HPV-p16 - bpa

advertisement

Human Papillomavirus in Cervical and Head-and-Neck Cancer

CME

Amanda Psyrri, MD Daniel DiMaio, MD

Nat Clin Pract Oncol 5(1):2431, 2008. © 2008 Nature Publishing Group

Summary and Introduction

Summary

Cervical cancer is a major cause of cancer mortality in women worldwide and is initiated by infection with high-risk human papillomaviruses (HPVs). High-risk

HPVs, especially HPV-16, are associated with other anogenital cancers and a subgroup of head-and-neck cancers. Indeed, HPV infection could account for the development of head-and-neck cancer in certain individuals that lack the classical risk factors for this disease (tobacco and alcohol abuse). This Review summarizes the main events of the HPV life cycle, the functions of the viral proteins, and the implications of HPV infection on their hosts, with an emphasis on carcinogenic mechanisms and disease outcomes in head-and-neck cancer. The demonstration that HPVs have a role in human carcinogenesis has allowed the development of preventive and therapeutic strategies aimed at reducing the incidence and mortality of HPV-associated cancers.

Introduction

Papillomaviruses are small non-enveloped DNA viruses that infect squamous epithelial cells. More than 200 papillomavirus types have been isolated and there are certainly additional types that have not yet been identified. These viruses have been found in many organisms, including humans. Human papillomaviruses (HPVs) give rise to a large spectrum of epithelial lesions, mainly benign hyperplasia (e.g. warts or papillomas) with low malignant potential.

There is a subgroup of HPVs, the ‘high-risk’ HPVs, which are associated with precancerous lesions. A small fraction of people infected with high-risk HPVs will develop cancers, which usually arise many years after the initial infection.

On the basis of epidemiological and molecular evidence, in 1995, the International Agency for Research on Cancer recognized that the highrisk HPV types 16 and 18 were carcinogenic in humans.

[1] Together, these two HPV types are responsible for approximately 70% of cervical cancer cases, and other high-risk

HPV types account for virtually all of the remaining cases of this disease.

[2] Cancer of the uterine cervix is, therefore, the malignancy most widely accepted as being associated with HPV infection. In addition, high-risk HPVs are associated with other anogenital carcinomas, including vulvar, anal, and penile cancers [3,4] and, as described in detail herein, some head-and-neck squamous cell carcinomas (HNSCCs).

[5]

High-risk HPVs cause cervical dysplasia and carcinoma in situ , which if locally confined can be successfully treated but if left untreated can progress to cervical carcinoma. There is usually a latency period of 10 years or more between the initial high-risk HPV infection and the development of cervical cancer.

[6]

This prolonged natural history allows preinvasive cervical lesions to be diagnosed using Pap smear screening programs. These screening programs have significantly decreased the incidence of cervical cancer in the developed world, but have not been widely adopted in the developing world. This inability to establish effective Pap smear programs in certain parts of the world contributes to the high incidence of cervical disease in these areas. Presumably, the combination of Pap smear screening with HPV-DNA testing will improve our ability to stage and manage cervical lesions. In addition, prevention of high-risk

HPV infection by prophylactic vaccination is likely to prevent many HPV-associated cancers.

HPV-associated cancers maintain and express the HPV viral genes even in advanced stages of disease, and repression of viral oncogene expression can prevent the growth or survival of cervical cancer cells. This finding raises the possibility that even late-stage cancers can be cured by HPV-targeted strategies, such as medicines that interfere with the expression or action of viral proteins and therapeutic vaccines that elicit a cytolytic immune response to cells expressing these proteins. This Review describes the principles of HPV-induced carcinogenesis in relation to cervical and head-and-neck carcinomas.

Life Cycle of the Papillomaviruses

The HPV genome is a double-stranded circular DNA molecule of 8,000 base pairs (bp) that encodes up to ten proteins. Only one DNA strand is transcribed into mRNA. The genome is divided into three portions: a

4,000 bp region that encodes proteins primarily involved in viral DNA replication and cell transformation; a ∼ 3,000 bp region that encodes the structural proteins of the virus particles; and a ∼ 1,000 bp noncoding region that contains the origin of viral

DNA replication and transcriptional regulatory elements.

[7]

Through wounds or abrasions, the papillomaviruses infect basal epithelial cells, which are the only actively dividing cells in the epithelial layer. The viral DNA is maintained in the nuclei of infected basal epithelial cells as a low-copy-number plasmid.

[8] Squamous epithelial cells normally undergo differentiation as they move from the basement membrane towards the surface epithelium, and HPV-DNA replicates to a high copy number only in terminally differentiated cells near the epithelial surface.

[8,9] Similarly, the late viral genes, which encode the L1 and L2 proteins that constitute the virus particle, are expressed only in the highly differentiated cells, where infectious progeny virus is produced and released.

[8]

Replication of the HPV genome is critically dependent on the host-cell DNA replication machinery, as is seen with other small DNA viruses. Although terminally differentiated epithelial cells are normally not able to support DNA synthesis, viral DNA replication takes place in these cells — HPVs encode E6 and E7 proteins that create a state competent for DNA replication.

