.; Simpson, A.; Humphries, A.; Burch, N.; Rodriguez-Justo, M.; Novelli, M.; Harrison, R.; Wright, N.A.; McDonald, S.A.; et al. Barrett’s metaplasia glands are clonal, contain multiple stem cells and share a common squamous progenitor. Gut 2012, 61, 1380–1389. [Google Scholar] [CrossRef]
  49. Quante, M.; Bhagat, G.; Abrams, J.A.; Marache, F.; Good, P.; Lee, M.D.; Lee, Y.; Friedman, R.; Asfaha, S.; Dubeykovskaya, Z.; et al. Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell 2012, 21, 36–51. [Google Scholar] [CrossRef] [Green Version]
  50. Josefsson, A.M.; Magnusson, P.K.; Ylitalo, N.; Sørensen, P.; Qwarforth-Tubbin, P.; Andersen, P.K.; Melbye, M.; Adami, H.O.; Gyllensten, U.B. Viral load of human papilloma virus 16 as a determinant for development of cervical carcinoma in situ: A nested case-control study. Lancet 2000, 355, 2189–2193. [Google Scholar] [CrossRef]
  51. Si, H.X.; Tsao, S.W.; Poon, C.S.; Wang, L.D.; Wong, Y.C.; Cheung, A.L. Viral load of HPV in esophageal squamous cell carcinoma. Int. J. Cancer J. Int. Cancer 2003, 103, 496–500. [Google Scholar] [CrossRef]
  52. Deng, Z.; Hasegawa, M.; Kiyuna, A.; Matayoshi, S.; Uehara, T.; Agena, S.; Yamashita, Y.; Ogawa, K.; Maeda, H.; Suzuki, M. Viral load, physical status, and E6/E7 mRNA expression of human papillomavirus in head and neck squamous cell carcinoma. Head Neck 2013, 35, 800–808. [Google Scholar] [CrossRef] [PubMed]
  53. Boshart, M.; Gissmann, L.; Ikenberg, H.; Kleinheinz, A.; Scheurlen, W.; zur Hausen, H. A new type of papillomavirus DNA, its presence in genital cancer biopsies and in cell lines derived from cervical cancer. EMBO J. 1984, 3, 1151–1157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Saunier, M.; Monnier-Benoit, S.; Mauny, F.; Ana Dalstein, V.; Briolat, J.; Riethmuller, D.; Kantelip, B.; Schwarz, E.; Mougin, C.; Prétet, J.L. Analysis of human papillomavirus type 16 (HPV16) DNA load and physical state for identification of HPV16-infected women with high-grade lesions or cervical carcinoma. J. Clin. Microbiol. 2008, 46, 3678–3685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Arias-Pulido, H.; Peyton, C.L.; Joste, N.E.; Vargas, H.; Wheeler, C.M. Human papillomavirus type 16 integration in cervical carcinoma in situ and in invasive cervical cancer. J. Clin. Microbiol. 2006, 44, 1755–1762. [Google Scholar] [CrossRef] [Green Version]
  56. Smith, J.S.; Lindsay, L.; Hoots, B.; Keys, J.; Franceschi, S.; Winer, R.; Clifford, G.M. Human papillomavirus type distribution in invasive cervical cancer and high-grade cervical lesions: A meta-analysis update. Int. J. Cancer 2007, 121, 621–632. [Google Scholar] [CrossRef]
  57. Chow, L.T.; Broker, T.R.; Steinberg, B.M. The natural history of human papillomavirus infections of the mucosal epithelia. APMIS 2010, 118, 422–449. [Google Scholar] [CrossRef]
  58. Ho, G.Y.F.; Bierman, R.; Beardsley, L.; Chang, C.J.; Burk, R.D. Natural History of Cervicovaginal Papillomavirus Infection in Young Women. N. Engl. J. Med. 1998, 338, 423–428. [Google Scholar] [CrossRef]
  59. Schiffman, M.; Castle, P.E. Human papillomavirus: Epidemiology and public health. Arch. Pathol. Lab. Med. 2003, 127, 930–934. [Google Scholar] [CrossRef]
  60. Bae, J.H.; Kim, C.J.; Park, T.C.; Namkoong, S.E.; Park, J.S. Persistence of human papillomavirus as a predictor for treatment failure after loop electrosurgical excision procedure. Int. J. Gynecol. Cancer 2007, 17, 1271–1277. [Google Scholar] [CrossRef]
  61. Rajendra, S.; Wang, B.; Pavey, D.; Sharma, P.; Yang, T.; Lee, C.S.; Gupta, N.; Ball, M.J.; Gill, R.S.; Wu, X. Persistence of Human Papillomavirus, Overexpression of p53, and Outcomes of Patients After Endoscopic Ablation of Barrett’s Esophagus. Clin. Gastroenterol. Hepatol. 2015, 13, 1364–1368.e5. [Google Scholar] [CrossRef]
  62. Ojesina, A.I.; Lichtenstein, L.; Freeman, S.S.; Pedamallu, C.S.; Imaz-Rosshandler, I.; Pugh, T.J.; Cherniack, A.D.; Ambrogio, L.; Cibulskis, K.; Bertelsen, B.; et al. Landscape of genomic alterations in cervical carcinomas. Nature 2014, 506, 371–375. [Google Scholar] [CrossRef] [Green Version]
  63. Agrawal, N.; Frederick, M.J.; Pickering, C.R.; Bettegowda, C.; Chang, K.; Li, R.J.; Fakhry, C.; Xie, T.X.; Zhang, J.; Wang, J.; et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 2011, 333, 1154–1157. [Google Scholar] [CrossRef] [Green Version]
  64. Kaye, P.V.; Haider, S.A.; James, P.D.; Soomro, I.; Catton, J.; Parsons, S.L.; Ragunath, K.; Ilyas, M. Novel staining pattern of p53 in Barrett’s dysplasia--the absent pattern. Histopathology 2010, 57, 933–935. [Google Scholar] [CrossRef]
  65. Scheffner, M.; Takahashi, T.; Huibregtse, J.M.; Minna, J.D.; Howley, P.M. Interaction of the human papillomavirus type 16 E6 oncoprotein with wild-type and mutant human p53 proteins. J. Virol. 1992, 66, 5100–5105. [Google Scholar] [CrossRef] [Green Version]
  66. Brennan, J.A.; Boyle, J.O.; Koch, W.M.; Goodman, S.N.; Hruban, R.H.; Eby, Y.J.; Couch, M.J.; Forastiere, A.A.; Sidransky, D. Association between cigarette smoking and mutation of the p53 gene in squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 1995, 332, 712–717. [Google Scholar] [CrossRef]
  67. Gillison, M.L.; Koch, W.M.; Capone, R.B.; Spafford, M.; Westra, W.H.; Wu, L.; Zahurak, M.L.; Daniel, R.W.; Viglione, M.; Symer, D.E.; et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J. Natl. Cancer Inst. 2000, 92, 709–720. [Google Scholar] [CrossRef]
  68. Lechner, M.; Frampton, G.M.; Fenton, T.; Feber, A.; Palmer, G.; Jay, A.; Pillay, N.; Forster, M.; Cronin, M.T.; Lipson, D.; et al. Targeted next-generation sequencing of head and neck squamous cell carcinoma identifies novel genetic alterations in HPV+ and HPV− tumors. Genome Med. 2013, 5, 49. [Google Scholar] [CrossRef] [Green Version]
  69. Agrawal, N.; Jiao, Y.; Bettegowda, C.; Hutfless, S.M.; Wang, Y.; David, S.; Cheng, Y.; Twaddell, W.S.; Latt, N.L.; Shin, E.J.; et al. Comparative Genomic Analysis of Esophageal Adenocarcinoma and Squamous Cell Carcinoma. Cancer Discov. 2012, 2, 899–905. [Google Scholar] [CrossRef] [Green Version]
  70. Smeets, S.J.; Braakhuis, B.J.; Abbas, S.; Snijders, P.J.; Ylstra, B.; van de Wiel, M.A.; Meijer, G.A.; Leemans, C.R.; Brakenhoff, R.H. Genome-wide DNA copy number alterations in head and neck squamous cell carcinomas with or without oncogene-expressing human papillomavirus. Oncogene 2006, 25, 2558–2564. [Google Scholar] [CrossRef] [Green Version]
  71. Richards, K.L.; Zhang, B.; Baggerly, K.A.; Colella, S.; Lang, J.C.; Schuller, D.E.; Krahe, R. Genome-wide hypomethylation in head and neck cancer is more pronounced in HPV-negative tumors and is associated with genomic instability. PLoS ONE 2009, 4, e4941. [Google Scholar] [CrossRef] [Green Version]
  72. Rajendra, S.; Wang, B.; Merrett, N.; Sharma, P.; Humphris, J.; Lee, H.C.; Wu, J. Genomic analysis of HPV-positive versus HPV-negative oesophageal adenocarcinoma identifies a differential mutational landscape. J. Med. Genet. 2016, 53, 227–231. [Google Scholar] [CrossRef] [PubMed]
  73. Parfenov, M.; Pedamallu, C.S.; Gehlenborg, N.; Freeman, S.S.; Danilova, L.; Bristow, C.A.; Lee, S.; Hadjipanayis, A.G.; Ivanova, E.V.; Wilkerson, M.D.; et al. Characterization of HPV and host genome interactions in primary head and neck cancers. Proc. Natl. Acad. Sci. USA 2014, 111, 15544–15549. [Google Scholar] [CrossRef] [Green Version]
  74. Rajendra, S.; Yang, T.; Xuan, W.; Sharma, P.; Pavey, D.; Lee, C.S.; Le, S.; Collins, J.; Wang, B. Active human papillomavirus involvement in Barrett’s dysplasia and oesophageal adenocarcinoma is characterized by wild-type p53 and aberrations of the retinoblastoma protein pathway. Int. J. Cancer 2017, 141, 2037–2049. [Google Scholar] [CrossRef] [Green Version]
  75. Rajendra, S.; Xuan, W.; Hufnagel, K.; Sharma, P.; Pavey, D.; Alhajjiri, N.; Rattan, A.; Wang, B. Antibodies against human papillomavirus proteins in Barrett’s dysplasia and intramucosal esophageal adenocarcinoma. Ann. N. Y. Acad. Sci. 2020, 1470, 44–56. [Google Scholar] [CrossRef]
  76. Smith, E.M.; Ritchie, J.M.; Pawlita, M.; Rubenstein, L.M.; Haugen, T.H.; Turek, L.P.; Hamsikova, E. Human papillomavirus seropositivity and risks of head and neck cancer. Int. J. Cancer 2007, 120, 825–832. [Google Scholar] [CrossRef]
  77. Kreimer, A.R.; Johansson, M.; Waterboer, T.; Kaaks, R.; Chang-Claude, J.; Drogen, D.; Tjønneland, A.; Overvad, K.; Quirós, J.R.; González, C.A.; et al. Evaluation of human papillomavirus antibodies and risk of subsequent head and neck cancer. J. Clin. Oncol. 2013, 31, 2708–2715. [Google Scholar] [CrossRef]
  78. Achour, M.; Zeghal, D.; Kochbati, L.; Kahla, S.; Zouari, F.; Maalej, M.; Oueslati, R. Antibody response for L1, E6 and E7 HPV 16 and HPV 18 antigens in Tunisian women with cervical cancer and controls. J. Immunoass. Immunochem. 2008, 29, 266–280. [Google Scholar] [CrossRef]
  79. Meschede, W.; Zumbach, K.; Braspenning, J.; Scheffner, M.; Benitez-Bribiesca, L.; Luande, J.; Gissmann, L.; Pawlita, M. Antibodies against early proteins of human papillomaviruses as diagnostic markers for invasive cervical cancer. J. Clin. Microbiol. 1998, 36, 475–480. [Google Scholar] [CrossRef] [Green Version]
  80. Reuschenbach, M.; Waterboer, T.; Wallin, K.L.; Einenkel, J.; Dillner, J.; Hamsikova, E.; Eschenbach, D.; Zimmer, H.; Heilig, B.; Kopitz, J.; et al. Characterization of humoral immune responses against p16, p53, HPV16 E6 and HPV16 E7 in patients with HPV-associated cancers. Int. J. Cancer J. Int. Cancer 2008, 123, 2626–2631. [Google Scholar] [CrossRef]
  81. Frazer, I.H. Interaction of human papillomaviruses with the host immune system: A well evolved relationship. Virology 2009, 384, 410–414. [Google Scholar] [CrossRef] [Green Version]
  82. Wang, S.S.; Schiffman, M.; Herrero, R.; Carreon, J.; Hildesheim, A.; Rodriguez, A.C.; Bratti, M.C.; Sherman, M.E.; Morales, J.; Guillen, D.; et al. Determinants of human papillomavirus 16 serological conversion and persistence in a population-based cohort of 10,000 women in Costa Rica. Br. J. Cancer 2004, 91, 1269–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Holzinger, D.; Wichmann, G.; Baboci, L.; Michel, A.; Höfler, D.; Wiesenfarth, M.; Schroeder, L.; Boscolo-Rizzo, P.; Herold-Mende, C.; Dyckhoff, G.; et al. Sensitivity and specificity of antibodies against HPV16 E6 and other early proteins for the detection of HPV16-driven oropharyngeal squamous cell carcinoma. Int. J. Cancer 2017, 140, 2748–2757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lagergren, J.; Wang, Z.; Bergström, R.; Dillner, J.; Nyrén, O. Human papillomavirus infection and esophageal cancer: A nationwide seroepidemiologic case-control study in Sweden. J. Natl. Cancer Inst. 1999, 91, 156–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Han, C.; Qiao, G.; Hubbert, N.L.; Li, L.; Sun, C.; Wang, Y.; Yan, M.; Xu, D.; Li, Y.; Lowy, D.R.; et al. Serologic association between human papillomavirus type 16 infection and esophageal cancer in Shaanxi Province, China. J. Natl. Cancer Inst. 1996, 88, 1467–1471. [Google Scholar] [CrossRef] [Green Version]
  86. Bjørge, T.; Hakulinen, T.; Engeland, A.; Jellum, E.; Koskela, P.; Lehtinen, M.; Luostarinen, T.; Paavonen, J.; Sapp, M.; Schiller, J.; et al. A prospective, seroepidemiological study of the role of human papillomavirus in esophageal cancer in Norway. Cancer Res. 1997, 57, 3989–3992. [Google Scholar]
  87. Dillner, J.; Knekt, P.; Schiller, J.T.; Hakulinen, T. Prospective seroepidemiological evidence that human papillomavirus type 16 infection is a risk factor for oesophageal squamous cell carcinoma. BMJ 1995, 311, 1346. [Google Scholar] [CrossRef] [Green Version]
  88. Rajendra, S.; Xuan, W.; Merrett, N.; Sharma, P.; Sharma, P.; Pavey, D.; Yang, T.; Santos, L.D.; Sharaiha, O.; Pande, G.; et al. Survival Rates for Patients with Barrett High-grade Dysplasia and Esophageal Adenocarcinoma with or Without Human Papillomavirus Infection. JAMA Netw. Open 2018, 1, e181054. [Google Scholar] [CrossRef]
  89. Fakhry, C.; Westra, W.H.; Li, S.; Li, S.; Cmelak, A.; Ridge, J.A.; Pinto, H.; Forastiere, A.; Gillison, M.L. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J. Natl. Cancer Inst. 2008, 100, 261–269. [Google Scholar] [CrossRef] [Green Version]
  90. Ang, K.K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tân, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef] [Green Version]
  91. Cao, F.; Zhang, W.; Zhang, F.; Han, H.; Xu, J.; Cheng, Y. Prognostic significance of high-risk human papillomavirus and p16(INK4A) in patients with esophageal squamous cell carcinoma. Int. J. Clin. Exp. Med. 2014, 7, 3430–3438. [Google Scholar]
  92. Kumar, R.; Ghosh, S.K.; Verma, A.K.; Talukdar, A.; Deka, M.K.; Wagh, M.; Bahar, H.M.; Tapkire, R.; Chakraborty, K.P.; Kannan, R.R.; et al. p16 Expression as a Surrogate Marker for HPV Infection in Esophageal Squamous Cell Carcinoma can Predict Response to Neo-Adjuvant Chemotherapy. Asian Pac. J. Cancer Prev. 2015, 16, 7161–7165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wang, W.L.; Wang, Y.C.; Lee, C.T.; Chang, C.Y.; Lo, J.L.; Kuo, Y.H.; Hsu, Y.C.; Mo, L.R. The impact of human papillomavirus infection on the survival and treatment response of patients with esophageal cancers. J. Dig. Dis. 2015, 16, 256–263. [Google Scholar] [CrossRef] [PubMed]
  94. Furihata, M.; Ohtsuki, Y.; Ogoshi, S.; Takahashi, A.; Tamiya, T.; Ogata, T. Prognostic significance of human papillomavirus genomes (type-16,-18) and aberrant expression of p53 protein in human esophageal cancer. Int. J. Cancer 1993, 54, 226–230. [Google Scholar] [CrossRef]
  95. Da Costa, A.M.; Fregnani, J.H.T.G.; Pastrez, P.R.A.; Mariano, V.S.; Silva, E.M.; Neto, C.S.; Guimarães, D.P.; Villa, L.L.; Sichero, L.; Syrjanen, K.J.; et al. HPV infection and p53 and p16 expression in esophageal cancer: Are they prognostic factors? Infect. Agents Cancer 2017, 12, 54. [Google Scholar] [CrossRef] [PubMed]
  96. Dreilich, M.; Bergqvist, M.; Moberg, M.; Brattström, D.; Gustavsson, I.; Bergström, S.; Wanders, A.; Hesselius, P.; Wagenius, G.; Gyllensten, U. High-Risk Hum. Papilloma Virus (HPV) Surviv. Patients Esophageal Carcinoma: A Pilot Study. BMC Cancer 2006, 6, 94. [Google Scholar] [CrossRef] [Green Version]
  97. Antonsson, A.; Green, A.C.; Mallitt, K.A.; O’Rourke, P.K.; Pandeya, N.; Pawlita, M.; Waterboer, T.; Neale, R.E. Prevalence and stability of antibodies to 37 human papillomavirus types—A population-based longitudinal study. Virology 2010, 407, 26–32. [Google Scholar] [CrossRef]
  98. Hippeläinen, M.; Eskelinen, M.; Lipponen, P.K.; Chang, F.; Syrjänen, K. Mitotic activity index, volume corrected mitotic index and human papilloma-virus suggestive morphology are not prognostic factors in carcinoma of the oesophagus. Anticancer Res. 1993, 13, 677–681. [Google Scholar]
  99. Rajendra, S.; Sharma, P.; Gautam, S.D.; Saxena, M.; Kapur, A.; Sharma, P.; Merrett, N.; Yang, T.; Santos, L.D.; Pavey, D.; et al. Association of Biomarkers for Human Papillomavirus with Survival Among Adults with Barrett High-grade Dysplasia and Esophageal Adenocarcinoma. JAMA Netw. Open 2020, 3, e1921189. [Google Scholar] [CrossRef]
  100. Arber, N.; Gammon, M.D.; Hibshoosh, H.; Britton, J.A.; Zhang, Y.; Schonberg, J.B.; Roterdam, H.; Fabian, I.; Holt, P.R.; Weinstein, I.B.; et al. Overexpression of cyclin D1 occurs in both squamous carcinomas and adenocarcinomas of the esophagus and in adenocarcinomas of the stomach. Hum. Pathol. 1999, 30, 1087–1092. [Google Scholar] [CrossRef]
  101. Langer, R.; Von Rahden, B.H.; Nahrig, J.; Von Weyhern, C.; Reiter, R.; Feith, M.; Stein, H.J.; Siewert, J.R.; Höfler, H.; Sarbia, M. Prognostic significance of expression patterns of c-erbB-2, p53, p16INK4A, p27KIP1, cyclin D1 and epidermal growth factor receptor in oesophageal adenocarcinoma: A tissue microarray study. J. Clin. Pathol. 2006, 59, 631–634. [Google Scholar] [CrossRef] [Green Version]
  102. Zhang, K.; Li, J.-T.; Li, S.-Y.; Zhu, L.-H.; Zhou, L.; Zeng, Y. Integration of human papillomavirus 18 DNA in esophageal carcinoma 109 cells. World J. Gastroenterol. 2011, 17, 4242–4246. [Google Scholar] [CrossRef] [PubMed]
  103. Boon, S.S.; Chen, Z.; Li, J.; Lee, K.Y.C.; Cai, L.; Zhong, R.; Chan, P.K.S. Human papillomavirus type 18 oncoproteins exert their oncogenicity in esophageal and tongue squamous cell carcinoma cell lines distinctly. BMC Cancer 2019, 19, 1211. [Google Scholar] [CrossRef] [Green Version]
  104. IARC Monographs Priorities Group. Advisory Group recommendations on priorities for the IARC Monographs. Lancet Oncol. 2019, 20, 763–764. [Google Scholar] [CrossRef] [PubMed]
  105. Gibson, M.K. Epidemiology and Pathobiology of Esophageal Cancer; Tanabe, K.K., Savarese, D.M.F., Eds.; UpToDate: Waltham, MA, USA, 2022; Available online: https://www.uptodate.com/contents/epidemiology-and-pathobiology-of-esophageal-cancer (accessed on 23 January 2022).
  106. Spechler, S.J.; Souza, R.F. Barrett’s Esophagus. N. Engl. J. Med. 2014, 371, 836–845. [Google Scholar] [CrossRef]
  107. Li, S.; Luk, H.Y.; Xia, C.; Chen, Z.; Chan, P.K.S.; Boon, S.S. Oesophageal carcinoma: The prevalence of DNA tumour viruses and therapy. Tumour Virus Res. 2021, 13, 200231. [Google Scholar] [CrossRef] [PubMed]
  108. White, J.R.; Ragunath, K.; Whitton, A.; Marsh, E.; Kaye, P.; Knight, G. Study to investigate the prevalence of human papillomavirus in Barrett’s oesophagus using a novel screening methodology. BMJ Open Gastroenterol. 2022, 9, e000840. [Google Scholar] [CrossRef]
Figure 1. E6/E7 oncogene analysis of hr-HPV 16 and 18 using a novel in-situ hybridization detection technique (RNAScope) in OAC (A) and Barrett’s dysplastic (B) tissue. Positive staining is diffusely present in the nuclei and cytoplasm as punctate/granular appearance. Oncogene activity is only evident in columnar cells. No staining seen in adjacent squamous tissue (B).
Figure 1. E6/E7 oncogene analysis of hr-HPV 16 and 18 using a novel in-situ hybridization detection technique (RNAScope) in OAC (A) and Barrett’s dysplastic (B) tissue. Positive staining is diffusely present in the nuclei and cytoplasm as punctate/granular appearance. Oncogene activity is only evident in columnar cells. No staining seen in adjacent squamous tissue (B).
Cancers 15 00873 g001
Table 1. Studies included in systematic review by Kunzman et al. (with permission) [35].
Table 1. Studies included in systematic review by Kunzman et al. (with permission) [35].
Geographic RegionNo. of Samples TestedOACBONo. of HPV-Positive SamplesHPV Prevalence (%)No. of Reports
USA23116233916.88
UK141687396.385
Germany2118083.794
Australia340228112226.42
Italy232303132
Turkey29290620.72
Mexico4517284293.31
Netherlands484801122.91
Iran4041251
Sweden27270518.51
South Africa110001
China5700001
Korea330001
France40400001
India550001
OAC, esophageal adenocarcinoma, BO, Barrett’s esophagus.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rajendra, S.; Sharma, P. Causal Link of Human Papillomavirus in Barrett Esophagus and Adenocarcinoma: Are We There Yet? Cancers 2023, 15, 873. https://doi.org/10.3390/cancers15030873

AMA Style

Rajendra S, Sharma P. Causal Link of Human Papillomavirus in Barrett Esophagus and Adenocarcinoma: Are We There Yet? Cancers. 2023; 15(3):873. https://doi.org/10.3390/cancers15030873

Chicago/Turabian Style

Rajendra, Shanmugarajah, and Prateek Sharma. 2023. "Causal Link of Human Papillomavirus in Barrett Esophagus and Adenocarcinoma: Are We There Yet?" Cancers 15, no. 3: 873. https://doi.org/10.3390/cancers15030873

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop