Abstract
Objective
Human papilloma virus (HPV) positive and HPV negative head and neck squamous cell cancer (HNSCC) are biologically distinct with a prognostic advantage for HPV positive patients compared to HPV negative cases. DNA promoter methylation is central to human diseases such as cancer, including HNSCC, with reported genome-wide hypomethylaton and promoter hypermethylation in HPV positive HNSCC tumors. The goal of this study was to identify differentially methylated genes in HPV positive versus HPV negative primary HNSCC genomes with clues to signaling networks.
Study Design
Laboratory-based study.
Setting
Primary care academic health care system.
Subjects and Methods
DNA from 4 HPV positive and 4 HPV negative freshly frozen primary HNSCC were subject to comprehensive genome-wide methylation profiling. Differentially methylated gene lists were examined using the Signal Transduction Pathways (canonical) filter in the Genomatix Pathway System (GePS).
Results
Twofold methylation differences were observed between HPV positive and HPV negative cases for 1168 genes. Pathway analysis applied to investigate the biological role of the 1168 differentially methylated genes revealed 8 signal transduction pathways forming a network of 66 genes, of which 62% are hypermethylated.
Conclusion
Our study reveals a predominant hypermethylation profile for genes in signal transduction pathways of HPV positive HNSCC tumor genomes. Because signaling events in the cell play a critical role in the execution of key biological functions, insights into how complex cellular signaling cascades and networks may be programmed in HNSCC are likely to be critical in the development of new biological agents designed to hit multiple targets.
Keywords: hypermethylation, gene networks, pathways, signal transduction
Background
The overwhelming majority of mucosal head and neck cancers are squamous cell carcinomas (HNSCC)1 that primarily develop in the oral cavity, pharynx, and larynx. Accurate and reliable stratification of HNSCC for prediction of outcomes has been challenging, mainly because of the numerous anatomic sites and subsites from which tumors can arise.
In the absence of a single risk factor attributable to developing HNSCC, the 2 well-studied important risk factors, tobacco and alcohol,2 are responsible for 72% of HNSCC cases.3 More recent epidemiological and laboratory evidence indicate the human papilloma virus (HPV) as a causative agent for some HNSCC4 and an independent risk factor for oropharyngeal cancer (OPSCC).5 A systematic review of 5046 patients with HNSCC reported an overall prevalence of HPV infection of 25.9% and concurs with a more recent meta-analysis of 5681 HNSCC.6 The prevalence of HPV infection was significantly higher among patients with OPSCC (35.6%) than among those with oral (23.5%) or laryngeal (24%) SCC.7 Approximately 95% of these HNSCC HPV subgroups contain high-risk HPV type 16 (HPV-16) genomic DNA sequences.8 Its contribution to neoplastic progression is predominantly through the action of the viral oncoproteins E6 and E7.9 Expression of these proteins is sufficient for the immortalization of primary human epithelial cells and induction of histologic atypia characteristic of pre-invasive HPV-associated squamous intraepithelial lesions.10 The biologic significance of HPV as another independent risk factor is underscored by the improved prognosis for patients with HPV positive HNSCC relative to HPV negative HNSCC,4,5 due in part to a better therapeutic response to chemoradiotherapy.11
Global characterization of the HNSCC methylome is beginning to uncover differential landscapes in HPV positive versus HPV negative tumors. HPV positive cells had higher CpG methylation both in nonrepetitive regions (genic and nongenic) and in repetitive regions.12,13 Querying differentially methylated genes into the Pathway analysis framework to identify distinct signaling pathway networks has the potential to provide a molecular basis for further exploration of these genes as differential targets in HPV positive and HPV negative HNSCC. In this type of analysis, a biological system is surveyed in the context of disease (or other interesting phenotypes) to identify gene groups associated with biological systems (bionetwork).14,15 Bionetwork coupling affords a strategic knowledge base approach and has been used to better understand the systems biology of disease processes and identify potential therapeutic targets.16–18 Signal transduction is the means by which cells respond to extracellular information. The major signaling systems have been conserved to a remarkable extent in all animals.
While it is becoming more firmly established that HPV positive HNSCC have better survival outcomes than HPV negative HNSCC, believed to be because of better response to chemo radiation,11 the underlying mechanism for these improved prognosis outcomes remains underexplored. In this exploratory study, we hypothesized that differentially methylated genes in HPV positive versus HPV negative tumors can point to biological processes encased within signaling networks functioning in HNSCC.
Materials and Methods
DNA from 4 HPV positive and 4 HPV negative freshly frozen primary HNSCC were subject to comprehensive genome-wide methylation profiling using the HumanMethylation27 BeadArray (San Diego, California). Tumor site and demographic characteristics are presented in Table 1. This study was approved by the Henry Ford Health System Institutional Review Board committee.
Table 1.
Cohort characteristics
Sample ID | Site | Age | Race | Gender | HPV16E6 | Smoking |
---|---|---|---|---|---|---|
31 | Larynx (supraglottis) | 51 | CA | F | Negative | Yes, past |
41 | Tongue | 41 | CA | M | Negative | Yes, past |
42 | Oral cavity | 54 | CA | M | Negative | Yes, current |
43 | Tongue | 71 | CA | M | Positive | Yes, current |
45 | Tongue | 49 | CA | M | Positive | Yes, past |
46 | Tongue | 46 | CA | M | Negative | Yes, current |
47 | Tongue | 54 | CA | M | Positive | No |
48 | Oral cavity | 52 | AA | M | Positive | Yes, current |
Abbreviations: CA, Caucasian American; AA, African American; F, female; M, male.
DNA was extracted according to the manufacturer’s protocol (Qiagen Inc, Chatsworth, California). Tumor HPV DNA concentrations were measured using real-time quantitative PCR (qPCR) as previously described.19,20 Briefly, primers and probes to a housekeeping gene (β-globin) are run in parallel to standardize the input DNA. By using serial dilutions, standard curves are developed for the HPV viral copy number using CaSki (American Type Culture Collection, Manassas, Virginia) cell line genomic DNA, known to have 600 copies/genome equivalent (6.6 pg of DNA/genome). The cut-off value for HPV16 positive status was ≥0.03 (≥3 HPV genome copy/100 cells).19–21
The Infinium 27k assays were performed at the Applied Genomics Technology Center (AGTC), which is part of Wayne State University School of Medicine’s Department of Obstetrics and Gynecology (Detroit, Michigan). The 27k platform measures methylation status of over 27,000 CpGs located in more than 14,000 gene promoters. The methylation score for each CpG is represented as a beta (β) value according to the fluorescent intensity ratio. Every β value is accompanied by a detection P-value. β values may take any value between 0 (non-methylated) and 1 (completely methylated) and is determined using the Genome Studio (Illumina, San Diego, California). Probes are discarded if this detection P-value is more than .05. The only corrections that are made to the data are background subtraction and normalization. The resulting beta values were exported into Microsoft Excel and JMP (SAS Institute, Cary, North Carolina) for data analysis.
The Genomatix Pathway System22 (GePS) uses information extracted from public and proprietary databases to display canonical pathways or to create and extend networks based on literature data. Genomatix Pathway System software was applied to build a network from differentially methylated genes. Biological processes enriched in the differentially methylated gene lists using the Signal Transduction Pathways (canonical) filter in GePS were identified and ranked starting with the lowest P-values determined using Genomatix analysis software.
Statistical Methods
To avoid gender-specific methylation bias, sex chromosome loci (n = 1092) were excluded, resulting in a final data set that consisted of 26,486 autosomal loci associated with 13,890 genes. Criteria for differential methylation was set as CpG sites with a ratio of the mean β for the HPV positive samples to the mean β for the HPV negative samples of at least 2.0 or less than 0.5 (ie, a twofold difference).
Results
Of the 26,486 autosomal CpG loci, the 1355 (5%) differentially methylated CpGs between HPV positive and HPV negative samples were assigned to 1168 genes (Figure 1, Supplemental Table 1, available at www.otojournal.org). Of the 1168 genes, 686 (59%) were hypermethylated, 467 (40%) were hypomethylated, and 15 (1%) had both hypo-and hypermethylation (ie, these 15 genes each had more than 1 CpG with a twofold change, and at least 1 of those was hypermethylation and 1 was hypomethylation).
Figure 1.
Of the 26,486 autosomal CpG loci, the 1355 (5%) differentially methylated CpGs between HPV positive and HPV negative samples were assigned to1168 genes: 686 (59%) were hypermethylated, 467 (40%) were hypomethylated.
Pathway analysis applied to investigate the biological role of the 1168 differentially methylated genes using the Signal Transduction Pathways (canonical) filter in GePS formed a network of 66 genes (Supplemental Figure 1), of which 62% are hypermethylated (41 of the 66) and 25 (39%) hypomethylated (Supplemental Table 2). Of the 66 genes, 62 were accounted for in 8 signal transduction canonical pathways (P <.05). These included the Wnt/Beta (β)-catenin degradation (P = .003) (Figure 2A), basic mechanism of action of peroxisome-proliferator-activated receptor (PPAR) elements (P = .006), E-cadherin (P = .018) (Figure 2B), RXR (retinoid X receptor) and RAR (retinoic acid receptor) heterodimerization (P = .036), ion channels and their functional role in vascular endothelium (P = .036), ErbB receptor (P = .04) (Figure 2C), signaling events mediated by stem cell factor receptor (c-Kit) (P = .04), and Integrin signaling (Filopodium formation) (P = .041) signaling events (Supplemental Table 3).
Figure 2.
(A, B, C): Yellow boxes: nodes in the network representing genes; edges: links between nodes indicating a relationship with corresponding genes. A: Beta-catenin degradation signaling; B: E-cadherin signaling events; C: ErbB receptor signaling network. Asterisked genes are hypermethylated.
Of the 66 genes, 8 were represented in more than 1 pathway (Supplemental Table 4) and included CAV1, CD44, EGFR, ERB2, GPC1, PPARA, PPARG, and SDC2.
Discussion
Molecular subtyping has shown that HPV positive HNSCC differ from HPV negative HNSCC in several ways. HPV positive HNSCC have genetic alterations that are indicative of HPV oncoprotein function8 and are characterized by wild-type TP53,23,24 wild-type CDKN2A (p16),25 and infrequent amplification of cyclin D,26–28 whereas the converse is true for HPV negative HNSCC. HPV positive HNSCC also differ from HPV negative HNSCC in their patterns of allelic29 and chromosomal30,31 loss and in their global gene expression profiles.26,32 Whole-exome (protein coding genes) mutational profiling33,34 recently confirmed mutations in TP53 as a potential genomic stratifier for HPV status.
To test the possible involvement of epigenetic modulation by HPV in HNSCC, we conducted a genome-wide DNA methylation analysis. In this hypothesis-generating study, albeit one with a small sample size, HPV status appears to modulate or influence promoter methylation as evidenced from this global examination of over 27,000 CpGs. The results point to HPV-associated genomic differences involving epigenetic events of differential DNA methylation that warrant consideration in addition to copy number changes and genomic mutation differences. The differentially methylated genes between HPV positive and HPV negative HNSCC in this study indicate more hypermethylated than hypomethylated profiles (59% vs 40%) and support higher gene promoter hypermethylation levels in HPV positive tumor cells.
From a clinical significance standpoint, recent studies are beginning to establish a mechanistic role for promoter methylation with improved survival outcomes in HPV positive HNSCC. Gubanova et al showed that promoter hypermethylation and concordant low SMG-1 expression correlated not only with HPV positive status and improved patient survival, but also enhanced response to radio therapy in HPV-positive HNSCC cell lines.35
Our study not only highlights and confirms previously reported studies of HPV-associated differentially methylated profiles, but also attempts to relate them to biological contexts of signal transduction pathways. The biological processes enriched within the differentially methylated genes point to likely functional consequences and biological roles as highlighted by the emergence of 8 signal transduction canonical pathways.
Pathway analysis has become the first choice for extracting and explaining the underlying biology for high throughput molecular measurements, as it reduces complexity and has increased explanatory power. Wnt/β-catenin signaling is a branch of a functional network that developed around a class of proteins called armadillo proteins and dates back to the first anaerobic metazoans.36 In vertebrates, Wnt signaling acts to prevent β-catenin degradation and promote its ability to activate transcription. In the canonical Wnt pathway, β-catenin acts as the central component.37,38 Wnt/β-catenin signaling is involved in a broad range of biological systems, including stem cells, embryonic development, and adult organs. Deregulation of components in this pathway has been implicated in a wide spectrum of diseases including a number of cancers and degenerative diseases.
Activation of the canonical Wnt pathway at multiple levels39 (plasma membrane, cytoplasm, or nucleus) supports transformation of HPV-infected primary human keratinocytes.40 Cytoplasmic and nuclear expression of β-catenin, a hallmark of the activated Wnt pathway, was reported in archived human cervical carcinoma samples.40–42 A follow-up study provides a potential link between activation of the Wnt signaling pathway and its contribution to HPV-mediated cervical cancer.43β-catenin is accumulated in the nucleus of HPV16-positive oropharyngeal cancer cells as a likely direct consequence of E6 and E7 HPV oncogene expression.44 In the absence of β-catenin mutations, epigenetic changes in Wnt pathway regulators are thought to explain the activation of the canonical Wnt pathway.45 Methylation of WNT-antagonists SFRP-1, SFRP-2, SFRP-4, SFRP-5, WIF-1, and DKK-3 in oral cancer implicates the WNT pathway in oral cancer pathogenesis.46 The latter supports the finding of our study, indicating hypermethylation of 11 of 17 Wnt/β-catenin pathways genes to include SFRP-4 and DKK-3 (Supplemental Table 3).
E-cadherin is a member of the cadherin family of transmembrane glycoproteins and mediates formation of adherens junctions in epithelial cells.47 Studies using normal epithelial cell cultures suggest that E-cadherin loss of function may provide a growth advantage.48,49 E-cadherin can dictate the activity of several mitogenic signaling pathways, as it differentially regulates EGFR/ERK, PI3-K/AKT, and β-catenin/TCF.49 In HNSCC, reduction of E-cadherin results in upregulation of epidermal growth factor receptor (EGFR) and its downstream signaling pathways.50
The EGFR gene is located at 7p12 and makes a 170-kD transmembrane glycoprotein.51 It is a member of the receptor protein tyrosine kinase family with several extra-cellular growth factor ligands, including epidermal growth factor (EGF) and transforming growth factor (TGF)α. Overexpression of EGFR is observed in 42% to 80% of HNSCC studied,52,53 and EGFR gene amplification occurs in up to 30% of HNSCC tumors.54,55 The majority of evidence suggests that increased EGFR expression and gene copy number are linked to poorer patient outcomes in HNSCC56–59 and may be useful in identifying subgroups of patients at high risk of tumor recurrence and in guiding therapy.52,60,61
Downregulation of the E-cadherin pathway with upregulation of EFGR as a consequence50 indicates cross-talk between E-cadherin and EGFR pathways. In our study, genes accounted for in both pathways were hypermethylated (7/7 genes in E-cadherin, 4/4 genes in the EGFR pathway), suggesting downregulation or loss of function of these pathways in HPV positive HNSCC.
Integrin signaling critically contributes to the progression, growth, and therapy resistance of malignant tumors, including HNSCC.62 Integrins are transmembrane cell surface receptors comprised of 18 α and 8 β subunits in close noncovalent association that form structural and functional bridges between the extracellular membrane (ECM) and cytoskeletal linker proteins within a cell, with roles in cell survival, proliferation, invasion, and cancer therapy resistance.63,64 Targeting of β1 integrins with inhibitory antibodies enhances the sensitivity to ionizing radiation and delays the growth of HNSCC cell lines in 3D cell culture and in xenografted mice65 and suggests that robust and selective pharmacological targeting of β1 integrins may provide therapeutic benefit to overcome tumor cell resistance to radiotherapy.
Peroxisome-proliferator-activated receptors (PPARs) are nuclear hormone receptors that mediate the effects of fatty acids and their derivatives at the transcriptional level. PPARs regulate gene expression by binding with RXR (retinoid X receptor) as a heterodimeric partner to specific DNA sequence elements termed PPRE (peroxisome proliferator response element).66 This heterodimeric transcription factor complex then binds to cognate sequences in promoter regions of target genes involved in the catabolism of fatty acids.67 Three isotypes, PPARα, PPARβ/δ, and PPARγ, have been identified. PPARs have been implicated in many normal and disease-related biologic processes relevant to the heart and vasculature, including lipid and energy metabolism, inflammation, embryo implantation, diabetes, and cancer.68 Increased expression of PPARβ/δ is reported in HNSCC.69
Retinoids are natural and synthetic vitamin A derivatives that regulate development,70 cell proliferation,71 and differentiation.72 Retinoids control cell proliferation and differentiation by binding to the nuclear retinoic acid receptors (RARs) or heterodimeric retinoid X receptors (RXRs) that in turn affect target gene expression.73 Retinoids have chemopreventive effects in various tumor types, including the skin, prostate, ovary, leukemia, and breast,74 and are useful tools to uncover therapeutic and chemopreventive pathways that can reduce carcinogenesis as illustrated by the UBE1L gene as a candidate-pharmacologic target for lung cancer chemoprevention.75
It is widely accepted that the PPARs must heterodimerize with RXR to carry out most of their functions, making PPARs integral partners of the RXR-dependent signaling network. Cross-talk between these 2 pathways is an active area of research investigation.76
Membrane ion channels are essential for many physiological processes. Recent studies have shown that abnormal expression and/or activity of a number of ion channels (eg, voltage-gated K+, Na+, Ca2+channels, transient receptor potential [TRP] channels, and epithelial Na+/degenerin family of ion channels) are involved in the growth/proliferation, migration, and/or invasion of cancer cells.77 Ion channels represent promising targets for developing novel and effective cancer therapies.
HNSCC cells have the ability to exploit diverse signaling pathways for growth advantage, cell survival, and evasion of apoptosis, with advancement along the tumorigenesis progression continuum. Current signal transduction based therapies under investigation in HNSCC include growth factor pathways (eg, epidermal growth factor) and nuclear receptor pathways (eg, RAR, RXR signaling).78–80 Cetuximab remains the sole FDA-approved molecular targeted therapy available for HNSCC, and though there are several new biological agents targeting EGFR and other pathways in the regulatory approval pipeline, the complexity of aberrant signaling in HNSCC may explain why interfering with only single steps in these pathways have not shown marked clinical response in HNSCC patients.
When compared to the genome, which is identical in every cell and tissue in the human body, the epigenome is highly variable over the life course, from tissue to tissue and from environment to environment.81 Also, unlike genes that are inactivated by nucleotide sequence variation, genes silenced by epigenetic mechanisms are still intact and thus retain the potential to be reactivated by environmental or medical intervention.81 There are several current human therapeutic intervention trials to reverse deleterious epigenetic changes. Some examples include epigenetic therapeutic trials to treat T-cell lymphoma based on reactivation of tumor suppressor genes82 and similar trials to prevent colorectal cancer by inhibiting the enzyme responsible for DNA methylation.83 Such therapies have shown promise in halting tumor growth by reactivation of the tumor suppressor gene(s) or by blocking progression of precancerous epigenetic lesions. Increased genome-wide methylation has been found to be more pronounced in HPV positive HNSCC13 and suggests additional treatment options for HPV positive tumors with demethylating drugs. Additionally, demethylating drugs in combination with therapeutic HPV DNA vaccines have been found to control more effectively a variety of HPV-associated malignancies due to the fact that DNA methylation is capable of decreasing expression of the encoded antigen of the DNA vaccines.84 In fact, preliminary studies already suggest that there is promise of improving preventative HPV DNA vaccine therapy by the addition of the demethylating drug 5-aza-2′ deoxycytidine.84 The potential for specific hypermethylated sites in HPV negative HNSCC may allow these patients to also benefit from demethylation treatment strategies.
An interesting feature of this study is the observation of a preponderance of methylated genes in the top ranked signal transduction pathways of HPV positive HNSCC tumor genomes. However, given the small sample size, this observation necessitates confirmation and validation in both studies with larger sample sizes that also include potential confounding factors (smoking, alcohol) and mechanistically designed studies from the standpoint of concordance of methylation status with gene expression and more importantly, the impact of gene methylation on the functional outcomes of the pathways they reside in.
Conclusion
The progressive acquisition of a malignant phenotype in HNSCC is dependent on the aberrant activation of multiple signaling pathways underscoring finely choreographed genomic instability events, likely influenced by etiological factors and risk habits, to achieve biological distinctiveness. The integration of the differentially methylated genes into known biological pathways is a versatile tool to gain insights into the biological complexity of genome-wide promoter methylation. In this hypothesis-generating pilot study, we were able to highlight genes and their association to signal transduction pathways germane to HNSCC. Signaling events in the cell play a critical role in the execution of key biological functions. In-depth understanding of how the complex cellular signaling cascades and networks may be programmed in HNSCC will be instrumental in the development of new biological agents designed to hit multiple targets.
Supplementary Material
Footnotes
Reprints and permission: sagepub.com/journalsPermissions.nav
Author Contributions
Maria J. Worsham, substantial contributions to conception and design, acquisition of data, analysis, and interpretation of data; drafting the article and revising it critically for important intellectual content; and final approval of the version to be published; Kang Mei Chen, substantial contributions to conception and design, acquisition of data, analysis, and interpretation of data; drafting the article and revising it critically for important intellectual content; and final approval of the version to be published; Tamer Ghanem, substantial contributions to conception and design, acquisition of data, and interpretation of data; drafting the article and revising it critically for important intellectual content; and final approval of the version to be published; Josena K. Stephen, substantial contributions to conception and design, acquisition of data, and interpretation of data; drafting the article and revising it critically for important intellectual content; and final approval of the version to be published; George Divine, substantial contributions to conception and design, acquisition of data, and analysis and interpretation of data; drafting the article and revising it critically for important intellectual content; and final approval of the version to be published.
This article was presented at the 2012 AAO-HNSF Annual Meeting & OTO EXPO; September 9–12, 2012; Washington, DC.
Disclosures
Competing interests: None.
Sponsorships: NIH.
Funding source: R01 DE15990.
Additional supporting information may be found at http://oto.sagepub.com/content/by/supplemental-data
References
- 1.Horner MJ, Ries LAG, Krapcho M, et al., editors. [Accessed March 29, 2010.];SEER cancer statistics review, 1975–2006, section 16: melanoma of the skin. 2009 http://seer.cancer.gov/csr/1975_2006/
- 2.Hashibe M, Brennan P, Benhamou S, et al. Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst. 2007;99:777–789. doi: 10.1093/jnci/djk179. [DOI] [PubMed] [Google Scholar]
- 3.Hashibe M, Brennan P, Chuang SC, et al. Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev. 2009;18:541–550. doi: 10.1158/1055-9965.EPI-08-0347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gillison ML, Lowy DR. A causal role for human papillomavirus in head and neck cancer. Lancet. 2004;363:1488–1489. doi: 10.1016/S0140-6736(04)16194-1. [DOI] [PubMed] [Google Scholar]
- 5.Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363:24–35. doi: 10.1056/NEJMoa0912217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Dayyani F, Etzel CJ, Liu M, et al. Meta-analysis of the impact of human papillomavirus (HPV) on cancer risk and overall survival in head and neck squamous cell carcinomas (HNSCC) Head Neck Oncol. 2010;2:15. doi: 10.1186/1758-3284-2-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kreimer AR, Clifford GM, Boyle P, et al. Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomarkers Prev. 2005;14:467–475. doi: 10.1158/1055-9965.EPI-04-0551. [DOI] [PubMed] [Google Scholar]
- 8.Gillison ML. Human papillomavirus-associated head and neck cancer is a distinct epidemiologic, clinical, and molecular entity. Semin Oncol. 2004;31:744–754. doi: 10.1053/j.seminoncol.2004.09.011. [DOI] [PubMed] [Google Scholar]
- 9.Munger K, Baldwin A, Edwards KM, et al. Mechanisms of human papillomavirus-induced oncogenesis. J Virol. 2004;78:11451–11460. doi: 10.1128/JVI.78.21.11451-11460.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Nguyen M, Song S, Liem A, et al. A mutant of human papillomavirus type 16 E6 deficient in binding alpha-helix partners displays reduced oncogenic potential in vivo. J Virol. 2002;76:13039–13048. doi: 10.1128/JVI.76.24.13039-13048.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fakhry C, Westra WH, Li S, et al. 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. doi: 10.1093/jnci/djn011. [DOI] [PubMed] [Google Scholar]
- 12.Sartor MA, Dolinoy DC, Jones TR, et al. Genome-wide methylation and expression differences in HPV(+) and HPV(−) squamous cell carcinoma cell lines are consistent with divergent mechanisms of carcinogenesis. Epigenetics. 2011;6:777–787. doi: 10.4161/epi.6.6.16216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Richards KL, Zhang B, Baggerly KA, et al. 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. doi: 10.1371/journal.pone.0004941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lum PY, Chen Y, Zhu J, et al. Elucidating the murine brain transcriptional network in a segregating mouse population to identify core functional modules for obesity and diabetes. J Neurochem. 2006;97(suppl 1):50–62. doi: 10.1111/j.1471-4159.2006.03661.x. [DOI] [PubMed] [Google Scholar]
- 15.Nilsson EE, Savenkova MI, Schindler R, et al. Gene bionet-work analysis of ovarian primordial follicle development. PLoS One. 2010;5:e11637. doi: 10.1371/journal.pone.0011637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen Y, Zhu J, Lum PY, et al. Variations in DNA elucidate molecular networks that cause disease. Nature. 2008;452:429–435. doi: 10.1038/nature06757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gargalovic PS, Imura M, Zhang B, et al. Identification of inflammatory gene modules based on variations of human endothelial cell responses to oxidized lipids. Proc Natl Acad Sci U S A. 2006;103:12741–12746. doi: 10.1073/pnas.0605457103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Horvath S, Zhang B, Carlson M, et al. Analysis of oncogenic signaling networks in glioblastoma identifies ASPM as a molecular target. Proc Natl Acad Sci U S A. 2006;103:17402–174067. doi: 10.1073/pnas.0608396103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stephen JK, Chen KM, Shah V, et al. Human papillomavirus outcomes in an access-to-care laryngeal cancer cohort. Otolaryngol Head Neck Surg. 2012;146:730–738. doi: 10.1177/0194599811434482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Stephen JSDG, Chen KM, Chitale D, Havard S, Worsham MJ. Significance of p16 in site-specific HPV positive and HPV negative head and neck squamous cell carcinoma. Cancer Clin Oncol. 2013;2:51–61. doi: 10.5539/cco.v2n1p51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Worsham MJ, Stephen JK, Chen KM, et al. Improved survival with HPV among African Americans with oropharyngeal cancer. Clin Cancer Res. doi: 10.1158/1078-0432.CCR-12-3003. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Frisch M, Klocke B, Haltmeier M, et al. LitInspector: literature and signal transduction pathway mining in PubMed abstracts. Nucleic Acids Res. 2009;37:W135–W140. doi: 10.1093/nar/gkp303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gillison ML, Koch WM, Capone RB, 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. doi: 10.1093/jnci/92.9.709. [DOI] [PubMed] [Google Scholar]
- 24.Hafkamp HC, Speel EJ, Haesevoets A, et al. A subset of head and neck squamous cell carcinomas exhibits integration of HPV 16/18 DNA and overexpression of p16INK4A and p53 in the absence of mutations in p53 exons 5–8. Int J Cancer. 2003;107:394–400. doi: 10.1002/ijc.11389. [DOI] [PubMed] [Google Scholar]
- 25.Licitra L, Perrone F, Bossi P, et al. High-risk human papillomavirus affects prognosis in patients with surgically treated oropharyngeal squamous cell carcinoma. J Clin Oncol. 2006;24:5630–5636. doi: 10.1200/JCO.2005.04.6136. [DOI] [PubMed] [Google Scholar]
- 26.Slebos RJ, Yi Y, Ely K, et al. Gene expression differences associated with human papillomavirus status in head and neck squamous cell carcinoma. Clin Cancer Res. 2006;12:701–709. doi: 10.1158/1078-0432.CCR-05-2017. [DOI] [PubMed] [Google Scholar]
- 27.Smith EM, Wang D, Kim Y, et al. P16INK4a expression, human papillomavirus, and survival in head and neck cancer. Oral Oncol. 2008;44:133–142. doi: 10.1016/j.oraloncology.2007.01.010. [DOI] [PubMed] [Google Scholar]
- 28.Ragin CC, Taioli E, Weissfeld JL, et al. 11q13 amplification status and human papillomavirus in relation to p16 expression defines two distinct etiologies of head and neck tumours. Br J Cancer. 2006;95:1432–1438. doi: 10.1038/sj.bjc.6603394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Braakhuis BJ, Snijders PJ, Keune WJ, et al. Genetic patterns in head and neck cancers that contain or lack transcriptionally active human papillomavirus. J Natl Cancer Inst. 2004;96:998–1006. doi: 10.1093/jnci/djh183. [DOI] [PubMed] [Google Scholar]
- 30.Smeets SJ, Braakhuis BJ, Abbas S, et al. 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. doi: 10.1038/sj.onc.1209275. [DOI] [PubMed] [Google Scholar]
- 31.Dahlgren L, Mellin H, Wangsa D, et al. Comparative genomic hybridization analysis of tonsillar cancer reveals a different pattern of genomic imbalances in human papillomavirus-positive and -negative tumors. Int J Cancer. 2003;107:244–249. doi: 10.1002/ijc.11371. [DOI] [PubMed] [Google Scholar]
- 32.Martinez I, Wang J, Hobson KF, et al. Identification of differentially expressed genes in HPV-positive and HPV-negative oropharyngeal squamous cell carcinomas. Eur J Cancer. 2007;43:415–432. doi: 10.1016/j.ejca.2006.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Stransky N, Egloff AM, Tward AD, et al. The mutational landscape of head and neck squamous cell carcinoma. Science. 2011;333:1157–1160. doi: 10.1126/science.1208130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Agrawal N, Frederick MJ, Pickering CR, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333:1154–1157. doi: 10.1126/science.1206923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gubanova E, Brown B, Ivanov SV, et al. Downregulation of SMG-1 in HPV-positive head and neck squamous cell carcinoma due to promoter hypermethylation correlates with improved survival. Clin Cancer Res. 2012;18:1257–1267. doi: 10.1158/1078-0432.CCR-11-2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Voronkov A, Krauss S. Wnt/beta-catenin signaling and small molecule inhibitors. Curr Pharm Des. 2013;19:634–664. doi: 10.2174/138161213804581837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480. doi: 10.1016/j.cell.2006.10.018. [DOI] [PubMed] [Google Scholar]
- 38.Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149:1192–1205. doi: 10.1016/j.cell.2012.05.012. [DOI] [PubMed] [Google Scholar]
- 39.Mosimann C, Hausmann G, Basler K. Beta-catenin hits chromatin: regulation of Wnt target gene activation. Nat Rev Mol Cell Biol. 2009;10:276–286. doi: 10.1038/nrm2654. [DOI] [PubMed] [Google Scholar]
- 40.Uren A, Fallen S, Yuan H, et al. Activation of the canonical Wnt pathway during genital keratinocyte transformation: a model for cervical cancer progression. Cancer Res. 2005;65:6199–6206. doi: 10.1158/0008-5472.CAN-05-0455. [DOI] [PubMed] [Google Scholar]
- 41.Rodriguez-Sastre MA, Gonzalez-Maya L, Delgado R, et al. Abnormal distribution of E-cadherin and beta-catenin in different histologic types of cancer of the uterine cervix. Gynecol Oncol. 2005;97:330–336. doi: 10.1016/j.ygyno.2004.12.062. [DOI] [PubMed] [Google Scholar]
- 42.Shinohara A, Yokoyama Y, Wan X, et al. Cytoplasmic/nuclear expression without mutation of exon 3 of the beta-catenin gene is frequent in the development of the neoplasm of the uterine cervix. Gynecol Oncol. 2001;82:450–455. doi: 10.1006/gyno.2001.6298. [DOI] [PubMed] [Google Scholar]
- 43.Bulut G, Fallen S, Beauchamp EM, et al. Beta-catenin accelerates human papilloma virus type-16 mediated cervical carcinogenesis in transgenic mice. PLoS One. 2011;6:e27243. doi: 10.1371/journal.pone.0027243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rampias T, Boutati E, Pectasides E, et al. Activation of Wnt signaling pathway by human papillomavirus E6 and E7 oncogenes in HPV16-positive oropharyngeal squamous carcinoma cells. Mol Cancer Res. 2010;8:433–443. doi: 10.1158/1541-7786.MCR-09-0345. [DOI] [PubMed] [Google Scholar]
- 45.Aguilera O, Munoz A, Esteller M, et al. Epigenetic alterations of the Wnt/beta-catenin pathway in human disease. Endocr Metab Immune Disord Drug Targets. 2007;7:13–21. doi: 10.2174/187153007780059450. [DOI] [PubMed] [Google Scholar]
- 46.Pannone G, Bufo P, Santoro A, et al. WNT pathway in oral cancer: epigenetic inactivation of WNT-inhibitors. Oncol Rep. 2010;24:1035–1041. doi: 10.3892/or.2010.1035. [DOI] [PubMed] [Google Scholar]
- 47.Takeichi M. Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol. 1993;5:806–811. doi: 10.1016/0955-0674(93)90029-p. [DOI] [PubMed] [Google Scholar]
- 48.Onder TT, Gupta PB, Mani SA, et al. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 2008;68:3645–3654. doi: 10.1158/0008-5472.CAN-07-2938. [DOI] [PubMed] [Google Scholar]
- 49.Georgopoulos NT, Kirkwood LA, Walker DC, et al. Differential regulation of growth-promoting signalling pathways by E-cadherin. PLoS One. 2010;5:e13621. doi: 10.1371/journal.pone.0013621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wang DSL, Huang D, Zhang H, Shin D, Chen Z. Downregulation of E-cadherin enhances proliferation of head and neck cancer through transcriptional regulation of EGFR. Molecular Cancer. 2011;10:116. doi: 10.1186/1476-4598-10-116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Modjtahedi H, Dean C. The receptor for EGF and its ligands—expression, prognostic value and target for therapy in cancer (review) Int J Oncol. 1994;4:277–296. doi: 10.3892/ijo.4.2.277. [DOI] [PubMed] [Google Scholar]
- 52.Grandis JR, Tweardy DJ. Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer. Cancer Res. 1993;53:3579–3584. [PubMed] [Google Scholar]
- 53.Ongkeko WM, Altuna X, Weisman RA, et al. Expression of protein tyrosine kinases in head and neck squamous cell carcinomas. Am J Clin Pathol. 2005;124:71–76. doi: 10.1309/BTLN5WTMJ3PCNRRC. [DOI] [PubMed] [Google Scholar]
- 54.Sheu JJ, Hua CH, Wan L, et al. Functional genomic analysis identified epidermal growth factor receptor activation as the most common genetic event in oral squamous cell carcinoma. Cancer Res. 2009;69:2568–2576. doi: 10.1158/0008-5472.CAN-08-3199. [DOI] [PubMed] [Google Scholar]
- 55.Kim S, Grandis JR, Rinaldo A, et al. Emerging perspectives in epidermal growth factor receptor targeting in head and neck cancer. Head Neck. 2008;30:667–674. doi: 10.1002/hed.20859. [DOI] [PubMed] [Google Scholar]
- 56.Rubin Grandis J, Melhem MF, Gooding WE, et al. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst. 1998;90:824–832. doi: 10.1093/jnci/90.11.824. [DOI] [PubMed] [Google Scholar]
- 57.Temam S, Kawaguchi H, El-Naggar AK, et al. Epidermal growth factor receptor copy number alterations correlate with poor clinical outcome in patients with head and neck squamous cancer. J Clin Oncol. 2007;25:2164–2170. doi: 10.1200/JCO.2006.06.6605. [DOI] [PubMed] [Google Scholar]
- 58.Chung CH, Ely K, McGavran L, et al. Increased epidermal growth factor receptor gene copy number is associated with poor prognosis in head and neck squamous cell carcinomas. J Clin Oncol. 2006;24:4170–4176. doi: 10.1200/JCO.2006.07.2587. [DOI] [PubMed] [Google Scholar]
- 59.Ang KK, Berkey BA, Tu X, et al. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res. 2002;62:7350–7356. [PubMed] [Google Scholar]
- 60.Grandis JR, Tweardy DJ. TGF-alpha and EGFR in head and neck cancer. J Cell Biochem Suppl. 1993;17F:188–191. doi: 10.1002/jcb.240531027. [DOI] [PubMed] [Google Scholar]
- 61.Grandis JR, Chakraborty A, Zeng Q, et al. Downmodulation of TGF-alpha protein expression with antisense oligonucleotides inhibits proliferation of head and neck squamous carcinoma but not normal mucosal epithelial cells. J Cell Biochem. 1998;69:55–62. [PubMed] [Google Scholar]
- 62.Eriksen JG, Steiniche T, Sogaard H, et al. Expression of integrins and E-cadherin in squamous cell carcinomas of the head and neck. Apmis. 2004;112:560–568. doi: 10.1111/j.1600-0463.2004.apm1120902.x. [DOI] [PubMed] [Google Scholar]
- 63.Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9–22. doi: 10.1038/nrc2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cordes N, Seidler J, Durzok R, et al. beta1-integrin-mediated signaling essentially contributes to cell survival after radiation-induced genotoxic injury. Oncogene. 2006;25:1378–1390. doi: 10.1038/sj.onc.1209164. [DOI] [PubMed] [Google Scholar]
- 65.Eke I, Deuse Y, Hehlgans S, et al. beta(1)Integrin/FAK/cortactin signaling is essential for human head and neck cancer resistance to radiotherapy. J Clin Invest. 2012;122:1529–1540. doi: 10.1172/JCI61350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Surapureddi S, Yu S, Bu H, et al. Identification of a transcriptionally active peroxisome proliferator-activated receptor alpha-interacting cofactor complex in rat liver and characterization of PRIC285 as a coactivator. Proc Natl Acad Sci U S A. 2002;99:11836–11841. doi: 10.1073/pnas.182426699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Libby P. Current concepts of the pathogenesis of the acute coronary syndromes. Circulation. 2001;104:365–372. doi: 10.1161/01.cir.104.3.365. [DOI] [PubMed] [Google Scholar]
- 68.Michalik L, Desvergne B, Wahli W. Peroxisome-proliferator-activated receptors and cancers: complex stories. Nat Rev Cancer. 2004;4:61–70. doi: 10.1038/nrc1254. [DOI] [PubMed] [Google Scholar]
- 69.Jaeckel EC, Raja S, Tan J, et al. Correlation of expression of cyclooxygenase-2, vascular endothelial growth factor, and peroxisome proliferator-activated receptor delta with head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 2001;127:1253–1259. doi: 10.1001/archotol.127.10.1253. [DOI] [PubMed] [Google Scholar]
- 70.Ross SA, McCaffery PJ, Drager UC, et al. Retinoids in embryonal development. Physiol Rev. 2000;80:1021–1054. doi: 10.1152/physrev.2000.80.3.1021. [DOI] [PubMed] [Google Scholar]
- 71.Lotan R, Lotan D, Sacks PG. Inhibition of tumor cell growth by retinoids. Methods Enzymol. 1990;190:100–110. doi: 10.1016/0076-6879(90)90014-r. [DOI] [PubMed] [Google Scholar]
- 72.Ross SA, Jones CS, De Luca LM. Retinoic acid and methotrexate specifically increase PHA-E-lectin binding to a 67-kDa glycoprotein in LA-N-1 human neuroblastoma cells. Int J Cancer. 1995;62:303–308. doi: 10.1002/ijc.2910620312. [DOI] [PubMed] [Google Scholar]
- 73.Shen PF. Molecular mechanisms of chemoprevention and therapy of cancer by retinoids. Front Biosci. 2004;9:2663–2670. doi: 10.2741/1425. [DOI] [PubMed] [Google Scholar]
- 74.Sun SY, Lotan R. Retinoids and their receptors in cancer development and chemoprevention. Crit Rev Oncol Hematol. 2002;41:41–55. doi: 10.1016/s1040-8428(01)00144-5. [DOI] [PubMed] [Google Scholar]
- 75.Pitha-Rowe I, Petty WJ, Feng Q, et al. Microarray analyses uncover UBE1L as a candidate target gene for lung cancer chemoprevention. Cancer Res. 2004;64:8109–8115. doi: 10.1158/0008-5472.CAN-03-3938. [DOI] [PubMed] [Google Scholar]
- 76.Chan LS, Wells RA. Cross-talk between PPARs and the partners of RXR: a molecular perspective. PPAR Res. 2009;2009:925309. doi: 10.1155/2009/925309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Li M, Xiong ZG. Ion channels as targets for cancer therapy. Int J Physiol Pathophysiol Pharmacol. 2011;3:156–166. [PMC free article] [PubMed] [Google Scholar]
- 78.Herbst RS, Hong WK. IMC-C225, an anti-epidermal growth factor receptor monoclonal antibody for treatment of head and neck cancer. Semin Oncol. 2002;29:18–30. doi: 10.1053/sonc.2002.35644. [DOI] [PubMed] [Google Scholar]
- 79.Kim ES, Kies M, Herbst RS. Novel therapeutics for head and neck cancer. Curr Opin Oncol. 2002;14:334–342. doi: 10.1097/00001622-200205000-00014. [DOI] [PubMed] [Google Scholar]
- 80.Harris G, Ghazallah RA, Nascene D, et al. PPAR activation and decreased proliferation in oral carcinoma cells with 4-HPR. Otolaryngol Head Neck Surg. 2005;133:695–701. doi: 10.1016/j.otohns.2005.07.019. [DOI] [PubMed] [Google Scholar]
- 81.Olden K, Isaac L, Roberts L. Neighborhood-specific epigenome analysis: the pathway forward to understanding gene-environment interactions. N C Med J. 2011;72:125–127. [PubMed] [Google Scholar]
- 82.Garber K. Breaking the silence: the rise of epigenetic therapy. J Natl Cancer Inst. 2002;94:874–875. doi: 10.1093/jnci/94.12.874. [DOI] [PubMed] [Google Scholar]
- 83.Kopelovich L, Crowell JA, Fay JR. The epigenome as a target for cancer chemoprevention. J Natl Cancer Inst. 2003;95:1747–1757. doi: 10.1093/jnci/dig109. [DOI] [PubMed] [Google Scholar]
- 84.Lu D, Hoory T, Monie A, et al. Treatment with demethylating agent, 5-aza-2′-deoxycytidine enhances therapeutic HPV DNA vaccine potency. Vaccine. 2009;27:4363–4369. doi: 10.1016/j.vaccine.2009.02.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.