Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Virol. Feb 12, 2018; 7(1): 1-9
Published online Feb 12, 2018. doi: 10.5501/wjv.v7.i1.1
Retinoic acid receptor beta promoter methylation and risk of cervical cancer
Chaninya Wongwarangkana, Nasamon Wanlapakorn, Jira Chansaenroj, Yong Poovorawan, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
ORCID number: Chaninya Wongwarangkana (0000-0001-5625-7612); Nasamon Wanlapakorn (0000-0002-7953-275X​); Jira Chansaenroj (0000-0003-2610-1355); Yong Poovorawan (0000-0002-2337-6807).
Author contributions: All authors equally contributed to this paper with conception and design of the study, literature review and analysis, drafting and critical revision and editing, and final approval of the final version.
Supported by Research Chair Grant from the National Science and Technology Development Agency, No. P-15-50004; the Center of Excellence in Clinical Virology, Chulalongkorn Unversity and King Chulalongkourn Memorial Hospital, No. GCE 5900930-005; and the Rachadapisek Sompote Fund of Chulalongkorn University for postdoctoral fellowships to Chaninya Wongwarangkana.
Conflict-of-interest statement: No potential conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Yong Poovorawan, MD, Professor, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, 1873 Rama IV Road Patumwan, Bangkok 10330, Thailand. yong.p@chula.ac.th
Telephone: +66-2-2564909 Fax: +66-2-2564929
Received: October 6, 2017
Peer-review started: October 6, 2017
First decision: November 7, 2017
Revised: November 8, 2017
Accepted: December 6, 2017
Article in press: December 6, 2017
Published online: February 12, 2018

Abstract

Cervical cancer is one of the leading causes of death in women worldwide, particularly in developing countries. Human papillomavirus has been reported as one of the key etiologic factors in cervical carcinoma. Likewise, epigenetic aberrations have ability to regulate cancer pathogenesis and progression. Recent research suggested that methylation has been detected already at precancerous stages, which methylation markers may have significant value in cervical cancer screening. The retinoic acid receptor beta (RARβ) gene, a potential tumor suppressor gene, is usually expressed in normal epithelial tissue. Methylation of CpG islands in the promoter region of the RARβ gene has been found to be associated with the development of cervical cancer. To investigate whether RARβ methylation is a potential biomarker that predicts the progression of invasive cancer, we reviewed 14 previously published articles related to RARβ methylation. The majority of them demonstrated that the frequency of RARβ promoter methylation was significantly correlated with the severity of cervical epithelium abnormalities. However, methylation of a single gene may not represent the best approach for predicting disease prognosis. Analyzing combinations of aberrant methylation of multiple genes may increase the sensitivity, and thus this approach may serve as a better tool for predicting disease prognosis.

Key Words: Methylation, Cervical cancer, Retinoic acid receptor beta, Human papillomavirus, Risk correlation, Promoter

Core tip: The frequency of retinoic acid receptor beta promoter methylation was significantly correlated with the severity of cervical epithelium abnormalities. However, a single gene may not represent the best approach for predicting disease prognosis. Thus, combinations of aberrant methylation of multiple genes may as a better tool for predicting disease.



INTRODUCTION

Cervical cancer is the leading cause of death in women worldwide. The prevalence is high in women in low- to middle-income countries[1]. In 2012, approximately 522000 women globally were diagnosed with cervical cancer, and the mortality rate due to cervical cancer was reported to be 266,000 cases/year[2]. The highest incidence occurred in sub-Saharan Africa while in Asia, cervical cancer remains the third most common cancer (after breast and lung cancer), with an estimated 285000 new cases and 144000 deaths in 2012[3]. The age-standardized incidence rates (ASRs) of cervical cancer estimated by GLOBOSCAN in 2012 indicated that the ASR is higher in less developed compared to more developed regions[4]. In Thailand, the age group with the highest incidence is 45-70 years[5].

Several studies had found that cervical cancer is preceded by a pre-invasive stage, in which abnormal cells are confined to the cervical epithelium. The pre-invasive stage is also known as cervical intraepithelial neoplasia (CIN). The 2014 Bethesda System categorizes squamous epithelial cell abnormalities as atypical squamous cell of undetermined significance (AS-CUS); low-grade squamous intraepithelial lesion (LSIL), which was previously known as CIN I; high-grade squamous intraepithelial lesion (HSIL), which was previously known as CIN II and III; or squamous cell carcinoma (SCC)[6]. SCC represents > 80% of cervical cancers, while adenocarcinoma (AC) accounts for the rest.

The standard method for screening for early-stage cervical neoplasia is cytological morphologic assessment of cervical scrapings. The sensitivity of the conventional Pap smear for identifying CIN II+ is 55.2%, while the sensitivity of liquid-based cytology is 57.1%[7]. High-risk human papillomavirus (HPV) DNA testing in combination with the conventional Pap smear increases the sensitivity. Furthermore, biomarkers of oncogenic progression would improve the accuracy of cancer progression predictions. Epigenetic biomarkers may help to fulfil this role, and they have the additional benefit predicting the stage of cervical carcinogenesis progression[8].

GENOME OF HPV

HPV is a small, non-enveloped and circular double-stranded DNA virus with a genome of approximately 8 kb in length[9]. The HPV genome comprises eight protein-coding genes and a noncoding region that is referred to as the regulatory long control region[10]. Only one strand of the DNA carries the protein-coding sequence[11]. Regarding the protein-coding genes, the genes are designated as early (E) or late (L) to indicate when the proteins are expressed in the viral life cycle[12]. The eight protein-coding gene consist of E1, E2, E4, E5, E6, E7, L1 and L2[9]. E1 and E2 are highly conserved and involved in viral DNA replication[13-15]. L1 and L2, which both have a high degree of sequence variation, encode for viral packaging proteins[16]. E4 releases the viral particle from the epithelial cells[17]. E6 and E7 are viral oncogenes that are involved in the integration of the HPV genome into the host genome[18]. There are more than 130 genotypes of HPV, which are categorized based on sequence variation in their L1 region[19]. Of the 130 genotypes, at least 40 genotypes infect the genital areas of humans via sexual transmission. HPV can also be classified into cutaneous or mucosal types[12]. The mucosal type can be subdivided into high-, intermediate-, or low-risk types[20].

HPV AND CERVICAL CANCER

The most important risk factor for cervical cancer is HPV infection, which has been found in 90.7% of cervical cancer patients worldwide[21]. HPV infection is a sexually transmitted disease. It has been estimated that more than 80% of sexually active women become infected with HPV, while more than 50% of young women become infected after they first have sexual intercourse[22]. The oncogenic potential of HPV depends on the genotype. HPV 16 and 18 are the most common types associated with invasive cervical cancer[23]. Other HPV genotypes have been found to be related to cancer, but their oncogenic risk differs among the various populations, geographic regions, and age groups.

At the country level, collecting baseline data on the local burden of specific HPV genotypes related to cervical cancer is important. This information can impact the local HPV vaccination policies. A meta-analysis revealed that HPV 16, 18, 31, 33, 45, 52, and 58 are responsible for more than 90% of cervical cancers worldwide[20]. These genotypes represent the baseline genotypes to include in a vaccine targeting the genotypes circulating in the population[4]. The current HPV vaccines were developed to prevent HPV infection, and thus prevent cervical carcinoma. HPV vaccines have been implemented in routine vaccination programs in several developed and developing countries worldwide[24]. To date, there have been three HPV vaccines in clinical use: Bivalent, quadrivalent, and nanovalent vaccines[25].

Other independent risk factors such as immunosuppression, individual lifestyle, and smoking have been found to be associated with the development of HPV-related cervical cancer[21,26]. Most HPV infection is transient, and clearance of the virus can occur spontaneously over a 3-year period[27]. However, in some cases, persistent infection can result in cervical cancer development. The transition from dysplasia to invasive carcinoma may take several years to decades to develop. HPV initially infects the basal layers of the epithelium through micro-wounds. The virus begins to replicate, and when infected daughter cells migrate to the upper layers of the epithelium, the viral late genes are activated, and viral DNA is packaged into capsids. Progeny virions are released to re-initiate infection, which can result in persistent and/or asymptomatic infection[28]. The integration of HPV into the host genome can lead to carcinogenic transformation. Certain regions of the human genome are favored for viral DNA insertion such as fragile sites, rupture points, translocation points, and transcriptionally active regions[29]. Moreover, the virus can induce epigenetic modification of viral and cellular genes, which affect their expression, leading to malignant cell transformation[30,31].

HOST GENETIC FACTORS AND CERVICAL CANCER

Diverse immunogenetic associations with HPV infection, persistence, and transformation have been extensively investigated. Recent studies have looked at multiple genes in various populations with different environment interactions[32]. HPV infection alone might not be sufficient for the development of cervical carcinoma, and certain antigen-processing machinery (APM) and single-nucleotide polymorphisms (SNPs) may lead to a smaller immunogenic peptide repertoire for presentation to local immune cells. This can result in further attenuation of cytokine and receptor expression, which leads to an ineffective overall immune response and progression to carcinoma[33]. The Genome-Wide Association Study (GWAS) for polymorphisms of host immune response genes showed that variation in several genes contributes to different risks of cervical cancer. The integrative approach, which is also known as systems biology, could help explain the complexity of host–virus interactions and provide a better understanding that may eventually lead to personalized prevention, diagnosis, and treatment[34-36].

The detection of methylated genes in cervical specmens is a feasible technique and represents a potential source of biomarkers that are of relevance to carcinogenesis. In particular, there are methylation markers that, among HPV-infected women, indicate the presence of CIN II+ and risk of cancer[37].

High expression levels of certain oncoproteins in cervical cells have been found to be associated with cervical carcinoma. One study found a strong correlation between centromere protein H (CENP-H) expression and cervical carcinoma in a Chinese population[38]. Another study found that expression of the B-cell-specific Moloney leukemia virus insert site 1 (Bmi-1), P16, and CD44v6 (a CD44 variant) were significantly higher in cervical carcinoma tissues compared with precancerous lesions and normal tissues[39]. In addition, abnormalities in the phosphatidylinositide 3-kinase (PI3K) pathway induced by mutations in PI3K catalytic subunit α (PIK3CA) were associated with shorter survival in cervical cancer patients[40]. Recently, deep sequencing of somatic mutations has identified several novel mutations in carcinoma cells, including E322K in the mitogen-activated protein kinase 1 (MAPK1) gene, inactivating mutations in the major histocompatibility complex, class I, B (HLA-B) gene, and mutations in F-box and WD repeat domain containing 7 (FBXW7), tumor protein p53 (TP53), and Erb-B2 receptor tyrosine kinase 2 (ERBB2)[41].

EPIGENETIC MECHANISMS AND RISK OF CANCER DEVELOPMENT

Recent studies also investigated epigenetic mechanisms related to HPV infection, including methylation of the host and viral genes, and chromatin modification in host cells[42]. Epigenetic mechanisms affect gene regulation without changing the genetic sequences, and these mechanisms have been increasingly found to be associated with cancer development[43]. The main epigenetic mechanism is methylation patterning, which occurs to various extents in different DNA and proteins. DNA methylation is a mechanism of gene regulation that typically occurs in CpG dinucleotide contexts, resulting in genomic instability. Methylation of heterochromatin and promoter regions is associated with decreased gene transcription. Several studies have found that DNA methylation frequently occurs in cervical cells but rarely in normal cells, suggesting that their methylation is highly related to the severity of cervical neoplasia[44]. Several markers have been evaluated extensively in studies involving women with precancerous and cancerous cervical lesions[44-46]. Epigenetic methylation in the promoter region of several tumor suppressor genes (TSGs) has been detected in precancerous cervical cells[47,48]. Genes that were found to be significantly associated with promoter methylation were RASSF1A and MGMT (involved in DNA repair), CDKN2A (involved in cell cycle control), PYCARD (involved in apoptosis), and APC and SFRP1 (involved in Wnt signaling)[49].

One striking conclusion of previous studies was that methylation frequencies for the same gene vary widely between studies. It was difficult to identify highly consistent results for most genes even when restricting analyses to studies of similar size or those that used common specimen sources or similar assays. This suggests that the frequency of certain methylation markers may also vary for reasons related to differences in populations, specific features of assay protocols, chance, or other unidentified factors. The most important prerequisite for a potential biomarker is that it must be reliable in its measurement. There is a possibility that the wide range of frequencies reported for some genes (in contrast to the more consistent measurement of a few other genes in similar studies) could be related to unreliable assays for these specific genes rather than biological variation. Another prerequisite for a good biomarker is that it has high sensitivity and high specificity for disease detection, resulting in a high positive predictive value. Several studies have proposed the use of methylated gene panels in order to obtain optimal assessment performance for cervical cancer screening[47,50].

Retinoic acid (RA) is an essential regulator of normal epithelial cell differentiation. The effect of RA is mediated by two types of nuclear receptors, the retinoic-acid receptor (RAR) family and retinoid-X receptor (RXR) family. Both of these receptor families have three members (alpha, beta, and gamma), which are encoded by distinct genes in vertebrates. The retinoic acid receptor beta (RARβ) gene encodes a nuclear receptor that binds RA and mediates cellular signaling. It is important during differentiation of stratified squamous epithelium, including cervical epithelium. It is considered to be a potential TSG. The RARβ gene is usually expressed in normal epithelial tissue. The direct roles of the RARβ protein include regulating gene expression and differentiation, immune modulation, and inducing apoptosis. Previous studies revealed that the RARβ gene is downregulated in high-grade lesions[51]. RARβ gene silencing was observed in carcinoma cells[52]. Recent research suggested that the RARβ protein can suppress cervical carcinogenesis and may play a role in the early development of cancer[51]. CpG methylation of the 5’ region of the RARβ gene contributes to gene silencing, and this methylation is associated with increased grades of SIL and invasive cervical cancer. Many studies have revealed that methylation of CpG islands in the promoter region of the RARβ gene induces repression of RARβ expression in several epithelial carcinomas, including cervical cancer[53-55].

The risk of cervical cancer due to RARβ methylation remains inconsistent across different studies[51,52,56]. Therefore, we reviewed previously published articles and summarized the relationship between RARβ promoter methylation and cervical cancer (Table 1).

Table 1 The summary of the articles that investigated the methylation of RARβ gene in tumor tissue from women diagnosed with squamous intraepithelial lesion and cervical cancer.
Ref.Year of publicationNationality of participantsSample sizeSource of samplesLab techniqueRARβ methylation results
Virmani et al[57]2001AmericanNormal/LSIL = 37Normal/LSIL/HSIL from liquid-based cytology specimenMSPRARβ methylation positive in
HSIL = 17ICC from biopsy tissueNormal/LSIL = 11% HSIL = 29%
ICC = 19ICC = 26%
Narayan et al[56]2003ColombiansNormal = 8Normal = cells from cervical swab LSIL/HSIL = formalin-fixed and paraffin-embeddedMSPRARβ methylation positive in
GermanLSIL = 9cervical tissuesNormal = 0% SCC/AC = 29.3%
AmericanHSIL = 30SCC/AC = tumor biopsiesImmuno-histochemistry of RARβ proteinImmunohistochemistry
SCC = 77LSIL; 11% showed low expression
AC = 5HSIL; 60% showed complete lack of expression
Gustafson et al[37]2004AmericanNormal = 11Liquid-based cytology specimenNested MSPRARβ methylation positive in
LSIL = 17Normal = 0% LSIL = 0%
HSIL = 11HSIL = 9.1%
Feng et al[58]2005SenegaleseNormal/ASCUS = 142Exfoliated cervical cells and tissue biopsyMSPRARβ methylation positive in
CIN I = 39Normal/ASCUS = 3.2%
CIN II = 23CIN I = 0%
CIN III = 23CIN II = 0%
ICC = 92CIN III = 15.8% ICC = 38.2%
Wisman et al[59]2006DutchNormal = 19Cervical scrapingQMSPThe percentage of RARβ methylation level above control ratio were detected in Normal = 0% SCC = 15% AC = 25%
SCC = 20
AC = 8
Choi et al[60]2007KoreanNormal = 37Normal cells were from hysterectomy due to myomaMSPRARβ methylation positive in
SCC = 37Cancer cells were from tissue after surgeryImmuno-histochemistry of RARβ proteinNormal = 0% SCC = 41%
Immunostaining normal = strong staining
SCC = 43% absent staining
Zhang et al[52]2007Japanese and ChineseNormal = 6Cervical tissue by biopsy or surgeryReal-time PCR for RARβ mRNARARβ expression level among normal cells: All were highly expressed
ICC = 17RARb2 expression level among cancer cells:
Semi-nested MSP13/17: Completely repressed
2/17: Highly repressed
2/17: Moderately down-regulated
Among 13 samples with completely repressed mRNA expression
9 promoter methylated, 4 unmethylated
Flatley et al[2]2009EnglishNormal = 58Exfoliated cervical cells and cervical biopsyNested MSPRARβ methylation positive in
CIN I = 68Normal = 6.5%
CIN II = 56CIN I = 42.6%
CIN III = 76CIN II = 6.3%
ICC = 50CIN III = 0% ICC = 15.9%
Kim et al[54]2010KoreanNormal = 41Liquid based cytology specimenMultiplex nested MSPRARβ methylation positive in
LSIL = 32Normal = 4.9% LSIL = 15.6%
HSIL = 67 SCC = 69HSIL = 46.3% SCC = 53.6%
Kim et al[61]2010KoreanNormal = 28Liquid based cytology specimenMultiplex QMSPRARβ methylation level
LSIL = 26Normal = 1.59+3.51% LSIL = 3.67+9.09%
HSIL = 45 SCC = 63HSIL = 21.93+20.10% SCC = 19.06+19.39%
Yang et al[62]2010DutchNormal = 20Biopsy tissueQMSPRARβ methylation positive (from tissue) in
LSIL = 20Normal = 85% LSIL = 65%
HSIL = 20Cervical scraping only available in subset of samplesHSIL = 75% SCC = 85% AC = 85%
SCC = 40RARβ methylation positive (from scraping) in
AC = 20Normal = 44% LSIL = 37.5%
HSIL = 55.6% SCC = 83.8% AC = 100%
The median methylation level increased significantly with the severity of lesion (P < 0.05)
Pathak et al[63]2012IndianNormal = 35Normal cells from hysterectomy SIL from excision ICC from tissue biopsyMSPRARβ methylation positive in
SIL = 27Normal = 11.4% SIL = 55.5% ICC = 57.8%
ICC = 38
Milutin Gašperov et al[64]2015CroatianNormal = 40Cervical scrapingMSPRARβ methylation positive in
CIN I = 40Normal = 62.5%
CIN II = 40CIN I = 35%
CIN III = 42CIN II = 61.5%
SCC = 8 AC = 3CIN III = 61.9% SCC/AC = 90%
Sun et al[51]2015ChineseNormal = 48Liquid based cytology specimenMethylation specific high resolution melting analysis (Quantitative)RARβ methylation positive in
CIN I = 54Normal = 31.3% CIN I = 35.2%
CIN II = 47CIN II and III = 28.2% SCC = 33.3%
CIN III = 56RARβ methylation level: none = 68.8%
SCC = 450-5% methylation = 26.4% 5-25% = 4.8%

Among the 14 articles reviewed, the majority of them (11/14) demonstrated that the frequency of RARβ promoter methylation was significantly correlated with severity of cervical epithelium abnormalities. Three studies did not concur with this finding. The first study was conducted in 2003 with a small sample size and no cancer patients were involved[37]. The other two studies were conducted in 2010 and 2015. Both studies found that normal tissue also had RARβ promoter methylation, which made it a poor predictor of progression to severe disease[62,64]. However, one of the two studies also investigated the level of methylation using quantitative methylation-specific PCR and found that although normal cells were methylated, the level of methylation increased in LSIL, HSIL, and invasive cancer tissue[62].

In addition, both Narayan et al[56] and Choi et al[60] found that RARβ promoter methylation was associated with cervical cancer prognosis. Narayan et al[56] found that 80% of the patients with RARβ methylation either died of cancer or only partly responded to treatment, while Choi et al[60] found that absence or reduction of RARβ protein expression was associated with a higher level of SCC antigen (P = 0.04) and more frequent lymph node metastasis (P = 0.023).

A study of the frequency of RARβ promoter methylation in urine and cervical samples from Senegalese women and cervical epithelial cell abnormalities found that methylation was significantly greater in abnormal specimens (and the results from the urine samples correlated with the results from the cervical swab samples)[58,65]. Another study by Zhang et al[52] compared the frequency of methylation with RARβ mRNA expression. The authors found that in normal cervical cells, the RARβ gene was highly expressed. In contrast, among 17 samples from patients with invasive cervical carcinoma, RARβ2 expression was completely repressed in 13 samples, highly repressed in 2 samples, and moderately downregulated in 2 samples. Among the 13 samples with completely repressed RARβ2 expression, the RARβ promoter region was methylated in 9 samples and unmethylated in 4 samples. The authors then further investigated the silencing mechanism and discovered that apart from methylation, repressive histone modifications also played a role in gene silencing, which could contribute to the development of cervical carcinoma.

Four studies performed a quantitative assessment of methylation. The first study was conducted in 2006 by Wisman et al[59], who found that the RARβ2 promoter was more methylated in cervical cancer than in control tissue. Four years later, Kim et al[61] found that the RARβ methylation level in normal tissue was 1.59% ± 3.51% whereas, in HSIL and SCC, it was 21.93% ± 20.10% and 19.06% ± 19.39%, respectively. The third study, by Yang et al[62], also highlighted that although the percentage of methylated samples was very high in normal tissue, the level of methylation correlated with disease severity. The last study was conducted by Sun et al[51] in 2015. They found that among 250 cervical samples from healthy individuals and patients with various stages of cervical epithelium abnormalities, the percentage of methylation in patients showed that 68.8% had no RARβ promoter methylation, 26.4% had 0%-5% methylation, and 4.8% had 5%-25% methylation. No samples had methylation levels above 25%.

In addition, two studies performed immunohistochemistry staining of the RARβ protein in cervical cells. Narayan et al[56] found that in the LSIL group, 11% had low RARβ expression whereas, in the HSIL group, 60% had a complete lack of RARβ expression. This finding suggested that the downregulation of the RARβ gene occurs early in the development of cervical carcinoma[56]. The second study was carried out by Choi et al[60], who discovered that all normal tissues highly expressed the RARβ protein, whereas no staining was detected in 43% of the SCC tissues.

Almost of cancer cell lines and primary cancer tissues examined, the RARβ2 was repressed. The repression was frequently associated with promoter methylation, which causes lack of gene expression. These results strongly support the hypothesis that promoter methylation is the epigenetic cause of RARβ2 repression in cervical cancers harboring methylated RARβ2 promoters. A DNA demethylating reagent can reactivate gene expression by inducing drastic demethylation of the promoter in repressed cells carrying a methylated promoter[44]. This consistency between promoter demethylation and RARβ2 derepression strongly suggests that the primary cause of RARβ2 repression is indeed promoter methylation.

Several hypotheses have been proposed regarding the mechanisms of DNA methylation that lead to silencing of genes. In some cancer cells and tissues examined, RARβ2 was repressed without promoter methylation. These facts indicate that although DNA methylation is the major epigenetic mechanism for gene silencing, there are other epigenetic silencing pathways independent of DNA methylation. RARβ2 is frequently silenced in cervical cancers by one of two epigenetic mechanisms. One is DNA methylation, a well-known epigenetic mechanism leading to transcriptional silencing of genes, while the other involves the formation of repressive histone modifications near the promoter, by unknown mechanisms independent of DNA methylation. At present, the initial causes of these epigenetic changes during carcinogenesis are unclear. RARβ2 silenced by promoter methylation can be reactivated by promoter hypomethylation. This result indicates the importance of examining promoter methylation if epigenetic modulation drugs are to be used for chemotherapy in patients with cervical cancers.

In conclusion, DNA methylation of TSGs likely contributes to the development of cancer. Although DNA methylation of only one gene may not represent the complete process of epigenetic silencing, it has been shown to be significantly correlated with cervical cancer. Analyzing combinations of aberrant hyper- or hypo-methylation of multiple genes may increase the sensitivity of prognoses. Thus, this approach may serve as a better tool for predicting disease progression. Risk factors should also be further characterized to better understand the pathogenesis of cervical carcinoma.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Virology

Country of origin: Thailand

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D, D

Grade E (Poor): 0

P- Reviewer: Chen C, Chen CJ, Ciotti M S- Editor: Ji FF L- Editor: A E- Editor: Li RF

References
1.  Ginsburg O, Bray F, Coleman MP, Vanderpuye V, Eniu A, Kotha SR, Sarker M, Huong TT, Allemani C, Dvaladze A. The global burden of women’s cancers: a grand challenge in global health. Lancet. 2017;389:847-860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 556]  [Cited by in F6Publishing: 560]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
2.  Flatley JE, McNeir K, Balasubramani L, Tidy J, Stuart EL, Young TA, Powers HJ. Folate status and aberrant DNA methylation are associated with HPV infection and cervical pathogenesis. Cancer Epidemiol Biomarkers Prev. 2009;18:2782-2789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 251]  [Reference Citation Analysis (0)]
3.  Vaccarella S, Laversanne M, Ferlay J, Bray F. Cervical cancer in Africa, Latin America and the Caribbean and Asia: Regional inequalities and changing trends. Int J Cancer. 2017;141:1997-2001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 84]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
4.  Wagner M, Bennetts L, Patel H, Welner S, de Sanjose S, Weiss TW. Global availability of data on HPV genotype-distribution in cervical, vulvar and vaginal disease and genotype-specific prevalence and incidence of HPV infection in females. Infect Agent Cancer. 2015;10:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
5.  Wilailak S, Lertchaipattanakul N. The epidemiologic status of gynecologic cancer in Thailand. J Gynecol Oncol. 2016;27:e65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
6.  Nayar R, Wilbur DC. The Pap Test and Bethesda 2014. Acta Cytol. 2015;59:121-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 122]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
7.  Arbyn M, Bergeron C, Klinkhamer P, Martin-Hirsch P, Siebers AG, Bulten J. Liquid compared with conventional cervical cytology: a systematic review and meta-analysis. Obstet Gynecol. 2008;111:167-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 260]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
8.  Fang J, Zhang H, Jin S. Epigenetics and cervical cancer: from pathogenesis to therapy. Tumour Biol. 2014;35:5083-5093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 93]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
9.  Zheng ZM, Baker CC. Papillomavirus genome structure, expression, and post-transcriptional regulation. Front Biosci. 2006;11:2286-2302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 289]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
10.  Bernard HU, Calleja-Macias IE, Dunn ST. Genome variation of human papillomavirus types: phylogenetic and medical implications. Int J Cancer. 2006;118:1071-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 177]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
11.  Graham SV. Human papillomavirus: gene expression, regulation and prospects for novel diagnostic methods and antiviral therapies. Future Microbiol. 2010;5:1493-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
12.  Sanclemente G, Gill DK. Human papillomavirus molecular biology and pathogenesis. J Eur Acad Dermatol Venereol. 2002;16:231-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 59]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
13.  Clertant P, Seif I. A common function for polyoma virus large-T and papillomavirus E1 proteins? Nature. 1984;311:276-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 117]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
14.  Lusky M, Fontane E. Formation of the complex of bovine papillomavirus E1 and E2 proteins is modulated by E2 phosphorylation and depends upon sequences within the carboxyl terminus of E1. Proc Natl Acad Sci USA. 1991;88:6363-6367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 82]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
15.  Piccini A, Storey A, Romanos M, Banks L. Regulation of human papillomavirus type 16 DNA replication by E2, glucocorticoid hormone and epidermal growth factor. J Gen Virol. 1997;78:1963-1970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
16.  Ma B, Roden RB, Hung CF, Wu TC. HPV pseudovirions as DNA delivery vehicles. Ther Deliv. 2011;2:427-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
17.  Doorbar J. The E4 protein; structure, function and patterns of expression. Virology. 2013;445:80-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 147]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
18.  Kessis TD, Connolly DC, Hedrick L, Cho KR. Expression of HPV16 E6 or E7 increases integration of foreign DNA. Oncogene. 1996;13:427-431.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Calleja-Macias IE, Kalantari M, Allan B, Williamson AL, Chung LP, Collins RJ, Zuna RE, Dunn ST, Ortiz-Lopez R, Barrera-Saldaña HA. Papillomavirus subtypes are natural and old taxa: phylogeny of human papillomavirus types 44 and 55 and 68a and -b. J Virol. 2005;79:6565-6569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
20.  Lorincz AT, Reid R, Jenson AB, Greenberg MD, Lancaster W, Kurman RJ. Human papillomavirus infection of the cervix: relative risk associations of 15 common anogenital types. Obstet Gynecol. 1992;79:328-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 652]  [Cited by in F6Publishing: 691]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
21.  Muñoz N, Bosch FX, de Sanjosé S, Herrero R, Castellsagué X, Shah KV, Snijders PJ, Meijer CJ; International Agency for Research on Cancer Multicenter Cervical Cancer Study Group. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med. 2003;348:518-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4165]  [Cited by in F6Publishing: 3980]  [Article Influence: 189.5]  [Reference Citation Analysis (0)]
22.  Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ, Muñoz N. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol. 1999;189:12-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
23.  Adefuye PO, Broutet NJ, de Sanjosé S, Denny LA. Trials and projects on cervical cancer and human papillomavirus prevention in sub-Saharan Africa. Vaccine. 2013;31 Suppl 5:F53-F59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Markowitz LE, Tsu V, Deeks SL, Cubie H, Wang SA, Vicari AS, Brotherton JM. Human papillomavirus vaccine INTRODUCTION--the first five years. Vaccine. 2012;30 Suppl 5:F139-F148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 222]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
25.  Bonanni P, Zanella B, Santomauro F, Lorini C, Bechini A, Boccalini S. Safety and perception: What are the greatest enemies of HPV vaccination programmes? Vaccine. 2017; pii: S0264-410X(17)30730-2 Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
26.  Plummer M, Peto J, Franceschi S; International Collaboration of Epidemiological Studies of Cervical Cancer. Time since first sexual intercourse and the risk of cervical cancer. Int J Cancer. 2012;130:2638-2644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
27.  Rositch AF, Koshiol J, Hudgens MG, Razzaghi H, Backes DM, Pimenta JM, Franco EL, Poole C, Smith JS. Patterns of persistent genital human papillomavirus infection among women worldwide: a literature review and meta-analysis. Int J Cancer. 2013;133:1271-1285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 144]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
28.  Narisawa-Saito M, Kiyono T. Basic mechanisms of high-risk human papillomavirus-induced carcinogenesis: roles of E6 and E7 proteins. Cancer Sci. 2007;98:1505-1511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 307]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
29.  Das P, Thomas A, Mahantshetty U, Shrivastava SK, Deodhar K, Mulherkar R. HPV genotyping and site of viral integration in cervical cancers in Indian women. PLoS One. 2012;7:e41012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
30.  Saavedra KP, Brebi PM, Roa JC. Epigenetic alterations in preneoplastic and neoplastic lesions of the cervix. Clin Epigenetics. 2012;4:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
31.  Sasagawa T, Takagi H, Makinoda S. Immune responses against human papillomavirus (HPV) infection and evasion of host defense in cervical cancer. J Infect Chemother. 2012;18:807-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
32.  Hong EP, Park JW. Sample size and statistical power calculation in genetic association studies. Genomics Inform. 2012;10:117-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 319]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
33.  Mehta AM, Mooij M, Branković I, Ouburg S, Morré SA, Jordanova ES. Cervical Carcinogenesis and Immune Response Gene Polymorphisms: A Review. J Immunol Res. 2017;2017:8913860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
34.  Chen D, Gyllensten U. Lessons and implications from association studies and post-GWAS analyses of cervical cancer. Trends Genet. 2015;31:41-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
35.  Chen D, Juko-Pecirep I, Hammer J, Ivansson E, Enroth S, Gustavsson I, Feuk L, Magnusson PK, McKay JD, Wilander E. Genome-wide association study of susceptibility loci for cervical cancer. J Natl Cancer Inst. 2013;105:624-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 126]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
36.  Yan Q. Immunoinformatics and systems biology methods for personalized medicine. Methods Mol Biol. 2010;662:203-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
37.  Gustafson KS, Furth EE, Heitjan DF, Fansler ZB, Clark DP. DNA methylation profiling of cervical squamous intraepithelial lesions using liquid-based cytology specimens: an approach that utilizes receiver-operating characteristic analysis. Cancer. 2004;102:259-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
38.  Weng MY, Li L, Feng SY, Hong SJ. Expression of Bmi-1, P16, and CD44v6 in Uterine Cervical Carcinoma and Its Clinical Significance. Cancer Biol Med. 2012;9:48-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
39.  Weng MY, Li L, Hong SJ, Feng SY. Clinical Significance of CENP-H Expression in Uterine Cervical Cancer. Cancer Biol Med. 2012;9:192-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
40.  Wright AA, Howitt BE, Myers AP, Dahlberg SE, Palescandolo E, Van Hummelen P, MacConaill LE, Shoni M, Wagle N, Jones RT. Oncogenic mutations in cervical cancer: genomic differences between adenocarcinomas and squamous cell carcinomas of the cervix. Cancer. 2013;119:3776-3783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 194]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
41.  Ojesina AI, Lichtenstein L, Freeman SS, Pedamallu CS, Imaz-Rosshandler I, Pugh TJ, Cherniack AD, Ambrogio L, Cibulskis K, Bertelsen B. Landscape of genomic alterations in cervical carcinomas. Nature. 2014;506:371-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 543]  [Cited by in F6Publishing: 595]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
42.  Di Domenico M, Giovane G, Kouidhi S, Iorio R, Romano M, De Francesco F, Feola A, Siciliano C, Califano L, Giordano A. HPV epigenetic mechanisms related to Oropharyngeal and Cervix cancers. Cancer Biol Ther. 2017; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
43.  Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3:415-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3397]  [Cited by in F6Publishing: 3607]  [Article Influence: 164.0]  [Reference Citation Analysis (0)]
44.  Wang KH, Lin CJ, Liu CJ, Liu DW, Huang RL, Ding DC, Weng CF, Chu TY. Global methylation silencing of clustered proto-cadherin genes in cervical cancer: serving as diagnostic markers comparable to HPV. Cancer Med. 2015;4:43-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
45.  Huang TH, Lai HC, Liu HW, Lin CJ, Wang KH, Ding DC, Chu TY. Quantitative analysis of methylation status of the PAX1 gene for detection of cervical cancer. Int J Gynecol Cancer. 2010;20:513-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
46.  Lai HC, Ou YC, Chen TC, Huang HJ, Cheng YM, Chen CH, Chu TY, Hsu ST, Liu CB, Hung YC. PAX1/SOX1 DNA methylation and cervical neoplasia detection: a Taiwanese Gynecologic Oncology Group (TGOG) study. Cancer Med. 2014;3:1062-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
47.  Clarke MA, Luhn P, Gage JC, Bodelon C, Dunn ST, Walker J, Zuna R, Hewitt S, Killian JK, Yan L. Discovery and validation of candidate host DNA methylation markers for detection of cervical precancer and cancer. Int J Cancer. 2017;141:701-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 53]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
48.  Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31:27-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1689]  [Cited by in F6Publishing: 1715]  [Article Influence: 114.3]  [Reference Citation Analysis (0)]
49.  Heyn H, Esteller M. DNA methylation profiling in the clinic: applications and challenges. Nat Rev Genet. 2012;13:679-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 567]  [Cited by in F6Publishing: 549]  [Article Influence: 45.8]  [Reference Citation Analysis (0)]
50.  Catarino R, Petignat P, Dongui G, Vassilakos P. Cervical cancer screening in developing countries at a crossroad: Emerging technologies and policy choices. World J Clin Oncol. 2015;6:281-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 146]  [Cited by in F6Publishing: 125]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
51.  Sun Y, Li S, Shen K, Ye S, Cao D, Yang J. DAPK1, MGMT and RARB promoter methylation as biomarkers for high-grade cervical lesions. Int J Clin Exp Pathol. 2015;8:14939-14945.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Zhang Z, Joh K, Yatsuki H, Zhao W, Soejima H, Higashimoto K, Noguchi M, Yokoyama M, Iwasaka T, Mukai T. Retinoic acid receptor beta2 is epigenetically silenced either by DNA methylation or repressive histone modifications at the promoter in cervical cancer cells. Cancer Lett. 2007;247:318-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
53.  Gurioli G, Salvi S, Martignano F, Foca F, Gunelli R, Costantini M, Cicchetti G, De Giorgi U, Sbarba PD, Calistri D. Methylation pattern analysis in prostate cancer tissue: identification of biomarkers using an MS-MLPA approach. J Transl Med. 2016;14:249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
54.  Kim JH, Choi YD, Lee JS, Lee JH, Nam JH, Choi C. Assessment of DNA methylation for the detection of cervical neoplasia in liquid-based cytology specimens. Gynecol Oncol. 2010;116:99-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
55.  Mariano FV, Egal ES, Pramio D, Fidalgo F, Sara É, Costa AF, de Oliveira Gondak R, Coletta RD, de Almeida OP, Kowalski LP, Victorino Krepischi AC, Altemani A. Evaluation of a subset of tumor suppressor gene for copy number and epigenitic changes in pleomorphic adenoma and carcinoma ex-pleomorphic adenoma carcinogenesis. Oral Surg Oral Med Oral Pathol Oral Radiol. 2016;122:322-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
56.  Narayan G, Arias-Pulido H, Koul S, Vargas H, Zhang FF, Villella J, Schneider A, Terry MB, Mansukhani M, Murty VV. Frequent promoter methylation of CDH1, DAPK, RARB, and HIC1 genes in carcinoma of cervix uteri: its relationship to clinical outcome. Mol Cancer. 2003;2:24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 171]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
57.  Virmani AK, Muller C, Rathi A, Zoechbauer-Mueller S, Mathis M, Gazdar AF. Aberrant methylation during cervical carcinogenesis. Clin Cancer Res. 2001;7:584-589.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Feng Q, Balasubramanian A, Hawes SE, Toure P, Sow PS, Dem A, Dembele B, Critchlow CW, Xi L, Lu H. Detection of hypermethylated genes in women with and without cervical neoplasia. J Natl Cancer Inst. 2005;97:273-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 144]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
59.  Wisman GB, Nijhuis ER, Hoque MO, Reesink-Peters N, Koning AJ, Volders HH, Buikema HJ, Boezen HM, Hollema H, Schuuring E. Assessment of gene promoter hypermethylation for detection of cervical neoplasia. Int J Cancer. 2006;119:1908-1914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 85]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
60.  Choi CH, Lee KM, Choi JJ, Kim TJ, Kim WY, Lee JW, Lee SJ, Lee JH, Bae DS, Kim BG. Hypermethylation and loss of heterozygosity of tumor suppressor genes on chromosome 3p in cervical cancer. Cancer Lett. 2007;255:26-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
61.  Kim JH, Choi YD, Lee JS, Lee JH, Nam JH, Choi C, Kweon SS, Fackler MJ, Sukumar S. Quantitative assessment of DNA methylation for the detection of cervical neoplasia in liquid-based cytology specimens. Virchows Arch. 2010;457:35-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
62.  Yang N, Nijhuis ER, Volders HH, Eijsink JJ, Lendvai A, Zhang B, Hollema H, Schuuring E, Wisman GB, van der Zee AG. Gene promoter methylation patterns throughout the process of cervical carcinogenesis. Cell Oncol. 2010;32:131-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 28]  [Reference Citation Analysis (0)]
63.  Pathak S, Bhatla N, Singh N. Cervical cancer pathogenesis is associated with one-carbon metabolism. Mol Cell Biochem. 2012;369:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
64.  Milutin Gašperov N, Sabol I, Planinić P, Grubišić G, Fistonić I, Ćorušić A, Grce M. Methylated Host Cell Gene Promoters and Human Papillomavirus Type 16 and 18 Predicting Cervical Lesions and Cancer. PLoS One. 2015;10:e0129452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
65.  Feng Q, Hawes SE, Stern JE, Dem A, Sow PS, Dembele B, Toure P, Sova P, Laird PW, Kiviat NB. Promoter hypermethylation of tumor suppressor genes in urine from patients with cervical neoplasia. Cancer Epidemiol Biomarkers Prev. 2007;16:1178-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]