search
for
 About Bioline  All Journals  Testimonials  Membership  News


Journal of Cancer Research and Therapeutics
Medknow Publications on behalf of the Association of Radiation Oncologists of India (AROI)
ISSN: 0973-1482 EISSN: 1998-4138
Vol. 7, Num. 3, 2011, pp. 251-255

Journal of Cancer Research and Therapeutics, Vol. 7, No. 3, July-September, 2011, pp. 251-255

Review Article

Human papilloma virus: A new risk factor in a subset of head and neck cancers

Manisha Bisht1, Sampan Singh Bist2

1 Department of Pharmacology, Himalayan Institute of Medical Sciences, Himalayan Institute Hospital Trust University, Jolly- Grant, Dehradun, India
2 Department of E.N.T and Head-Neck Surgery, Himalayan Institute of Medical Sciences, Himalayan Institute Hospital Trust University, Jolly- Grant, Dehradun, India
Correspondence Address: Manisha Bisht, Department of Pharmacology, Himalayan Institute of Medical Sciences, Himalayan Institute Hospital Trust University, Jolly Grant, Doiwala, Dehradun, Uttrakhnad - 248 140, India, manishabisht@yahoo.co.in

Code Number: cr11067

PMID: 22044803

DOI: 10.4103/0973-1482.87004

Abstract

Head and neck cancer is the sixth most common malignancy worldwide. Tobacco smoking and alcohol consumption are two well known behavioral risk factors associated with head and neck cancer. Recently, evidence is mounting that infection with human papilloma virus, most commonly human papilloma virus-16 is responsible for a subset of head and neck squamous cell carcinoma especially tumors of tonsillar origin. The molecular pathway used by human papilloma virus to trigger malignant transformation of tissue is different from that of other well known risk factors, i.e. smoking and alcohol, associated with squamous cell carcinoma. Apparently, these subsets of patients with human papilloma virus positive tumor are more likely to have a better prognosis than human papilloma virus negative tumor. Considering this fact, the human papilloma virus infection should be determined in all oropharyngeal cancers since it can have a major impact on the decision making process of the treatment.

Keywords: Human papilloma virus, oropharyngeal carcinoma, squamous cell carcinoma

Introduction

Almost 6.5 lakh patients worldwide are diagnosed with head and neck cancer each year, and nearly 90% of these cancers are squamous-cell carcinomas. [1] Smoking and alcohol are the two main causative factors accounting for approximately 80% of oral, oropharyngeal and laryngeal carcinomas. The association between human papilloma virus (HPV) and head and neck squamous cell carcinoma (HNSCC) has been under investigation for at least 20 years. The morphological similarity of genital and oral HPV associated lesions was one of the early findings that raised the possibility that HPV might be involved in oral and laryngeal squamous cell carcinomas (SCCs). This fact was first postulated in 1983, and later confirmed in 1985. [2] Since then, there is mounting evidence that points to a much stronger association between oropharyngeal carcinomas, especially those of tonsillar origin, and infection with high risk strain of HPV, most commonly HPV-16. [3] This review describes the principles of HPV induced carcinogenesis in HNSCC and its clinical implications.

Human Papilloma Virus

HPVs are small non-enveloped Deoxyribonucleic acid (DNA) viruses that show particular affinity for human squamous cell epithelium. There are more than 120 different identified strains of HPV. [4] HPVs give rise to a large spectrum of epithelial lesions, ranging from benign hyperplasia (e.g. warts or papillomas) with low malignant potential to precancerous lesions having high malignant potential. HPV-1, 2 and 4 are associated with common warts, whereas HPV-6 and 11 are associated with respiratory papillomas. [5] There is a subgroup of HPVs, the ′high-risk′ HPVs (HPV-16, 18, 33 and 35) which are associated with precancerous lesions. [4] In 1995, the International Agency for Research on Cancer, on the basis of epidemiological and molecular evidence, recognized that mainly two HPV types 16 and 18 were carcinogenic in humans. [6] The genome of HPV consists of double stranded DNA enclosed in a viral capsid and its replication is critically dependent on the host-cell DNA replication machinery. HPV DNA can exist within the human cell in an integrated or an episomal form. Either state can lead to malignant progression. [4] The HPVs genome encodes for several different proteins. L1 and L2 code for the viral capsid proteins while E6 and E7 proteins create a state competent for DNA replication. The oncogenic potential of HPV is attributed to its ability to insert E6 and E7 into host cellular genomes. As a result of this integration, the function of two tumor suppressor genes is abrogated. The E6 protein binds and induces the degradation of the p53 tumor suppressor protein via a ubiquitin-mediated process, while the HPV-E7 protein binds and destabilizes the retinoblastoma (Rb) tumor suppressor protein. [7],[8] This in turn leads to defects in apoptosis, DNA repair mechanism, cell cycle regulation and finally to cellular immortalization, thus inducing the malignant phenotype. L1 protein forms viral capsid proteins that trigger an immune reaction against the virus, but do not contain the rest of the harmful viral genome. This property is being exploited to produce a vaccine against HPV. [5]

Molecular Mechanism of Action

Functional disruption of tumor suppressor genes p53 and pRb are common finding in SCC. However, there is a difference between the specific mechanism by which HPV inactivates p53 and pRb as compared to the other well known etiological agents of SCC-tobacco and alcohol. Excess alcohol or tobacco exposure produces mutational or epigenetic inactivation of TP53, p16 and RB1 genes in a multistep progression from normal cell to dysplasia to carcinoma. Whereas in HPV-induced tumors, HPV-E6 and E7 proteins inactivate p53 and pRb pathways, without inactivation of their genes, and since pRb negatively regulates p16 expression there is upregulation of its expression. Thus, tobacco/alcohol-associated SCC is associated with downregulation of p16 protein and TP53 gene mutation, whereas HPV-associated SCC is associated with wild-type TP53 and RB1 genes and upregulation of p16 protein levels. [9] In fact, p16 levels are so consistently over expressed that it has been proposed as a surrogate marker for high risk HPV infection that could replace the labour of intensive and expensive process of HPV DNA detection and typing. [10]

Epidemiology

A wide range of HPV prevalence (0-100%), along with the presence of HPV in normal oral mucosa have been reported in literature. The prevalence of HPV in HNSCC ranges from 11% to 44%. [11] A recent systematic review of literature revealed that the overall prevalence of HPV infection was 25.9% in HNSCC, and it was significantly higher among patients with oropharyngeal SCC (35.6%) as compared to those with oral (23.5%) or laryngeal (24.0%) SCC. [12] When HPV presence is further stratified by tumor site, HPV has been found in 17% - 40% of base of tongue tumors, and in 21-100% of tonsillar tumors with an average prevalence in tonsillar tumor of 50%. [3],[4],[11] In contrast, only 2.3% to 25% of oral cavity SCCs are reported as being positive for HPV. [11] HPV-16 is the most frequently detected HPV type in HNSCC, found in up to 90% of HPV-positive cases. [13] Although HPV-16 is usually found in isolation, there may be concurrent infection with other oncogenic HPV strains such as 18, 33 and 35. HPV-18 has been found in 6-11%, whereas HPV-33 has been isolated in 1-4% of oropharyngeal SCCs. HPV- 35 has been found even less frequently. [14] There also seems to be geographic variation as to which HPV type is most common in tumors. In Greece, HPV-18 was the most common strain isolated, whereas in India, HPV-16 and 18 were found at equal rates. [3]

Route of Oral Human Papilloma Virus Transmission

The mode of HPV transmission in HNSCC is not very clear. Various theories have proposed and multiple pathways of transmission have been suggested, including perinatal transmission, autoinfection from oral genital contact by hand and sexual transmission by oral genital contact. Several studies suggest that oral HPV infection is sexually acquired. [15] However, transmission of the virus by direct oral contact or by other means could not be excluded. In one study involving married couples with healthy oral mucosa, the results suggested that the oral route is an important means of HPV transmission between partners; one spouse had a 10-fold risk of acquiring oral HPV infection if the other spouse had persistent oral HPV infection. [16]

Site of HPV Related Tumor

There is evidence that specific epithelial areas of oropharynx such as the squamous columnar junction at the level of the tonsillar crypts, which is similar to the transformation zone of the uterine cervix from which most cervical cancers originate, have a stronger association with HPV due to the easy exposure of the basal cells. [17] It is unclear, however, whether HPV-associated cancer of the tonsils originates from crypt or surface epithelium, although data suggest that HPV inhabits the normal crypt epithelia. [18] The possibility that tonsillar tissue in Waldayer′s ring, which is important as an antigen-presenting site, is the reservoir of HPV, cannot be excluded.