[10] The E6 protein of the high-risk HPV binds and induces the degradation of the p53 tumor suppressor protein via a ubiquitin-mediated process, [11,12] while the HPV-E7 protein binds and destabilizes the retinoblastoma (Rb) tumor suppressor protein and related proteins.

[13,14] The E6 and E7 proteins also interact with other cellular targets.

[7] Together, these effects promote cell-cycle progression and viral

DNA replication in differentiated keratinocytes.

[15,16]

The papillomavirus E1 and E2 proteins are required for viral DNA replication and papilloma formation.

[17] E1 is an ATP-dependent helicase that initiates viral replication in cooperation with the E2 protein.

[18-20] In addition, the E2 protein can function as a transcriptional repressor of E6 and E7 oncogene expression, [21] and has an essential role in the segregation of viral DNA plasmids as cells divide through its association with cellular chromatin-associated proteins.

[22,23]

HPV and Cervical Carcinoma

The observation in 1842, in Italy, that cervical cancer developed almost exclusively in married women and was rare in nuns, was the first hint that a sexually transmitted agent was associated with the pathogenesis of cervical cancer.

[24]

Harald zur Hausen’s laboratory was the first to demonstrate that cervical cancer biopsies and cervical cancer cell lines contained HPV-DNA sequences.

[25-27] HPV-16 is the most prevalent high-risk HPV type and is found in approximately

50% of cervical cancer cases. HPV-18 is the second most prevalent high-risk HPV, with other HPV types being isolated from fewer cases of cervical cancer.

[2]

Several lines of evidence indicate that high-risk HPV types have an essential role in cervical carcinogenesis. First, almost all cervical carcinomas contain and express high-risk HPV sequences, frequently in an integrated form.

[2,28,29] Second, epidemiological studies demonstrated that persistent infection with a highrisk HPV dramatically increases the risk of cervical carcinoma.

[28,30] Third, molecular and cellular studies demonstrated that high-risk HPVs encode the E6 and

E7 oncoproteins, which interfere with well-established tumor suppressor pathways.

[7,31] Fourth, the E6 and E7 oncogenes are continuously expressed in cancer cells and are required for proliferation and survival of cervical cancer cell lines.

[7,32-34] Fifth, transgenic mice expressing high-risk HPV sequences develop cervical carcinoma following exposure to estrogen.

[35,36] Finally, vaccination that prevents persistent high-risk HPV-16 and HPV-18 infection in women also prevents the development of precancerous cervical lesions.

[37-39]

The continuous expression of high-risk E6 and/or E7 genes causes genomic instability, due in part to the ability of the viral proteins to disrupt tumor suppressor pathways.

[7,40] In addition, the E6 oncoprotein interferes with DNA repair enzymes, and the E7 oncoprotein can induce structural and numerical chromosome abnormalities by the disruption of centrosome synthesis.

[41,42] This genetic instability can cause the emergence of tumorigenic cells. Despite the accumulation of cellular mutations during carcinogenesis, repression of E6 and E7 expression in cervical carcinoma cell lines results in restoration of Rb and p53 activity and is sufficient to induce cell growth arrest or apoptosis.

[32,33,43]

HPV and Head-and-Neck Carcinoma

In addition to the strong association between HPV infection and cervical carcinoma, mounting epidemiological, molecular and clinical evidence indicates that high-risk HPVs (especially HPV.16) account for the development of headand-neck carcinoma in some individuals who do not have the classical risk factors for this disease (i.e. a history of tobacco use and/or alcohol consumption).

[5,44.47] According to Surveillance, Epidemiology, and End Results (SEER) data, the annual incidence of base-of-tongue and tonsil cancers in the USA increased by 2.1% and 3.9%, respectively, from 1973 to 2001 among white men and women aged 20-44 years, whereas incidences at other sites declined.

[48,49] Similarly, the incidence of tonsillar cancer increased by approximately 2-3% per year among men younger than 60 years from 1975 through 1998.

[50] This increase in the incidence of oropharyngeal cancer was paralleled by an increase in certain sexual behaviors. In the US, between the periods 1976-1980 and 1988-1994, herpes simplex-2 seroprevalence, a surrogate marker of sexual activity, increased by 30%.

[51] This change in the demographics of patients with head-and-neck cancer is consistent with a role for genital HPVs in the pathogenesis of oropharyngeal squamous cell carcinoma (OSCC) in individuals whose sexual practices are typically associated with sexual transmission of the virus. Several studies suggest that oral HPV infection is sexually acquired.

[44,52] A recent hospital-based case-control study of 100 newly diagnosed patients with OSCC and

200 control patients without cancer revealed that a high (i.e. 26 or more) lifetime number of vaginal-sex partners and 6 or more lifetime oral-sex partners were associated with OSCC (odds ratios 3.1 and 3.4, respectively).

[44] The authors conclude that there is a strong association between oral HPV infection and

OSCC among subjects with or without tobacco or alcohol use, the established risk factors for this disease. Although oral —genital contact may be responsible for HPV transmission, transmission through direct mouth-to-mouth contact or other means could not be excluded. An increased risk of HPV-associated OSCC in individuals with a history of HPV-associated anogenital cancers and in husbands of women with in situ carcinoma and invasive cervical cancer also suggests that sexual transmission of HPV infection to the oral cavity can occur.

[47,53]

Markers of HPV infection are also associated with increased risk of OSCC.

[44,54,55]

D’Souza et al . showed that OSCC was significantly associated with oral HPV infection and with HPV-16 L1 seropositivity among patients both with and without a history of heavy tobacco and alcohol use.

[44] In a Swedish study, oral highrisk HPV infection was associated with dramatically increased risk of OSCC development (odds ratio 230; 95% CI 44-1,200), after adjustment for alcohol and tobacco use.

[55] In a separate, nested case-control study, HPV-16-seropositivity conferred a greater than 14-fold increased risk of subsequent development of

OSCC.

[54] Studies that failed to show an association between sexual habits, oral HPV infection and HNSCC include cohorts with a less than 25% incidence of

HPVDNA positivity, where the risk association might be attenuated.

[52,56] Studies that restricted enrollment to patients with OSCC showed a clear association between sexual behaviors, oral HPV infection and HNSCC.

[44,46,57] Tonsillar crypts seem particularly susceptible to transformation by HPV, which is similar to the transformation zone of the uterine cervix, the location in which most cervical cancers originate.

[58]

Establishing the link between HPV and a subset of OSCC has been difficult because of the heterogeneity of OSCC, and the fact that only a fraction of cases are HPVassociated. Syrjänen et al . observed that some oral squamous cell cancers have morphological and immunohistochemical features indicative of

HPV infection.

[59] This finding was the first hint that HPV may be involved in the pathogenesis of a subset of HNSCC.

[59] Since HPV-16 DNA was first detected in an invasive HNSCC in 1985, [60] HPV sequences have been repeatedly detected in a variable proportion of HNSCC, from as few as 10% to 100%.

[61] This disparity might reflect the different anatomic locations of tumors and the techniques used to detect HPV-DNA. HPV-associated OSCC tend to be poorly differentiated, often basaloid in histology, and frequently present at an advanced stage.

HPV-16 is the most prevalent genotype in cervical carcinoma, and is also the most frequently detected HPV type in HNSCC, found in up to 90% of HPVpositive cases.

[5,45] Some reports have indicated that the HPV subtypes associated with OSCC are similar but not identical to HPVs found in cervical carcinoma.62-64 Chen et al . showed that HPV-associated HNSCC harbor HPV-16 subtypes with characteristic changes in their promoter/enhancer region that render them particularly active in oral keratinocytes.

[62]

Transcription of HPV-16 E6/E7 mRNA in tonsillar carcinomas is not necessarily dependent on viral DNA integration, and the viral DNA is predominately in episomal form.

[65] It is not clear how the viral DNA can remain in cancer tissues as episomes with high copy numbers. A report by Van Tine and coauthors provided evidence that the HPVE2 protein might serve as an ‘anchor’ to bind episomal HPV to cellular mitotic spindles, thus, ensuring maintenance of the episomal state.

[66]

Clinical Implications of Hpvassociated Head-and-Neck

Carcinoma

In a mechanism similar to that seen in cervical cancer, viral oncoprotein-mediated abrogation of the p53 and pRb pathways obviates the need for mutational inactivation of TP53 and RB1 genes in HPV-associated OSCC. HPV-induced cancers are, therefore, associated with wild-type TP53 and RB1 genes and low levels of p53 and pRb proteins. The p16 tumor suppressor gene (CDKN2A) is also often upregulated in these cancers because it is negatively regulated by pRb.

[67,68] Overexpression of p16 has been repeatedly reported in HPV-associated cancers. In one study of cervical and genital lesions, high levels of p16 protein expression were associated with high-risk HPV infection.

[69] A recent study of lymph-node metastases in HNSCC reported that p16 protein overexpression is a surrogate marker for oropharyngeal origin and HPV-association.

[70] By contrast, loss of p16 protein expression is a common and early event in tobacco-related HNSCC. Thus, tobacco/alcohol-associated OSCC are associated with downregulation of p16 protein and TP53 gene mutation, [71] whereas HPV-associated OSCC are associated with wild-type TP53 and RB1 genes and upregulation of p16 protein levels.

Several lines of clinical evidence also suggest that HPV-associated OSCC could be biologically distinct from classical OSCC.

[45,72] Tobacco-associated OSCC are more frequent in men, while men and women are at equal risk of HPV-associated OSCC. In addition, patients with HPV-associated OSCC are often nonsmokers and nondrinkers and on average 5 years younger than their tobacco-use-associated counterparts.

[58]

HPV-DNA detection per se in an OSCC does not prove causal association. Only HPV DNA that is transcriptionally active is biologically and clinically relevant.

To address the role of HPV-16 expression in OSCC, we studied a cohort of 107 OSCC samples for HPV-16-DNA viral load using real-time PCR.

[45] In addition, we constructed a tissue array composed of these tumors and studied expression of p53, pRb and p16 using a quantitative in situ method of protein analysis. We hypothesized that for HPV-DNA-positive cases, p16 expression status would identify those that were biologically relevant. These experiments delineated three biologically and clinically distinct types of OSCC: class I, HPV-negative/p16 nonexpressing; class II, HPV-positive/p16 nonexpressing; and class III, HPV-positive/p16 expressing oropharyngeal tumors. Only patients in class III had significantly lower p53 and pRb expression. The 5-year survival in class III was 79%, significantly higher than in the other two classes (20% and 18%, P = 0.0095). Diseasefree survival for class III was 75% compared with

15% and 13% for classes I and II, respectively (P = 0.0025). The 5-year local recurrence was 14% in class III compared with 45% and 74% (P = 0.03).

Multivariate survival analysis confirmed the prognostic value of the threeclass model. Thus, only the HPV-positive/p16 expressing OSCC tumors (class III) fit the cervical carcinogenesis model, and these tumors are associated with a favorable prognosis.

The classic oncogenic insult in HNSCC has been attributed to excess alcohol or tobacco exposure producing mutational or epigenetic inactivation of TP53, p16 and RB1 in a multistep progression from normal cell to dysplasia to carcinoma. On the basis of our results, we can refine a model for HPV-associated oropharyngeal cancer.

[72] In these HPV-induced tumors, oncogenic HPV-E6 and HPV-E7 proteins inactivate p53 and pRb pathways, with subsequent upregulation of p16 expression. This model eliminates the need for mutational inactivation of the RB1 and TP53 genes and the pathways they control. In addition to the previously described models, [72] we also describe a novel class II of OSCC, where HPV-16 DNA is present, but p53, p16 and pRb expression is similar to class I tumors that lack HPV-DNA.

[45] These class II tumors might arise when tobacco/alcohol-related tumors are infected by high-risk HPVs. It is not clear whether these tumors represent a group that is biologically distinct from HPV-negative tumors.

HPV-associated OSCC are associated with a better prognosis than HPV-negative tumors in the majority of studies.

[5,73.78] In our study, HPVpositivity confers a

60% to 80% reduction in risk of death from cancer compared with HPVnegative tumors.

[45] What are the clinical implications of a role of sexually transmitted

HPV in a subset of head-and-neck cancers with a relatively favorable prognosis? Prophylactic vaccines that prevent persistent cervical HPV.16 infections might be effective in preventing these cases of head-and-neck cancer as well, either indirectly by eliminating an anogenital source of virus or directly by protecting the oropharyngeal epithelium itself from infection. Similar benefits could arise from strategies that inhibit viral replication; for example, from drugs that block the ability of the E1 and E2 proteins to drive viral DNA replication. In addition, viral etiology might provide opportunities to treat established cancers.

Thus, antiviral measures being developed to treat cervical cancer may well be effective in HPV-associated OSCC.

Detection of high-risk E6/E7 mRNA or protein would be the ideal test for classifying a tumor as truly HPV-associated, but this determination is not feasible in formalin-fixed, paraffin-embedded tissue. Determination of p16 expression status by immunohistochemistry could serve as a reasonable surrogate marker for biologically relevant high-risk HPV infection. The addition of p16 immunohistochemistry to existing protocols to determine HPV DNA presence may, therefore, prove useful in the classification of OSCC.

The favorable outcome of HPV-induced oropharyngeal cancers might be attributable to the absence of field cancerization or enhanced radiation sensitivity.

[77]

The term ‘field cancerization’ is used to describe the presence of carcinogeninduced early genetic changes in the epithelium from which multiple independent lesions arise, leading to the development of multifocal tumors.

[79] In addition, unlike tobacco-associated oropharyngeal cancers that harbor mutant TP53, the apoptotic response of HPV-associated tumors to radiation and chemotherapy might be intact, as seems to be the case in some cervical cancers.

[80] It might be important to distinguish HPV-associated HNSCC with favorable prognosis from poor prognosis tobacco/alcohol-associated HNSCC, especially as specific anti-HPV treatment becomes available. Careful clinical trials are required to determine the optimum management of HPV-associated HNSCC, compared with

HPV-independent HNSCC. The diagnosis of HPV-associated OSCC should be considered in all OSCCs, especially those arising from the tonsils in patients without a history of tobacco use or alcohol abuse, and in immunocompromised patients and those with basaloid or poorly differentiated tumors.

Prophylactic and Therapeutic HPV Vaccination

As described in detail in a recent review, prophylactic HPV vaccines have been developed on the basis of recombinant expression and selfassembly of the major capsid protein, L1, into immunogenic virus-like particles that resemble authentic virions but are noninfectious.

[81] One of the vaccines, Gardasil® (Merck

& Co., Whitehouse Station, NJ) protects against HPV types 6, 11, 16 and 18, and the other vaccine, Cervarix® (GlaxoSmithKline, Rixensart, Belgium) protects against HPV types 16 and 18. HPV.6 and HPV-11 are low-risk virus types that cause approximately 90% of genital warts. Several randomized placebo-controlled trials in human volunteers demonstrated that these prophylactic vaccines elicited high levels of neutralizing antibody and significantly reduced the incidence of persistent HPV-16 and HPV-18 infections and associated moderate-to-high grade cervical intraepithelial neoplasia CIN2/3.

[37-39]

Followup for 4.5 years demonstrated 100% efficacy in the prevention of HPV-16- and HPV-18 CIN2/3 and persistent infections among subjects that were strictly adherent to the study protocol.

[82] Prophylactic vaccination against high-risk HPV types will eventually prevent a significant number of cervical carcinomas, although owing to the prolonged course of cervical carcinogenesis, reduction in cancer incidence will not be profound for several years. These vaccines might also reduce the incidence of HPV-associated oropharyngeal cancers, as well as cancers of the anus, vulva, vagina, and penis. Indeed, the presence of HPV-16-DNA in a relatively high fraction of HPV-associated HNSCC suggests that the currently available vaccines may be particularly effective in preventing this disease.

Despite the promise of these vaccines, there are several issues that still need to be addressed. First, the duration of protection is unknown. Second, virus-like particle vaccines are relatively expensive, and vaccine delivery in the developing world will be difficult. Third, the vaccines will only protect against the highrisk HPV types that the vaccine targets (i.e. HPV-16 and HPV.18) and women must, therefore, continue to undergo Pap smear screening, because they could still be infected by high-risk HPV types that are not included in these vaccines. Finally, these prophylactic vaccines are likely to provide limited benefits to women already persistently infected with high-risk HPV.

[82]

Therapeutic HPV vaccination strategies are also being developed. The high-incidence of cervical cancer in immunosuppressed individuals suggests that cellmediated immunity has a major role in controlling cervical carcinogenesis. The generation of a cell-based immune response to E6 and/or E7 might, therefore, result in destruction of HPV-associated precancerous and cancerous cells that express these viral proteins. Various therapeutic vaccination strategies can elicit a cytolytic CD8 + T-cell response directed against high-risk HPV-E6 and/or HPV.E7 oncoproteins in humans and mice.

[83] In mice, this practice leads to a decrease in the growth of transplantable tumors expressing these viral antigens.

[84] This approach, however, has not yet been successful against CIN and cervical carcinoma in humans.

[85]

Conclusions

An improved mechanistic understanding of the virologic basis for cervical and some head-andneck and other anogenital cancers will help to prevent and treat these diseases. The widespread use of prophylactic HPV vaccines combined with vigilant Pap smear and HPV-DNA testing has the potential to eliminate many of these cancers. Molecular classification of tumors is likely to provide important new information that will allow a better estimate of prognosis and may well influence treatment decisions. In the future, antiviral pharmaceutical approaches and therapeutic vaccination may allow effective, nontoxic therapy. We are optimistic that the tools to control these cancers will soon be available.

Key Points

High-risk human papillomaviruses encode E6 and E7 oncoproteins that bind and degrade p53 and pRb tumor suppressors, respectively

HPV-associated cancers maintain and express the HPV viral genes even in advanced stages of disease, and repression of viral oncogene expression can prevent the growth and survival of cervical cancer cells

The annual incidence of oropharyngeal cancer in the US increased between 1973 and 2001 in younger individuals, in parallel with an increase in sexual practices that are typically associated with the sexual transmission of HPV

HPV-16 is the most prevalent genotype in cervical carcinoma, and is also the most frequently detected HPV type in HNSCC, found in up to 90% of HPV-positive cases

HPV-associated oropharyngeal squamous-cell cancers have a better prognosis than HPVnegative tumors

Prophylactic vaccination against high-risk HPV types will eventually prevent a significant number of cervical carcinomas and possibly HNSCC, although owing to the prolonged course of carcinogenesis, the reduction in cancer incidence will not be profound for several years

Acknowledgements

Charles P Vega, University of California, Irvine, CA, is the author of and is solely responsible for the content of the learning objectives, questions and answers of the Medscapeaccredited continuing medical education activity associated with this article.

Funding

Research in the authors’ laboratories was supported by grants from the NIH (CA16038) and the Virginia Alden Wright Fund.

Reprint Address

Amanda Psyrri, Departments of Medicine/Oncology, Yale Cancer Center, Yale University School of Medicine, PO Box 208020, New Haven, CT 06520-8020,

USA diamando.psyrri@yale.edu

References

1.

2.

3.

4.

5.

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans (1995) Human papillomaviruses. IARC Monogr Eval Carcinog Risks

Hum 64: 1-378 zur Hausen H (2002) Papillomaviruses and cancer: from basic studies to clinical application.

Crum CP et al . (1997) Pathobiology of vulvar squamous neoplasia.

Nat Rev Cancer

Curr Opin Obstet Gynecol 9: 63-69

2: 342-350

Kayes O et al . (2007) Molecular and genetic pathways in penile cancer. Lancet Oncol 8: 420-429

Gillison ML et al . (2000) Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer

Inst 92: 709-720

Goldie SJ et al . (2003) A comprehensive natural history model of HPV infection and cervical cancer to estimate the clinical impact of a 6.

7.

8.

9. prophylactic HPV-16/18 vaccine. Int J Cancer 106: 896-904

Munger K et al . (2004) Mechanisms of human papillomavirus-induced oncogenesis. J Virol 78: 11451-11460

Stubenrauch F and Laimins LA (1999) Human papillomavirus life cycle: active and latent phases. Semin Cancer Biol 9: 379-386

Bedell MA et al . (1991) Amplification of human papillomavirus genomes in vitro is dependent on epithelial differentiation. J Virol 65: 2254-2260

10. Cheng S et al . (1995) Differentiation-dependent up-regulation of the human papillomavirus E7 gene reactivates cellular DNA replication in suprabasal differentiated keratinocytes. Genes Dev 9: 2335-2349

11. Werness BA et al . (1990) Association of human papillomavirus types 16 and 18 E6 proteins with p53. Science 248: 76-79

12. Scheffner M et al . (1990) The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 63:

1129-1136

13. Dyson N et al . (1989) The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243: 934-937

14. Boyer SN et al . (1996) E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res 56: 4620-4624

15. Flores ER et al . (2000) The human papillomavirus type 16 E7 oncogene is required for the productive stage of the viral life cycle. J Virol 74:

6622-6631

16. McLaughlin-Drubin ME et al . (2005) The role of the human papillomavirus type 18 E7 oncoprotein during the complete viral life cycle. Virology

338: 61-68

17. Wu X et al . (1994) Papilloma formation by cottontail rabbit papillomavirus requires E1 and E2 regulatory genes in addition to E6 and E7 transforming genes. J Virol 68: 6097-6102

18. Yang L et al . (1993) The E1 protein of bovine papilloma virus 1 is an ATP-dependent DNA helicase. Proc Natl Acad Sci USA 90: 5086-5090

19. Hughes FJ and Romanos MA (1993) E1 protein of human papillomavirus is a DNA helicase/ATPase. Nucl Acids Res 21: 5817-5823

20. Mohr IJ et al . (1990) Targeting the E1 replication protein to the papillomavirus origin of replication by complex formation with the E2 transactivator. Science 250: 1694-1699

21. Thierry F and Yaniv M (1987) The BPV1-E2 trans-acting protein can be either an activator or a repressor of the HPV18 regulatory region. EMBO

J 6: 3391-3397

22. Skiadopoulos MH and McBride AA (1998) Bovine papillomavirus type 1 genomes and the E2 transactivator protein are closely associated with mitotic chromatin. J Virol 72: 2079-2088

23. You J et al . (2004) Interaction of the bovine papillomavirus E2 protein with Brd4 tethers the viral DNA to host mitotic chromosomes. Cell 117:

349-360

24. Scotto J and Bailar JC III (1969) Rigoni-Stern and medical statistics: a nineteenth-century approach to cancer research. J Hist Med Allied Sci 24:

65-75

25. Boshart M et al . (1984) A new type of papillomavirus DNA is present in genital cancer biopsies and in cell lines derived from cervical cancer.

EMBO J 3: 1151-1157

26. Durst M et al . (1983) A papillomavirus DNA from a cervical carcinoma and its prevalence in cancer biopsy samples from different geographic regions. Proc Natl Acad Sci USA 80: 3812-3815

27. Gissmann L et al . (1983) Human papillomavirus types 6 and 11 DNA sequences in genital and laryngeal papillomas and in some cervical cancers. Proc Natl Acad Sci USA 80: 560-563

28. Walboomers JM et al . (1999) Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189: 12-19

29. Cullen AP et al . (1991) Analysis of the physical state of different human papillomavirus DNAs in intraepithelial and invasive cervical neoplasm. J

Virol 65: 606-612

30. Schlecht NF et al . (2001) Persistent human papillomavirus infection as a predictor of cervical intraepithelial neoplasia. JAMA 286: 3106-3114

31. Halbert CL et al . (1991) The E7 gene of human papillomavirus type 16 is sufficient for immortalization of human epithelial cells. J Virol 65: 473-

478

32. Hwang ES et al . (1993) Inhibition of cervical carcinoma cell line proliferation by the introduction of a bovine papillomavirus regulatory gene. J

Virol 67: 3720-3729

33. Desaintes C et al . (1997) Expression of the papillomavirus E2 protein in HeLa cells leads to apoptosis. EMBO J 16: 504-514

34. von Knebel Doeberitz M et al . (1988) Correlation of modified human papilloma virus early gene expression with altered growth properties in C4-1 cervical carcinoma cells. Cancer Res 48: 3780-3786

35. Arbeit JM et al . (1994) Progressive squamous epithelial neoplasia in K14-human papillomavirus type 16 transgenic mice.

J Virol 68: 4358-4368

36. Brake T and Lambert PF (2005) Estrogen contributes to the onset, persistence, and malignant progression of cervical cancer in a human papillomavirus-transgenic mouse model. Proc Natl Acad Sci USA 102: 2490-2495

37. Harper DM et al . (2004) Efficacy of a bivalent L1 virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and 18 in young women: a randomised controlled trial. Lancet 364: 1757-1765

38. Villa LL et al . (2005) Prophylactic quadrivalent human papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in young women: a randomised doubleblind placebo-controlled multicentre phase II efficacy trial. Lancet Oncol 6: 271-278

39. Koutsky LA and Harper DM (2006) Chapter 13: current findings from prophylactic HPV vaccine trials. Vaccine 24 (Suppl 3): S114-S121

40. Durst M et al . (1987) Molecular and cytogenetic analysis of immortalized human primary keratinocytes obtained after transfection with human papillomavirus type 16 DNA. Oncogene 1: 251-256

41. Duensing S and Munger K (2004) Mechanisms of genomic instability in human cancer: insights from studies with human papillomavirus oncoproteins. Int J Cancer 109: 157-162

42. Srivenugopal KS and Ali-Osman F (2002) The DNA repair protein, O(6)-methylguanine-DNA methyltransferase is a proteolytic target for the E6 human papillomavirus oncoprotein. Oncogene 21: 5940-5945

43. Goodwin EC and DiMaio D (2000) Repression of human papillomavirus oncogenes in HeLa cervical carcinoma cells causes the orderly reactivation of dormant tumor suppressor pathways. Proc Natl Acad Sci USA 97: 12513-12518

44. D’Souza G et al . (2007) Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356: 1944-1956

45. Weinberger PM et al . (2006) Molecular classification identifies a subset of human papillomavirus-associated oropharyngeal cancers with favourable prognosis. J Clin Oncol 24: 736-747

46. Andl T et al . (1998) Etiological involvement of oncogenic human papillomavirus in tonsillar squamous cell carcinomas lacking retinoblastoma cell cycle control. Cancer Res 58: 5-13

47. Frisch M and Biggar RJ (1999) Aetiological parallel between tonsillar and anogenital squamous-cell carcinomas. Lancet 354: 1442-1443

48. Shiboski CH et al . (2005) Tongue and tonsil carcinoma: increasing trends in the US population ages 20-44 years. Cancer 103: 1843-1849

49. Frisch M et al . (2000) Changing patterns of tonsillar squamous cell carcinoma in the United States. Cancer Causes Control 11: 489-495

50. Canto MT and Devesa SS (2002) Oral cavity and pharynx cancer incidence rates in the United States, 1975-1998. Oral Oncol 38: 610-617

51. Fleming DT et al . (1997) Herpes simplex virus type 2 in the United States, 1976 to 1994. N Engl J Med 337: 1105-1111

52. Schwartz SM et al . (1998) Oral cancer risk in relation to sexual history and evidence of human papillomavirus infection. J Natl Cancer Inst 90:

1626-1636

53. Hemminki K et al . (2000) Tonsillar and other upper aerodigestive tract cancers among cervical cancer patients and their husbands. Eur J Cancer

Prev 9: 433-437

54. Mork J et al . (2001) Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med 344: 1125-

1131

55. Hansson BG et al . (2005) Strong association between infection with human papillomavirus and oral and oropharyngeal squamous cell carcinoma: a populationbased case-control study in southern Sweden. Acta Otolaryngol 125: 1337-1344

56. Herrero R et al . (2003) Human papillomavirus and oral cancer: the International Agency for Research on Cancer multicenter study. J Natl Cancer

Inst 95: 1772-1783

57. Smith EM et al . (2004) Age, sexual behavior and human papillomavirus infection in oral cavity and oropharyngeal cancers. Int J Cancer 108: 766-

772

58. McNeil C (2000) HPV in oropharyngeal cancers: new data inspire hope for vaccines. J Natl Cancer Inst 92: 680-681

59. Syrjanen K et al . (1983) Morphological and immunohistochemical evidence suggesting human papillomavirus (HPV) involvement in oral squamous cell carcinogenesis. Int J Oral Surg 12: 418-424

60. de Villiers EM et al . (1985) Papillomavirus DNA in human tongue carcinomas. Int J Cancer 36: 575-578

61. Steinberg BM and DiLorenzo TP (1996) A possible role for human papillomaviruses in head and neck cancer. Cancer Metastasis Rev 15: 91-112

62. Chen Z et al . (1997) Mutations in the long control region of human papillomavirus DNA in oral cancer cells, and their functional consequences.

Cancer Res 57: 1614-1619

63. Maitland NJ et al . (1987) Detection of human papillomavirus DNA in biopsies of human oral tissue. Br J Cancer 56: 245-250

64. Yeudall WA and Campo MS (1991) Human papillomavirus DNA in biopsies of oral tissues. J Gen Virol 72: 173-176

65. Mellin H et al . (2002) Human papillomavirus type 16 is episomal and a high viral load may be correlated to better prognosis in tonsillar cancer. Int

J Cancer 102: 152-158

66. Van Tine BA et al . (2004) Human papillomavirus (HPV) origin-binding protein associates with mitotic spindles to enable viral DNA partitioning.

Proc Natl Acad Sci USA 101: 4030-4035

67. Li Y et al . (1994) Transcriptional repression of the D-type cyclin-dependent kinase inhibitor p16 by the retinoblastoma susceptibility gene product pRb. Cancer Res 54: 6078-6082

68. Hara E et al . (1996) Regulation of p16CDKN2 expression and its implications for cell immortalization and senescence. Mol Cell Biol 16: 859-867

69. Agoff SN et al . (2003) p16(INK4a) expression correlates with degree of cervical neoplasia: a comparison with Ki-67 expression and detection of high-risk HPV types. Mod Pathol 16: 665-673

70. Begum S et al . (2003) Detection of human papillomavirus in cervical lymph nodes: a highly effective strategy for localizing site of tumor origin.

Clin Cancer Res 9: 6469-6475

71. Sidransky D (1995) Molecular genetics of head and neck cancer. Curr Opin Oncol 7: 229-233

72. Gillison ML and Shah KV (2001) Human papillomavirusassociated head and neck squamous cell carcinoma: mounting evidence for an etiologic role for human papillomavirus in a subset of head and neck cancers. Curr Opin Oncol 13: 183-188

73. Schwartz SR et al . (2001) Human papillomavirus infection and survival in oral squamous cell cancer: a population-based study. Otolaryngol Head

Neck Surg 125: 1-9

74. Paz IB et al . (1997) Human papillomavirus (HPV) in head and neck cancer: an association of HPV16 with squamous cell carcinoma of

Waldeyer’s tonsillar ring. Cancer 79: 595-604

75. Licitra L et al . (2006) High-risk human papillomavirus affects prognosis in patients with surgically treated oropharyngeal squamous cell carcinoma. J Clin Oncol 24: 5630-5636

76. Ritchie JM et al . (2003) Human papillomavirus infection as a prognostic factor in carcinomas of the oral cavity and oropharynx. Int J Cancer 104:

336-344

77. Lindel K et al . (2001) Human papillomavirus positive squamous cell carcinoma of the oropharynx: a radiosensitive subgroup of head and neck carcinoma. Cancer 92: 805-813

78. Li W et al . (2003) Human papillomavirus positivity predicts favourable outcome for squamous carcinoma of the tonsil. Int J Cancer 106: 553-558

79. Slaughter DP et al . (1953) Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer 6: 963-968

80. Butz K et al . (1996) Cellular responses of HPV-positive cancer cells to genotoxic anti-cancer agents: repression of E6/E7-oncogene expression and induction of apoptosis. Int J Cancer 68: 506-513

81. Wheeler CM et al . (2007) Advances in primary and secondary interventions for cervical cancer: human papillomavirus prophylactic vaccines and testing. Nat Clin Pract Oncol 4: 224-235

82. FUTURE II Study Group (2007) Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 356:

1915-1927

83. Pinto LA et al . (2003) Cellular immune responses to human papillomavirus (HPV)-16 L1 in healthy volunteers immunized with recombinant HPV-

16 L1 virus-like particles. J Infect Dis 188: 327-338

84. Feltkamp MC et al . (1993) Vaccination with cytotoxic T lymphocyte epitope-containing peptide protects against a tumor induced by human papillomavirus type 16-transformed cells. Eur J Immunol 23: 2242-2249

85. Borysiewicz LK et al . (1996) A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet 347: 1523-1527

Authors and Disclosures

As an organization accredited by the ACCME, Medscape, LLC requires everyone who is in a position to control the content of an education activity to disclose all relevant financial relationships with any commercial interest. The ACCME defines "relevant financial relationships" as financial relationships in any amount, occurring within the past 12 months, including financial relationships of a spouse or life partner, that could create a conflict of interest.

Medscape, LLC encourages Authors to identify investigational products or off-label uses of products regulated by the US Food and Drug Administration, at first mention and where appropriate in the content.

Author

Amanda Psyrri, MD

Assistant Professor, Departments of Medicine and Oncology, Yale University School of Medicine, New

Haven, Connecticut

Disclosure: Amanda Psyrri, MD, has disclosed no relevant financial relationships.

Daniel DiMaio, MD

Waldemar Von Zedtwitz Professor of Genetics; Professor of Therapeutic Radiology, Yale University

School of Medicine, New Haven, Connecticut

Disclosure: Daniel DiMaio, MD, has disclosed no relevant financial relationships.

CME Author

Charles P. Vega, MD

Associate Professor; Residency Director, Department of Family Medicine,

University of California, Irvine

Disclosure: Charles P. Vega, MD, has disclosed that he has served as an advisor or consultant to Novartis, Inc.

Registration for CME credit, the post test and the evaluation must be completed online.

To access the activity Post Test and Evaluation link, please go to:

http://www.medscape.com/viewprogram/8391_index

Download