Life time risk of head and neck squamous cell carcinoma with human papilloma virus infection

It is important to note that most people with HPV infection do not develop cancer. Research in women with cervical HPV infection clearly shows that 80% of women are able to clear the infection spontaneously as a result of normal immune system mechanisms. Similarly, whether persistent HPV infection in the head and neck region is a risk factor for oropharyngeal cancer has not been rigorously demonstrated. One study has shown a 14-fold increase in the risk of oropharyngeal cancer among patients seropositive for HPV-16 L1 protein, which is a measure of lifetime exposure to HPV-16 and suggested that exposure to HPV can precede the appearance of oropharyngeal cancer by 10 or more years. [19]

Diagnosis of Human Papilloma Virus Infection

The productivity of HPV DNA from oral samples when performing the molecular detection is quite low. It is therefore essential that all the procedures employed are highly sensitive, specific and reliable. The rate of HPV detection in HNSCC varies according to the method used and the tissue examined. Both oral tissue samples and exfoliated cells can be used for the HPV detection. Fresh frozen tissue (−70 o C) offers the highest yield of DNA as opposed to paraffin embedded or formalin fixed tissue, as these latter methods result in degradation of DNA, especially after prolonged storage. [5] Oral mucosal exfoliated cells can be obtained by either superficial brushing/scraping or oral rinse. The latter method is more efficient in terms of cell yield, DNA containing nucleated cells and is less invasive than that of biopsy. [20] Commercial mouthwashes seem to be more efficient than glucose or saline in terms of DNA yield, quality and productivity. [21] Polymerase chain reaction (PCR) is the most sensitive method available for HPV detection. Quantitative PCR offers the additional advantage of differentiating low level infection from contamination, as well as determining the viral load in the specimen. [22] Other methods such as southern blotting and In-situ hybridization should be avoided as they lead to lower HPV rates. However, there are certain inherent limitations in diagnosis of HPV associated oropharyngeal SCC. Firstly, the oral cavity is an extremely rarely a site of HPV related carcinomas, and secondly, detection of HPV DNA does not in itself prove a causal relationship. The mere presence of HPV DNA is not sufficient to relate it with carcinoma; rather, an evidence of biologically active HPV justifies the causation of SCC. 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. Since the levels of p16 are consistently associated with HPV infection, determination of p16 expression status by immunohistochemistry could serve as a reasonable surrogate marker for biologically relevant high-risk HPV infection. More recently, p16-positive/HPV-negative oropharyngeal SCC has also been isolated and therefore its reliability as a surrogate marker has been challenged. [23] There is still a need to enhance the standardization of the approach in terms of types of oral specimen examined, sampling method applied and HPV molecular assay employed.

Classification of Oropharyngeal Squamous Cell Carcinomas

HPV-DNA detection per se in an oropharyngeal SCC does not prove causal association. Only transcriptionally, active HPV DNA is biologically and clinically relevant as a risk factor of oropharyngeal SCC. According to one study it was hypothesized that for HPV-DNA-positive cases, p16 expression status would identify those that were biologically relevant. [13] This study delineated three biologically and clinically distinct types of oropharyngeal SCC: Class I, HPV-negative/p16 non expressing; class II, HPV-positive/p16 non expressing; and class III, HPV-positive/p16 expressing oro pharyngeal tumors. Only patients in class III had significantly lower p53 and pRb expression. Thus, only the HPV-positive/p16 expressing oropharyngeal SCC tumors (class III) are said to be the actual HPV associated SCC.

Clinic pathologic features of human papilloma virus related oropharyngeal carcinoma [24]

HPV + tumors are distinct clinically and pathologically. They are more common in young patients (<40 years) with a male to female ratio of 4:1. They usually present as a small or occult primary tumor with advanced neck disease. Microscopically, they are non-keratinizing squamous cell carcinomas with basaloid features, excessive mitosis and comedo type necrosis. The tumors have a distinct immunohistochemical profile characterized by strong and diffuse p16 reactivity, low or negative p53 staining and high Ki67 labeling scores. HPV + carcinomas are more radio-sensitive and have a better prognosis than the classical keratinizing SCC.

Clinical Implication of Human Papilloma Virus

Researchers have tried to delineate whether there is a difference in the clinical profile of patients with HPV positive tumors. HPV-associated oral SCC patient′s are often nonsmokers and nondrinkers and on average 5 years younger than their tobacco-use-associated counterparts. [17] There is controversy regarding the issue of whether HPV positivity is more common in patients with a decreased history of tobacco and alcohol consumption. Regardless, HPV has been shown to have an additive or even synergistic effect with tobacco and alcohol in increasing the risk of HNSCC. [14] Various studies have indicated that patient who have had more sexual partners and who engaged in oral sex practices are more likely to have HPV positive tumors. [4],[14] The association of HPV positive tumors with patient′s age, tumor grade, tumor stage or nodal involvement is not well defined. [3],[11] It appears that patients with HPV positive tumors have a better clinical prognosis. [3],[4],[5],[11] In one study, it was seen that the 5-year survival in HPV positive tumors was 79%, significantly higher than HPV negative tumors (20%, P=0.0095). Disease free survival was 75% compared with 15% (P=0.0025). The 5-year local recurrence was 14% compared with 45% (P=0.03). [13] The favorable outcome of HPV-induced oropharyngeal cancers might be attributable to the wild type Rb and p53 genes, absence of field cancerization or an increased radiosensitivity. [25],[26] HPV-positive carcinomas usually do not contain any p53 and Rb mutations, and overexpression of p16 protein has been reported in HPV-associated cancers.

The term ′field cancerization′ is used to describe the presence of carcinogen induced early genetic changes in the epithelium which lead to the development of multiple independent lesions. [27] Recently, there is also evidence that HPV positive cell line is extremely cisplatin resistance. [28] p16 and p53 protein expression, and high-risk human papillomavirus (HPV-HR) types have been associated with better survival in head and neck cancer (HNC). Evidence suggests that multiple molecular pathways need to be targeted to improve the poor prognosis of HNC. More recently a study examined the individual and joint effects of tumor markers for differences in predicting HNC survival. It revealed better predictive value of multiple molecular markers in HNC patients. It was seen that p16+/p53-/HPV-HR had best prognosis and the p16−/p53+/HPV- group had the lowest overall survival (84% vs. 60%, P<0.01) and disease-specific survival (86% vs. 66%, P<0.01). [29]

Role of Human Papilloma Virus Prevention

In recent years, two HPV vaccines have been developed. One of the vaccines is quadrivalent, and protects against HPV types 6, 11, 16 and 18. It is advised for reduction in genital warts along with cervical cancer. The other vaccine protects against HPV types 16 and 18 and is indicated for reduction in precancerous cervical lesions and cancer incidence. The impressive range of protection of these vaccines ranging from 86-100% has been recently reported. [30],[31] It is predicted that 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. It is thought that these vaccines might have a broader implication, and also reduce the incidence of oral and genital HPV infection and thereby HPV-related oropharyngeal, anal, vulvar, vaginal and penile cancers. Because HPV seems to be a distinct etiological factor for a certain percentage of head and neck cancer, the possibility that a subset of HNSCC might be prevented by HPV vaccination is worth considering.

Potential of Gene Therapy

The preventive HPV vaccines may not be useful for the treatment of existing diseases. Therefore, it is necessary to develop effective therapies to take care of already infected patients. One approach is to develop therapeutic HPV vaccination strategies eliciting cytolytic T cell response against high risk HPV-E6 and E7 oncoproteins. This approach, however, has not yet been successful against carcinoma in situ and cervical carcinoma in humans. [32] Another approach is to develop gene therapy directed against HPV-E6 and E7 oncogenes to tackle HPV positive cervical tumors. [33] The various methods include the use of E6 short interfering RNA, antisense RNA to E6 and E7 genes and mutated E2 protein that acts as cancer cell specific inducer of apoptosis. [34] It was found that when E6 and E7 protein expression is suppressed, p53 and Rb protein expression increase, the HPV infected cells undergo apoptosis and tumorigenicity is greatly suppressed. However, further understanding of HPV induced oncogenesis is required, for the appropriate use of gene therapy to treat HPV associated disease in humans.

Conclusion

Recent data confirms that infection with HPV-16 and to a lesser extent other high risk strains of HPV is an independent risk factor for a subset of oropharyngeal SCCs, especially once arising from the tonsil. Research results also trend in the direction that there is association between riskier sexual behavior, oral HPV infection and HNSCC. HPV-associated oropharyngeal SCCs are likely to have a better prognosis than HPV negative tumors. 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. Since HPV-16-DNA is present in relatively high fraction of HPV-associated HNSCC, effective vaccines targeted against this virus will soon hopefully be a part of the adjuvant therapy used to treat these tumors. However, future research is required to find out the associations of HPV infection with tobacco and alcohol exposure, appropriate screening methods for HPV infection and finally the impact of anti HPV vaccines on HPV-related oropharyngeal carcinoma.

References

1.Ferlay J, Bray F, Pisani P, Parkin DM. Globocan 2002: Cancer incidence and Prevalence worldwide. IARC Cancer Base no. 5, version 2.0. Lyon, France: IARC; 2004.  Back to cited text no. 1    
2.Loning T, Ikenberg H, Becker J, Gissmann L, Hoepfer I, Zur Hausen H. Analysis of oral papillomas, leukoplakias, and invasive carcinomas for human papillomavirus type related DNA. J Invest Dermatol 1985;84:417-20.  Back to cited text no. 2    
3.Ringstrom E, Peters E, Hasegawa M, Posner M, Liu M, Kelsey KT. Human papillomavirus type 16 and squamous cell carcinoma of the head and neck. Clin Cancer Res 2002;8:3187-92.  Back to cited text no. 3    
4.Gillison ML. Human papillomavirus-associated head and neck cancer is a distinct epidemiologic, clinical and molecular entity. Semin Oncol 2004;31:744-54.  Back to cited text no. 4  [PUBMED]  [FULLTEXT]
5.Dahistrand H, Dahlgren L, Lindquist D, Munck-Wikland E, Dalianis T. Presence of Human papillomavirus in tonsillar cancer is a favorable prognostic factor for clinical outcome. Anticancer Res 2004;24:1829-35.  Back to cited text no. 5    
6.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 1995;64:1-378.  Back to cited text no. 6  [PUBMED]  
7. Scheffner M, Werness BA, Huibregtse JM, Levine AJ, Howley PM. The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 1990;63:1129-36.  Back to cited text no. 7    
8.Boyer SN, Wazer DE, Band V. E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res 1996;56:4620-4.  Back to cited text no. 8  [PUBMED]  [FULLTEXT]
9.Sidransky D. Molecular genetics of head and neck cancer. Curr Opin Oncol 1995;7:229-33.  Back to cited text no. 9  [PUBMED]  
10.Wittenkindt C, Gultekin E, Weissenborn SJ, Dienes HP, Pfister HJ, Klussmann JP. Expression of p16 protein is associated with human papillomavirus status in tonsillar carcinomas and has implications on survival. Adv Otorhinolaryngol 2005;62:72-80.  Back to cited text no. 10    
11. Dahlgren L, Dahistrand H, Lindquist D, Högmo, A, Björnestål L , Lindholm J, et al. Human papillomavirus is more common in base of tongue than in mobile tongue cancer and is favourable prognostic factor in base of tongue cancer patients. Int J Cancer 2004;112:1015-9.  Back to cited text no. 11    
12.Kreimer AR, Clifford GM, Boyle P, Franceschi S. Human papillomavirus types in head and neck squamous cell carcinomas worldwide: A systematic review. Cancer Epidemiol Biomarkers Prev 2005;14:467-75.  Back to cited text no. 12  [PUBMED]  [FULLTEXT]
13.Weinberger PM, Yu Z, Haffty BG, Kowalski D, Harigopal M, Brandsma J, et al. Molecular classification identifies a subset of human papillomavirus-associated oropharyngeal cancers with favourable prognosis. J Clin Oncol 2006;24:736-47.  Back to cited text no. 13  [PUBMED]  [FULLTEXT]
14.Herrero R, Castellsague X, Pawlita M, Lissowska J, Kee F, Balaram P, et al. Human papillomavirus and oral cancer: The International Agency for Research on Cancer multicentre study. J Natl Cancer Inst 2003;95:1772-83.  Back to cited text no. 14    
15.D'Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, et al. Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 2007;356:1944-56.  Back to cited text no. 15  [PUBMED]  [FULLTEXT]
16.Rintala M, Grénman S, Puranen M, Syrjänen S. Natural history of oral human papillomasvirus infections in female and male partners: A prospective Finnish HPV Family Study. J Clin Virol 2006;35:89-94.  Back to cited text no. 16    
17. McNeil C. Human papillomavirus in oropharyngeal cancers: New data inspire hope for vaccines. J Natl Cancer Inst 2000;92:680-1.  Back to cited text no. 17  [PUBMED]  [FULLTEXT]
18. Hammarstedt L, Lindquist D, Dahlstrand H, Romanitan M, Dahlgren LO, Joneberg J, et al. Human papillomavirus as a risk factor for the increase in incidence of tonsillar cancer. Int J Cancer 2006;119:2620-3.  Back to cited text no. 18  [PUBMED]  [FULLTEXT]
19. Mork J, Lie AK, Glattre E, Hallmans G, Jellum E, Koskela P, et al. Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med 2001;344:1125-31.  Back to cited text no. 19  [PUBMED]  [FULLTEXT]
20.Lawton G, Thomas S, Schonrock J, Monsour F, Frazer I. Human papillomaviruses in normal oral mucosa: A comparison of methods for sample collection. J Oral Pathol Med 1992;21:265-9.  Back to cited text no. 20  [PUBMED]  
21.Heath EM, Morken NW, Campbell KA, Tkach D, Boyd EA, Strom DA. Use of buccal cells collected in mouthwash as a source of DNA for clinical testing. Arch Pathol Lab Med 2001;125:127-33.  Back to cited text no. 21  [PUBMED]  [FULLTEXT]
22.Ha PK, Califano JA. The role of human papillomavirus in oral carcinogenesis. Crit Rev Oral Biol Med 2004;15:188-96.  Back to cited text no. 22  [PUBMED]  [FULLTEXT]
23.Perrone F, Gloghini A, Cortelazzi B, Bossi P, Licitra L, Pilotti S. Isolating p16-positive/HPV-negative Oropharyngeal Cancer: An Effort Worth Making. Am J Surg Pathol 2011;35:774-7.  Back to cited text no. 23  [PUBMED]  [FULLTEXT]
24.El-Mofty SK. Human papillomavirus (HPV) related carcinomas of the upper aerodigestive tract. Head Neck Pathol 2007;1:181-5.  Back to cited text no. 24  [PUBMED]  [FULLTEXT]
25.Termine N, Giovannelli L, Rinaldi G, Campisi G. Human papillomavirus infection and head and neck squamous cell carcinoma: Current debates. J Stomatol Invest 2008;2:27-33.  Back to cited text no. 25    
26.Lindel K, Beer KT, Laissue J, Greiner RH, Aebersold DM. Human papillomavirus positive squamous cell carcinoma of the oropharynx: A radiosensitive subgroup of head and neck carcinoma. Cancer 2001;92:805-13.  Back to cited text no. 26  [PUBMED]  
27.Slaughter DP, Southwick H, Smejkol W. Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer 1953;6:963-8.  Back to cited text no. 27    
28. Hoffmann TK, Sonkoly E, Hauser U, Van Lierop A, Whiteside TL, Klussmann JP, et al. Alterations in the p53 pathway and their association with radio- and chemosensitivity in head and neck squamous cell carcinoma. Oral Oncol 2008;44:1100-9.  Back to cited text no. 28  [PUBMED]  [FULLTEXT]
29.Smith EM, Rubenstein LM, Hoffman H, Haugen TH, Turek LP. Human papillomavirus, p16 and p53 expression associated with survival of head and neck cancer. Infect Agent Cancer 2010;5:4  Back to cited text no. 29    
30.Garland SM, Hernandez AM, Wheeler CM, Perez G, Harper DM, Leodolter S, et al. Females united to unilaterally reduce endo/ecto cervical disease (FUTURE - I) investigators: Qudrivalent vaccine against Human papillomaviruses to prevent anogenital diseases. N Engl J Med 2007;356:1928-43.  Back to cited text no. 30    
31.Harper DM, Franco EL, Wheeler CM, Moscicki AB, Romanowski B, Roteli- Martins CM, et al. Sustained efficacy upto 4.5 years of a bivalent L1 virus like particle vaccine against Human papillomavirus types 16 and18: Follow up from a randomized control trial. Lancet 2006;367:1247-55.  Back to cited text no. 31    
32.Borysiewicz LK, Fiander A, Nimako M, Man S, Wilkinson GW, Westmoreland D, et al. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet 1996;347:1523-7.  Back to cited text no. 32  [PUBMED]  
33.Yamoto K, Yamada T, Kizaki M, Ui Tei K, Natori Y, Fujino M, et al. New highly potent and specific E6 andE7 siRNAs for treatment of HPV 16 positive cervical cancer. Cancer Gene Ther 2008;15:140-53.  Back to cited text no. 33    
34.Green KL, Brown C, Roeder GE, Southgate TD, Gaston K. A cancer cell specific inducer of apoptosis. Hum Gene Ther 2007;18:547-61.  Back to cited text no. 34  [PUBMED]  [FULLTEXT]

Copyright 2011 - Journal of Cancer Research and Therapeutics

Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil