Abstract
Background: Human papillomavirus (HPV) infection is considered as the most common viral sexually transmitted infection worldwide. This poses an increasingly interdisciplinary medical challenge. Since there is vast scattered information in databases about HPV and the correlated diseases, we decided to collect useful data so that the experts can get a more comprehensive view of HPV.
Methods: In this article, HPV-associated diseases, prevalence, prevention, and new treatments are discussed. The retrieved articles reporting the latest data about the required information for our review were selected through searching in Web of Science, Scopus, Medline (PubMed), EMBASE, Cochrane Library, Ovid, and CINHAL with language limitations of English and German.
Results: There are 2 groups of HPVs: (1) low-risk HPV types that can lead to genital warts, and (2) high-risk HPV types that are involved in HPV-associated oncogenesis. About 70% of all sexually active women are infected and most of these infections heal within many weeks or months. In the case of HPV-persistence, a risk of preneoplasia or carcinoma exists. These types of viruses are responsible for the existence of genitoanal, gastrointestinal, urinary tract, and head and neck tumors. There is still no definite successful treatment. The detection of HPV-related condylomata occurs macroscopically in women and men, and the diagnosis of the precursors of cervical carcinoma in women is possible by Pap smear.
Conclusion: For extragenital manifestations, there is no structured early detection program. Meanwhile, studies on HPV vaccines confirm that they should be used for the primary prevention of HPV-dependent diseases. However, we need more research to find out the real advantages and disadvantages of vaccines.
Keywords: Human Papilloma Virus, Cancer, Epidemiology, Warts, Vaccines, Virology, Diagnostic
↑ What is “already known” in this topic:
There are numerous papers in the field of sexual transmitted diseases (STDs), especially HPV. However, most of them do not have comprehensive data involving the main facts. Some of them have only evaluated the epidemiology or carcinogenesis or screening methods. And some have only assessed one type of cancer correlated with HPV. Thus, there is no distinct information among concepts.
→ What this article adds:
This study presents comprehensive updated data about HPV, the laboratory methods, and all its correlated cancers. These combined data are highly useful for the clinicians and laboratory specialists. This is the main reason that we decided not to divide this article into several papers. The readers do not need to look for scattered information to find their required data. A vast part of the information is available in tables and figures that are easily reachable by the readers.
Introduction
Sexually transmitted infections (STIs) are initially developed by sexual contact and have a high rate of morbidity and mortality worldwide, affecting 50% to 70% of sexually active individuals (1, 2). Human papillomaviruses (HPVs) are a large and diverse group of epitheliotropic double-stranded DNA viruses (3). There are up to 225 types of HPVs divi ded into 5 groups (α, ß, γ, µ, and ν) (4). The exact classification of each group is shown in Figure 1. A subgroup of about 15 of the α-types (high-risk (HR)-HPV types) can lead to invasive carcinomas (5, 6).
Fig. 1.
HPV classification: Red color: High-risk HPVs, Green color: Low-risk HPVs, Yellow colors: probably high-risk HPVs, Blue color: Unknown
Persistent HPV infection is one of the important sexual transmitted diseases (STDs) associated with more than 5% of all cancers in the world (7). In other words, globally more than half of all malignancies related to infection are caused by HPV (8). Approximately 90% of the HPV viruses clear or become dormant in 1 to 2 years after infection. Statistics show that the majority of women who had a positive test for a high-risk HPV serotype developed cervical cancer after 3 to 5 years (9). HPVs mostly cause nonpersistent acute infections. Hence, like other oncoviruses, there is a striking gap between the times of diagnosis of the chronic infection and its early stages (10). Some studies report that infection with at least 1 high-risk HPV will occur during the lifetime of 60% of sexually active individuals (11). They are often eliminated by the immune system within 1 or 2 years after exposure (12). The virus in the remaining cases that persists for a long time affords lesions that can bring malignancies (13). Therefore, early diagnosis of HPV infection and HPV induced lesions is highly important to prevent cancer development (14). In this review, we tried to collect all data about the prevalence, mechanism of action and carcinogenesis, the correlated disases as well as a brief overview about laboratory studies, screening, diagnosis, and therapies of each disease. This collection has been prepared by the newest data of valid databases and websites, such as Centers for Disease Control and Prevention (CDC) and human papillomavirus (HPV) centers. This information can help us to be aware of several factors when a patient with HPV comes to us for screening or treatment.
Methods
Search Syntax and Search Strategy
All original published articles were searched in international databases, including Web of Science, Scopus, Medline (PubMed), EMBASE, Cochrane Library, Ovid, and CINHAL to retrieve all articles reporting the latest data about epidemiology, diagnostic methods, and treatment of diseases related to HPV that were not discussed comprehensively in previous studies. A search of these databases was performed by the researchers with hand searching through the reference lists and gray literature. We searched from January 1985 to March 2020, with language limitation of English and German. All related components were related to the following keywords: “Human Papilloma Virus”, “HPV”, “Epodemiology”, “Carcinogenesis”, “Diagnostic methods”, “Screening methods”, “Treatment”, “Cervical cancer”, “Cervical neoplasm”, “Cervical malignancy”, “Vulvar and vaginal cancer”, “Vulvar and vaginal neoplasm”, “Vulvar and vaginal malignancy”, “Anal cancer”, “Anal neoplasm”, “Anal malignancy”,”Esophageal cancer”, “Esophageal neoplasm”, “Esophageal malignancy”, “Colorectal cancer”, “Colorectal neoplasm”, “Colorectal maliganany”, “Prostate cancer”, “Prostate neoplasm”, “Prostate malignancy”, “Urothelial cancer”, “Urothelial eoplasm”, “Urothelial malignancy”, “Testicular cancer”, “Testicular neoplasm”, “Testicular malignancy”, “Renal cancer”, “Renal neoplasm”, “Renal malignany”, “Penile cancer”, “Penile neoplasm”, “Penile malignancy”, “Head and neck squamous cell carcinoma”, “Head and neck cancer”, “Head and neck neoplasm”, “Head and neck malignancy”, “cutaneous squamous cell carcinoma”, “Warts”, “HPV Vaccines”, and “HPV DNA Tests.
Screening and Selection
TheReference Manager bibliographic software was applied to manage the searched citations. Duplicate entries were checked by considering the title of the published papers, authors, year of publication, and specifications of the sources types. In case of questionable records, the texts were compared. We reviewed the initial search results, and after reviewing each article by title and available abstract, some of the articles were excluded. Evaluating the papers under consideration was based on the inclusion and exclusion criteria by 2 researchers separately (Sh.N. and Maryam S.). About 546 papers were retrieved; 353 of which were selected and evaluated to extract the required information. In case of finding no data in the studies available in databases, we tried to apply valid websites such as HPV center or CDC that have been cited in references.
Eligibility Criteria
All observational studies that assessed the prevalence of HPV and related cancers, screening and diagnostic methods, and total therapeutic managements were included. We excluded duplicate citations, oral presentation, posters, and articles where their abstract and full-text were not available. The original articles and reviews were included.
Data Extraction
Three steps of assessment for titles, abstracts, and full-texts were done. The full-text of each selected article was retrieved for detailed evaluation. Data were extracted using a checklist involving publication year, authors, type of cancer, laboratory tests, screening methods, diagnostic methods, and treatment. All steps from search to final data extraction were followed independently by 2 research experts (Sh. N., Marzieh S).
Results
Epidemiology and Transmission
About 50% to 80% of sexually active females will be infected with HPV during their lifetime (15). The estimated global HPV prevalence is 11.7%. South Africa (17.4.0%), Eastern Africa (33.6%), Eastern Europe (21.4%), Western Europe (9.0%), Eastern Europe (21.4%), and Caribbean (35.4%) showed the highest HPV prevalences (16, 17). Female sex workers (FSWs) are among the most susceptible groups to acquire HPV infection and develop cervical intraepithelial neoplasia and cervical cancer. In a meta-analysis done by Farhmand et al, it is demonstrated that the pooled HPV prevalence was 42.6%. HPV-16, HPV-52, and HPV-53 were the most common high-risk HPV types found among FSWs (18). Since there are no definite antiviral treatments for HPV, the high prevalence of genital HPV has been a great concern in the world (19). Another research evaluating 645 sexually active innercity young females reported a cervical HPV prevalence of 54% (20). Assessments of 12.7 million cancers occurring globally in 2008 showed that HPV infection is related to almost 100% of cervical cancers, 90% to 93% of anal canal cancers, 12% to 63% of oropharyngeal cancers, 40% to 64% of vaginal cancers, 40% to 51% of vulvar cancers, and 36% to 40% of penile cancers. Virtually 5% (610 000) of the cases had HPV associated with anogenital or oral cancers. Nonmelanoma skin cancers are the increased risk of cutaneous HPV types. The initial risk factor for occurring the anogenital and oral HPV infection among males and females is sexual behavior (21).
HPV Infection and Carcinogenesis
HPV infection can be persistent and carcinogenic through integrating the viral DNA into the host genome. Then, it omits the early and late HPV genes known as E2, E4, E5, L1, and L2. Two early viral proteins have oncogenic potentials: E6 and E7 (22, 23). The high-risk HPVs (HR-HPVs) that are also carcinogens have a great ability to remain in human keratinocytes in vitro through a chronic status. These viruses penetrate the cervical epithelial cells and express the oncogenic protein E6 and E7, leading to the inactivation of host regulatory proteins p53 and then retinoblastoma protein (RB). The viral antigen is a very specific type of antigen marking tumor cells. It is presented in viral-mediated carcinogenesis. Thus, the point that should be considered is that the best target to have an antitumor therapeutic goal is this antigen that is expressed just by the infected cells. This is somehow a way to block the autoimmunity cascade. Viral antigens can also act along with tumor-associated antigens in case of the dependence of oncogenes on expressing viral oncoproteins in infected cells. Scientists try to develop anticancer gene therapy because of the relationship between HPV infection and cervical cancer (24). When HPV starts to integrate into the host genome, the malignant transformation occurs. Hereby, E6 and E7 are expressed. Therefore, they have been targeted by several types of vaccines. It is proven that vaccines have a satisfactory result against these antigens in HPV-induced cervical dysplasia (25-27). They are applied in the clinical trial phase to treat cervical and head and neck cancers (28). The integration of the genome to the cell is necessary to induce cervical cancer. So persistent HPV infection is not the only or sufficient factor to this point (29). HPV 18 has a great power to integrate into the host genome leading to malignancy (30-32). The fragile sites of chromosome have this integration that causes disruption to the open-reading frame of E2 and less commonly E1, E4 or E5 (29, 30). Tables 1 and 2 present complete data of the statistics as well as present screening methods, diagnostic methods, and available treatments of different types of HPV-related diseases.
Table 1. The Statistics of Community, Prevalence, Incidence, Age, Types of Dependent and Mortality of HPV-related Cancers and Warts .
| Type of disease | Prevalence |
Incidence (per year) |
In which age it mostly occurs? | Related types | Mortality |
| Cervical cancer |
The 3rd most common cancer in women (156) From 31.6% in eastern Africa to a low prevalence of 6.2% in southeastern Asia (17) |
569,847 in 2018 (156) | 15 to 44 year (156) | 16, 18 (157) | 311,365 deaths in 2018 (156) |
|
Vulvar and vaginal cancer |
Rare (158, 159) 5.6%, 4.4% (160) |
27000 and 13000 new cases in 2008 respectively (156) |
Females aged 45 to 49 years old (161) |
16, 18 (162) |
880 and 400 deaths in 2007 respectively (162) |
|
Anal cancer |
Rare (156) 1.5 % of GI cancers (163) and more than 10% in HPV positive patients (164) |
27000 new cases (165) |
65-69 years old (166) |
16,18 (167) |
1- and 5-year mortality rates of 40% and 80%, respectively (163) |
|
Esophageal cancer |
The 8th most common cancer (168) 11.7%-38.9% (169, 170) |
500000 new cases (168) |
Mean age 65 years old (171) |
16, 57 (172) |
406000 deaths each year (168) |
|
Colorectal cancer |
The third most common in men and 2d most common in women (173) For prevalence rate Data not found |
141,270 new cases in 2016 (166) |
65-79 years old (174) |
16 (175, 176) |
52,286 deaths each year (166) |
|
Prostate cancer |
The most common cancer in men (177) 15% (178) |
160000 new cases (177) |
72-74 years old (179) |
16 (180) |
11,710 each year in the US between 1988–92 (181) |
|
Urothelial cancer |
The 4th most common cancer (182) 3%-50% (183-185) |
150,350 new cases (182) |
≥65 years old (186) |
16, 18 (187) |
132,432 worldwide in 2000 (186) |
|
Testicular cancer |
1% of all male cancers (188) 0.066% (166) |
14.9 per 100,000 (166) |
20-39 (166) |
16 (189-191) |
2% (188) |
|
Renal cancer |
The 6th most common cancer in men and the 10th most common cancer in women (160) 0.3% (166) |
22.8 per 100,000 men and 11.6 per 100,000 women each year (166) |
65-84 years old (166) |
Data not found |
About 9,000 men and 5,000 women each year (166) |
|
Penile cancer |
Rare (156) 6% (192) |
22000 new cases (156) |
50-70 years old (156) |
16, 18, 31, 33 (193) |
30% (194) |
|
Head and neck squamous cell carcinoma |
Less than 5% of all cancers (195) 6.9% (196) |
644,000 new cases each year (195) |
≤45 years old (195) |
16 (197) |
2.2% (195) |
|
Cutaneous squamous cell carcinoma |
The 2d most common form of skin cancers (198) 9-14% in men and 4-9% in women (195) |
700,000 new cases each year in US (199) |
In ages>35 years old (200) |
ß-HPV such as 5 and 8 (200, 201) |
2.1% (198) |
|
Warts |
One of the common sexually transmitted Peak prevalence of 53.8% (202) |
14,100,000 new cases each year (166) |
Peak prevalence 20- to 24 (202) |
6, 11, 42, 43, 44 (114) |
Significant source of morbidity and mortality worldwide (203). (Data about the exact rate was not found) |
Table 2. Pathogenesis, screening methods, diagnostic methods, treatment, and some useful information in brief. Abbreviations: ESCC, esophageal squamous cell cancer; EAC, esophageal adenocarcinoma; GEJAC, gastroesophageal junction adenocarcinoma; BCG: Bacillus Calmette-Guerin; CT scan, computed tomography scan; MRI, magnetic resonance imaging.
| Type of Disease | Pathogenesis | Screening & Diagnostic Methods | Treatment | Additional Information |
| Cervix | Cervical intraepithelial neoplasia exists due to chronic infection that can lead to cervical cancer (204) |
Pap smear (149) Colposcopy, cervical biopsy (149), avoid unnecessary excisions (205) |
Ursodeoxycholic acid (UDCA), chenodeoxycholic acid (CDCA), synthetic CDCA derivatives like HS-1199 and HS-1200 and the system of the cholic acid-functionalized star-shaped PLGA-b- TPGS (CA-PLGA-b-TPGS), polymeric nanoparticles control delivery of the drug, such as Docetaxel (149) | Progression of precursor lesions for cervical cancer takes more than 10 years (160, 206, 207) |
| Vulvar and vaginal | Vulvar and vaginal intraepithelial neoplasia (VIN and VaIN) (208, 209) |
No screening methods are available (4) Direct visual examination, biopsy and histopathological examination (210) |
Surgery, external or internal radiation therapy, and systemic or regional chemotherapy (211) | The current treatment strategies, unfortunately, are not successful. The relapse rate is high (158) |
| Anal | Strongly links to a complex inflammatory process leading to anal cancers of squamous cell origin (167) |
Anal Papanicolaou smears (pap) and Southern blotting (212) High-resolution anoscopy (HRA) (212) |
An organized team to plan chemotherapy, radiation therapy and surgery (5) | It has a ratio of female to male being as high as 5:1 (165). HPV was positive in 83%-95% of patients (213). HPV is the reason for 90% of SCCA (214, 215) |
| Esophageal cancer | Chronic infection leads to ESCC (216) |
In high incidence area regular endoscopy (171) Endoscopy and biopsy (217) |
Chemotherapy, radiation therapy and surgery (217) | A significant relation between HPV and ESCC but not with EAC and GEJAC. Identification of HPV in this malignancy can be helpful for better response and outcome (216) |
| Colorectal cancer | Chronic infection leads to mutations in glandular cells of the colorectal mucosa of colon and rectum (218) |
Fecal occult blood test and colonoscopy (219) Colonoscopy and biopsy (220) |
Chemotherapy, radiation therapy and surgery (220) | Identification of HPV in this malignancy can be helpful for better response and outcome (216) |
| Prostate cancer | Still unclear (150) |
Prostate-specific antigen (PSA) (179) Tissue biopsy (217) |
Chemotherapy, radiotherapy and surgery and androgen deprivation therapy (217) | It is important to keep HPV infection in mind when encounters with unusual disease manifestations of the urogenital tract (221) |
| Urothelial cancer | Still unclear (222) |
No recommended screening method (223) Applying CT urography or MRI for upper urinary tract evaluation and cystoscopy for lower urinary tract evaluation (224) |
Intravesical chemotherapy or intravesical BCG (224) | Urothelial cancer is an overarching term that describes a number of tumors that arise from the urothelial lining of the bladder, renal pelvis, ureters, and urethra (225) |
| Testicular cancer | It leads not directly to testicular cancer but can provide a status of higher vulnerability induced by the tumor (226) |
No recommended screening method (227) Transscrotal ultrasonography (228) |
Orchiectomy, chemotherapy and radiotherapy (229) | No Additional information |
| Renal cancer | Still unclear (230-232) |
No recommended screening method (182) Renal CT scan with and without contrast or MRI with and without gadolinium enhancement (233) |
Surgery, chemotherapy, immunotherapy, or targeted therapy (182) | No Additional information |
| Penile cancer | Penile intraepithelial neoplasia (PIN) (234) |
Data not found for screening methods No imagining technique can exactly detect micrometastatic lymph nodes. So, invasive inguinal lymph node diagnosis is recommended for all tumor stages from pT1G2 (235) |
Surgical methods (circumcision, wide local excision, and glans resurfacing), T-cell immune checkpoint inhibitors, and HPV genome targeting strategies (236, 237) | Penile SCC has four subgroups: warty, basaloid, keratinizing, and verrucous. Only the first two groups (warty and basaloid) are related to HPV (238, 239) |
| Head and neck squamous cell carcinoma | Still unclear (240-242) |
P16 IHC, FISH, HPV genome detection in biopsy specimens (243) Endoscopy (nasopharyngolaryngoscopy, esophagoscopy, and bronchoscopy, as appropriate) and biopsies (195) |
EGFR TKI and low-dose radiation, trans-oral robotic surgery, many reduction surgeries and post-operative adjuvant therapies based on pathologic staging (244) | HPV-positive OPSCCs have a better therapeutic response and prognosis (240-242) |
| Cutaneous squamous cell carcinoma (CSCC) | CSCC is mostly seen in patients with epidermodysplasia verruciformis (EV). EV is an autosomal recessive genodermatosis existed through mutations in EVER 1 and EVER 2 genes (200, 201) |
A total-body examination of the skin is the only screening test available (195) Biopsy (195) |
Surgical excision, cryotherapy, curettage, electrodesiccation, topical treatments (e.g imiquimod, 5-FU, ingenol mebutate, diclofenac, and retinoids) and radiation therapy (245) | Although most of CSCC have an excellent prognosis, some of them are susceptible to have poor outcomes (198) |
| Warts (Condyloma acuminate) | Available in the text. |
Regular physical examination, cytology/viral detection (246) Histologic examination of biopsy specimens (118) |
Topical and systemic. Trichloroacetic acid (TCA) is the best (124, 125) | Available In the text. |
Molecular Diagnosis of HPV
The molecular techniques are the main instrument to detect HPV DNA because of hard cultivation of HPV in culture systems. Although the E1 gene is used, the L1 and L2 late genes encode viral capsid proteins are used for HPV genotypes detection. The diagnostic techniques for detection and genotyping of HPV were classified as shown in Figure 2. All the commercially available tests are listed in Table 3.
Fig. 2.
The schematic classification of diagnostic techniques for detection and genotyping of HPV
Table 3. The classification of commercially available diagnostic techniques for the detection and genotyping of HPV .
|
The classification of diagnostic techniques for detection and genotyping of HPV |
The commercial available | ||
|
detection based methods nucleotide acid |
Nucleic acid amplification |
Conventional PCR based methods |
❖ PCR, Nested-PCR, multiplex-PCR, PCR-RFLP |
|
PCR following by hybridization |
❖ The LINEAR ARRAY HPV Genotyping Test (Roche), ❖ AMPLICOR HPV test (Roche), ❖ INNO-LiPA HPV Genotyping Extra (Innogenetics) ❖ Microplate colorimetric hybridization assay (MCHA) |
||
|
PCR-based fluorescent bead array |
❖ Microarray-based HPV genotyping assays ○ The PapilloCheck HPV Screening Test(greiner bio), ○ Clart HPV 2 – papillomavirus clinical arrays( Genomica), ○ HPV GenoArray Test Kit ( Hybribio), ○ GeneTrack HPV DNA Chip( Daejeon), ○ GeneSQUARE HPV Microarray,( Kurabo) ○ INFINITI HPV-HR QUAD (AutoGenomics), ○ PANArray HPV Genotyping Chip( Panagene), ○ GG HPVCHIP ( Goodgene) ❖ Suspension array based HPV genotyping assays ○ The Multiplex HPV Genotyping Kit (Multimetrix, Heidelberg) ○ digene HPV Genotyping LQ Test(Qiagen) |
||
|
Real-time PCR based HPV genotyping |
❖ Human papillomavirus Real-Time PCR Kit ( GenoID), ❖ Abbott RealTime High-Risk HPV(Abbott molecular), ❖ QIAscreen HPV PCR(QIAGEN) ❖ TRUPCR® HPV High Risk Kit (TRUPCR Diagnostics) ❖ cobas 4800 HPV test (Roche) |
||
|
HPV E6/E7 mRNA-based screening assays |
❖ PreTect HPV-Proofer (NorChip), ❖ NucliSENS EasyQ HPV V1 assay (bioMérieux) ❖ APTIMA HPV Assay (Gen-Probe) |
||
|
Signal amplification |
Signal amplification technologies include branched DNA (bDNA) |
❖ Branched DNA (bDNA), initially developed by Chiron and marketed by Bayer Diagnostics and recently by The Quantivirus(®) HPV E6/E7 RNA 3.0 assay |
|
|
Hybrid capture (HC) assays |
❖ hybrid capture (HC) assays ( Digene Corporation, QIAGEN) ❖ Cervista HPV test ( Invader) |
||
|
Noucleotid hybridization |
Southern blot hybridization |
❖ Just use in research laboratory |
|
|
in situ hybridization (ISH) |
❖ No commercial product were found and Just use in research laboratory |
||
|
Immune- biochemical based methods |
HPV Serology-ELISA assay |
Direct, indirect and competitive |
❖ Biocompare, Cusabio, Abbexa, Creative-diagnostics, |
|
HPV Neutralization assay |
high-throughput pseudovirion-based neutralization assay (HT-PBNA) |
❖ No commercial product were found |
|
1. Nucleic Acid Detection-based Methods
1.1. Nucleic Acid Amplification-based Methods
These methods are generally based on the polymerase chain reaction that is used for amplifying, detecting, and typing the HPV DNA by the use of degenerate primers MY09/MY11 or PGMY09/11, GP5+/6+ and SPF10 to amplify the viral capsid L1 gene (33-35).
1. The Conventional PCR-based Methods: The conventional PCR based methods are single or double nested-PCR (36, 37) multiplex-PCR (38) and nested-PCR-RFLP assay (34). Then, the PCR product is amplified by targeting a type-specific DNA sequence or treatment with restriction enzyme to determine the specific sequence existing in the sample related to a specific type of HPV (39, 40)
2. The PCR Following by Hybridization-based Methods: The methods-based amplification of target DNA following hybridization include traditional PCR in situ hybridization (PISH), microplate colorimetric hybridization assay (MCHA), the linear array for HPV genotyping, and the reverse line hybridization. The PISH technique is the typical PCR performed on the slide of intact paraffin-embedded tissue, and then hybridized with specific DNA probes (41, 42). The MCHA is a method based on PCR followed by colorimetric hybridization to type-specific probes on microplates (43). In the reverse line blot assays and the linear array HPV genotyping, after the amplifying step, the sequences are fixed on a membrane strip and detected by type-specific probe (39, 44).
3. PCR-based Fluorescent-based Array: Microarray-based HPV genotyping assays employ the PCR to amplificate the viral genome fragment and then hybridize with several HPV-specific oligonucleotide probes attached on the surface of an insoluble supporter like bead or DNA chip (45-53). The suspension array genotyping assays use bead-based technology, which is based on the use of polystyrene beads dyed with 2 spectrally distinct fluorophores (red and infrared), and each bead set is coupled with a specific oligonucleotide probe for 1 HPV type. The HPV sequences are amplified, denatured, and hybridized with the bead-bound probes. Then, hybridized biotinylated amplicons that are labeled by using phycoerythrin and streptavidin are served as a reporter fluorophore. The bead sets are then read and analyzed on a Luminex analyzer (54-60).
4. The Real-time PCR-based HPV Genotyping-based Methods: The real-time PCR is reliable and sensitive, with high accuracy and validity in HPV-DNA detection and genotyping. Also, the viral load quantification and the capability of multisample qualification with different fluorochromes is the advantage. In this method the fluorescent probes in cooperation with PCR primers allow for quantification of the viral genome and are presented in a sample as the name “viral load”(61, 62) (63-65). The cobas 4800 HPV test uses multiplex real-time PCR and nucleic acid hybridization with 4 different fluorescent reporter probes that concurrently detects the L1 gene (64, 66).
5. HPV E6/E7 mRNA-based Screening Assays: The detection of viral mRNA is based on transcription-mediated amplification of full-length E6/E7 transcripts accomplished by target capture. Reverse-transcriptase-PCR incorporates is a RT step following real-time quantitative PCR. The oncoproteins E6 and E7 are the most relevant transcripts for diagnostic and carcinogenesis follow-up. (67, 68). The main techniques used to detect mRNA for E6/E7 oncogenes are 3 commercial assays shown in Table 3 (45, 68-71).
1.2 Signal Amplification
Signal amplification describes methods-based probe molecule, which are hybridized to the target nucleic acid sequence and generate the signal related to amplification rate (72). Signal amplification technologies include branched DNA (bDNA) and hybrid capture (HC) assays (73, 74). The hybrid capture method is the most widely-used signal amplification method that briefly samples DNA hybridized with the cocktail of RNA probes. The RNA-DNA hybrids indicate the presence of HPV DNA and subsequently it is revealed by the nonradioactive signal-amplification method to aid detection (73-76). The Digene HCII technology served as the second version of hybrid capture (77).The bDNA assay directly measures nucleic acid molecules and is based on binding the subset of “target probes” bound to specific nucleotide sequences (exist in the sample) as in situ hybridization (bDNA ISH) (72, 74). The Cervista HPV test is a signal amplification method that uses 2 types of isothermal reactions, which is briefly based on the enzymatic cleaves the FRET oligonucleotides between the fluorophore and quencher molecule, resulting in the production of a fluorescence signal (39, 42, 74).
1.3 Noucleotid Hybridization-based Methods
The nonamplified HPV techniques include in southern/dot blot hybridization, and in situ hybridization (ISH). This method is generally time-consuming, requires more skill, necessary equipments, and is not as sensitive and reliable as the molecular methods. briefly, in HPV DNA detection by southern blot, the sample extracted DNA is digested by restriction enzymes and then runs in agarose gel electrophoresis to separate the digested DNA based on the size, and then is transferred to a nitrocellulose or nylon membrane and finally hybridized with cloned HPV genomic probes labeled with isotopic (P32) or nonisotopic (digoxigenin) techniques. The detection procedure in ISH occurs right on the fixed nuclei of infected cells (in situ) and hybridization reaction is evaluated microscopically (78, 79) In comparison with PCR, the blot hybridization-based method has higher specificity but is less sensitive (42, 80, 81).
2. Immune-biochemical-based Methods
1. HPV serology — ELISA assay: The serological tests to determine the HPV virus are performed based on VLPs (virus-like particles)(82-84), and the sensitivity is about 50%(85). Three forms of Elisa method include (1) Direct assays binding to HPV VLPs on the microplate; (2) indirect assays binding to HPV VLPs to the microplate via anti-VLP antibodies; and (3) Competitive assay by which the antigen is coated on fluorescent beads (Luminex-based assays) exposed to the sample (54, 58, 86).
2. HPV Neutralization Assay: High-throughput pseudovirion-based neutralization assay (HT-PBNA) with excellent repeatability and run-to-run reproducibility was developed for HPVs (86). HPV neutralization assays rely on neutralization of one of the following items: authentic virions, pseudotyped virions that are capsids carrying a reporter gene on their surface, and pseudovirions (PsVs) that have encapsidated reporter genes to assess anti-VLP conformational antibodies neutralizing activity. The neutralization assays are the gold standard to assess the protective potential of antibodies induced by HPVs vaccines in experimental systems (87-89).
The Potential Biomarkers for HPV Detection
The biomarkers related to high-risk HPV infection can help to enhance the sensitivity of cervical cytology screening, reduce false-negative diagnoses, monitoring and prognosis of related diseases.
The potential biomarker in cervical cancer includes in p16INK4a, Ki-67 that are the target of E6 and E7 oncoprotein (90-93). There are many molecular targets of E6 and E7, such as Wnt/β-catenin/Notch (94, 95), PI3K/AKT/mTOR pathway (96), P53, and PRb(97, 98). The molecular targets of HPV E5 oncoprotein are the cell surface receptors like EGFR (99, 100), p21Wafl/Sdil/Cip(101), p27KIP1(102), COX-2, VEGF, and Cav-1(102). Besides, the host microRNAs have been affected by HPV proteins E5, E6, and E7. Increased expression of miR-16, miR-25, miR-92a, and miR-378, and decreased expression of miR-22, miR-15a, miR-15b, miR-21and miR29a miR-16, miR-27a, miR-29a, and miR-100 are attributed to viral oncoprotein E6 or E7 (103-108). The miRNA-944(109) and miRNA-155(110) overexpression as well as downregulation of miRNA-375 (111) can potentially be served as a biomarker for cervical cancer follow-up. Dysregulation of miR-375/AEG-1 Axis by HPV high-risk 16 and 18 E6/E7 promotes cellular proliferation, migration, and invasion in cervical cancer (112).
Warts
Condyloma acuminate (CA) or wart is one of the most common sexually transmitted diseases in the world (15). The incubation period is 3 weeks to 8 months, and the clinical manifestation takes 2 to 3 months (113). The viruses producing genital warts are usually the low-risk types of HPV: 6, 11, 42, 43, and 44. types 6 and 11 are detected in 90% of cases. A study in 2009 has reported that 31% of patients with CA have a high-risk HPV as coinfection (114). A cohort study in 2019 reprted that the high-risk HPV genotypes could also be associated with warts. Particularly, a large number of patients with CIN2-3 and milder lesions with CIN1 were carriers of a virus of a HPV-HR genotype (115). Typically, CA is mostly discrete, skin-colored, brown or whitish, pedunculated,or broad-based with a variation in size from 1 millimeter to several centimeters (116). They are usually found on the external genitalia, perineum, perianal area, and oral cavity (117). Warts are most often diagnosed through their clinical appearance. Laboratory tests for the presence of HPV are not recommended for the diagnosis of CA. Histologic examination of biopsy specimens can be performed to rule out intraepithelial or invasive squamous cell carcinomas SCCs, which can coexist with or appear similar to anogenital warts (118). Their feature in histologic evaluation involves epidermal hyperplasia, parakeratosis, koilocytosis, and papillomatosis (119). Unfortunately, there is not any definite antiviral therapy to treat anogenital warts. Recurrence rate is high (25%-65%), which is may be due to the widespread infection and subclinical lesions (120). Also, most of the treatments are time-consuming and uncomfortable (121). Since there are no specific therapy options, the therapist should consider many points for the treatment such as the number, size, morphology, anatomic location, patient preference and side effects (122). The available therapies are effective for 60% to 90% of nonimmunosuppressed patients (121, 123). About trichloroacetic acid (TCA), it is investigated that 71% to 79% of cases regressed from high grade squamous intraepithelial lesion (HSIL) to low-grade squamous intraepithelial lesion (LSIL) or complete resolution when using TCA (124). The current treatments used for warts are as below:
1. Topical, destructive, surgical: podophyllotoxin and podophyllin, imiquimod, sinecatechins, intralesional immunotherapy with skin test antigens, cidofovir, 5-fluorouracil (5-FU), TCA, cryotherapy, potassium hydroxide, surgical excision, laser therapy, and photodynamic therapy
2. Systemic: interferon, isotretinoin (125).
The Risk of Infertility
Infertility means the inability of a couple to have a gestational process after 1 year of unprotected sex. Approximately 10% to 20% of couples at the reproductive age are suffering from infertility. Also, 50% of the reasons for infertility among couples relate to male factors, such as sexual dysfunction, congenital dysplasia, endocrine disorders, varicocele, immune factors, and sexually transmitted infections (126-128). It is established that there is a correlation between cervical disease and pregnancy complications (129). Nonetheless, we do not have any definite evidence recognizing the influence of HPV infection on pregnancy, such as preeclampsia, preterm labor, or fetal growth restriction (130). In men, the story is as in women. The literature supporting a link between HPV and infertility is still controversial (131). Seminal HPV infection is common worldwide, which may be correlated with the risk of male infertility through affecting sperm abnormalities, such as low sperm count and motility. Some studies have reported that HPV16 and 52 in men and HPV 58 in women are the most common types of HPVs leading to infertility (24, 132).
Vaccination
There is no definite therapy for HPV infection. Sometimes it is eliminated on its own. Some scientists believe that it cannot be cleared completely and may only switch to undetectable levels (133). We have several types of therapeutic vaccines evaluated in preclinical and clinical trials: live vector, protein or peptide, nucleic acid, and cell-based vaccines (134). However, 3 HPV vaccines are currently available in the vaccination program: cervarix (bivalent vaccine for HPV 16, 18), gardasil (quadrivalent vaccine for HPV 6, 11, 16, 18), and nonavalent vaccine (for HPV 6, 11, 16, 18, 31, 33, 45, 52, 58). These vaccines can target between 2 and 7 oncogenic HPV serotypes (135). They promise, in the long-term (30-50 years), to reduce the incidence of disease associated with HPV vaccine types (136). Some studies have proved that HPV vaccination is a secure and efficient method to prevent cancer (137, 138). Steben et al in 2018 evaluated 10 years of clinical experience in Canada. The results indicated that the prevalence of HPV types 16, 18, 6, and 11 was lower in qHPV-vaccinated than unvaccinated individuals (1.5% vs 11%). The risk of anogenital warts incidence had a decrease by up to about 45% in vaccinated population in cohort studies and the incidence of cervical intraepithelial neoplastic had a significant decrease by up to 86%. These researchers concluded that the programs of HPV vaccination constitute an effective and useful public health initiative (139). Unfortunately, there is limited vaccination coverage of large populations in the world. Therefore, a gross unvaccinated population remains at a high risk of HPV-associated disease (14). Additionally, some researches have demonstrated that current vaccines will not be useful for preventing all types of HPV-related cancers (140). Also, the HPV subtype distribution in cervical cancer varies throughout the world (141, 142). Japanese scientists in 2019 reported a significant number of adolescent girls complaining of unusual symptoms after HPV vaccination. The vast majority of them had psychiatric illness in the absence of any pathologic findings in radiological images or laboratory tests. So the recommendation of vaccination was withdrawn by the Japanese Ministry of Public Health (143). At present, the vaccination program is implemented in Argentina, Australia, Austria, Belgium, Bhutan, Brazil, Brunei, Darussalam, Canada, Colombia, Cook Islands, Czech Republic (the), Denmark, Fiji, France, Germany, Greece, Iceland, Ireland, Israel, Italy, Japan, Kiribati, Latvia, Lesotho, Luxembourg, Malaysia, Marshall Islands (the), Mexico, Micronesia, Netherlands, New Zealand, Norway, Palau, Panama, Paraguay, Portugal, Rwanda, San Marino, Singapore, Slovenia, Spain, Sweden, Switzerland, United Kingdom of Great Britain, the former Yugoslav Republic of Macedonia, Uganda, and Northern Ireland, Trinidad and Tobago, United States of America (the), and Uruguay (144).
Discussion
HPV transmission risk is increased among multipartners. It can originate from a genital area and extend to other parts like the anus region. Neonates can be infected by their mothers during pregnancy or the delivery period through vertical and perinatal transmission (145). The transmitted couples are sexually more active with a history of not using condoms regularly. It shows that condoms can be effective, along with the prophylactic vaccines. Despite the 3% of transmitting couples, more than 50% of nontransmitting couples presented that during the previous 4 months they used condoms in 100% of intercourses (146). So condoms can be used to protect against HPV but since they do not cover every possible HPV-infected area of the body, they cannot offer full protection (147). According to a cohort study done by Hernandez et al, genital transmission from women to men occurs more frequently than from men to women. The primary source of transmission to the cervix is the penis. The cervix and urethra are the primary sources of infection to male genitals. Sexual transmission implicates also the scrotum, the anus of women, and the hands of both genders. In heterosexual transmission the anus of women plays an important role as a major source and target. It is found that HPV transmission does not definitely influence the target organs because it depends on the tissue or genotype differences or both (146).
Epidemiologist research finds that HR-HPVs play an important role in cervical cancer and bacterial vaginosis (148, 149). After the integration of the genome, E6 oncogene is expressed. Many cells have changed by the E6 protein, leading to inhibition of apoptosis and increased telomerase function. This is why E6 protein can prolong cellular lifespan. It functions as a transcriptional activator accompanied by this prolonged lifespan and transforms cells (150). HR-HPV E7 allows the cell to increase its transforming activities. Therefore, human keratinocytes are immortalized because they interact with factors that regulate cell growth (151). Based on the our results, the expression of E6 and E7 can be blocked by E2. After disrupting, it enables uninhibited E6 and E7 oncoprotein activity (31). The differences between low-risk and high-risk HPVs in producing warts or cancers is mainly in E6 and E7 function. In low-risk HPVs, E6/E7 expression stimulates cell cycle entry in the upper epithelial layers (differentiating cells), allowing genome amplification in both low and high-risk HPVs. Also, basal cell proliferation may still be stimulated by growth factors, but not E6/E7. There is little or no expression of E6/E7. About high-risk HPVs, E6/E7 expression stimulates additional cell cycle entry and cell proliferation in the basal and parabasal epithelial layers leading to neoplasia. E6/E7 inhibit immune response to tolerate viral gene expression (152). HPV vaccines provide a promising primary approach to prevent malignancies. Because HPV acquisition generally occurs soon after first sexual activity, vaccine effectiveness will be lower in older age groups because of prior infections. Evidence suggests that although HPV vaccination is safe for adults aged 27 to 45 years, population benefit would be minimal; nevertheless, some adults who are not adequately vaccinated might be at risk for new HPV infection and might benefit from vaccination in this age range. Vaccination is routinely recommended at age 11 or 12 years; it can be given starting at age 9 years. Although catch-up HPV vaccination is recommended for all persons through age 26 years who are not adequately vaccinated, it is not recommended for all adults aged >26 years. Instead, shared clinical decision-making regarding HPV vaccination is recommended for some adults aged 27 to 45 years who are not adequately vaccinated. HPV vaccines are not licensed to be used in adults aged >45 years (153).
Strengths
This was a comprehensive review study involving 353 valid publications evaluating all important aspects of HPVs and their related cancers. Most of the studies have evaluated only 1 or 2 types of cancers correlated with HPVs. The need for easily accessible comprehensive data in this field seemed to be necessary. Thus, we tried to provide such data for clinicians and laboratory specialists working on HPV infection.
Limitations
The major limitations of this review were lack of data in some countries and nonvalid data in developing countries. Some papers had poor or unclear information or evaluation. In some cases, it was not possible to find valuable data. Thus, we applied the phrase “not found” for such cases.
Conclusion
About 50% to 80% of sexually active females will be infected with HPV during their lifetime. The global HPV prevalence has been estimated to beabout 11.7%. The major burden of HPV infection is the carcinogenic effect of high-risk HPVs. Since there is no definite treatment for HPV, the high prevalence of genital HPV has been a great concern in the world. At present, vaccination has been introduced as the best prevention and treatment method for HPV infection. If complete effectiveness of vaccination is expected, future vaccines should be multivalent for all described oncogenic HPV types. Nonetheless, these vaccines will be much more expensive than current formulations (14). Although some researchers have reported positive clinical effectiveness of vaccines in reducing malignancy, large population-based clinical studies of these vaccines are necessary to assess the true impact of vaccination (135, 154, 155). We believe that routine use of HPV vaccines needs much more care and assessment because there are many doubts and questions about these vaccines.
Conflict of Interests
The authors declare that they have no competing interests.
Cite this article as: Soheili M, Keyvani H, Soheili M, Nasseri Sh. Human papilloma virus: A review study of epidemiology, carcinogenesis, diagnostic methods, and treatment of all HPV-related cancers. Med J Islam Repub Iran. 2021 (22 May);35:65. https://doi.org/10.47176/mjiri.35.65
Footnotes
Conflicts of Interest: None declared
Funding:None
References
- 1.Trottier H, Franco EL. The epidemiology of genital human papillomavirus infection. Vaccine. 2006 Mar 30;24:S4–15. doi: 10.1016/j.vaccine.2005.09.054. [DOI] [PubMed] [Google Scholar]
- 2.Yarbrough ML, Burnham CA. The ABCs of STIs: an update on sexually transmitted infections. Clin Chem. 2016;62(6):811–23. doi: 10.1373/clinchem.2015.240234. [DOI] [PubMed] [Google Scholar]
- 3.Egawa N, Doorbar J. The low-risk papillomaviruses. Virus Res. 2017;231:119–27. doi: 10.1016/j.virusres.2016.12.017. [DOI] [PubMed] [Google Scholar]
- 4.Haley CT, Mui UN, Vangipuram R, Rady PL, Tyring SK. Human oncoviruses: Mucocutaneous manifestations, pathogenesis, therapeutics, and prevention: Papillomaviruses and Merkel cell polyomavirus. J Am Acad Dermatol. 2019;81(1):1–21. doi: 10.1016/j.jaad.2018.09.062. [DOI] [PubMed] [Google Scholar]
- 5.Krzowska-Firych J, Lucas G, Lucas C, Lucas N, Pietrzyk Ł. An overview of Human Papillomavirus (HPV) as an etiological factor of the anal cancer. J Infect Public Health. 2019 Jan-Feb;12(1):1–6. doi: 10.1016/j.jiph.2018.06.005. [DOI] [PubMed] [Google Scholar]
- 6.von Knebel Doeberitz, Prigge ES. Role of DNA methylation in HPV associated lesions. Papillomavirus Res . 2019 Jun;7:180–183. doi: 10.1016/j.pvr.2019.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D, Plummer M. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012 Jun;13(6):607–15. doi: 10.1016/S1470-2045(12)70137-7. [DOI] [PubMed] [Google Scholar]
- 8.Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health. 2016 Sep;4(9):e609–16. doi: 10.1016/S2214-109X(16)30143-7. [DOI] [PubMed] [Google Scholar]
- 9.de Sanjose S, Quint WG, Alemany L, Geraets DT, Klaustermeier JE, Lloveras B. et al. Human papillomavirus genotype attribution in invasive cervical cancer: a retrospective cross-sectional worldwide study. Lancet Oncol. 2010 Nov;11(11):1048–56. doi: 10.1016/S1470-2045(10)70230-8. [DOI] [PubMed] [Google Scholar]
- 10.Alizon S, Murall CL, Bravo IG. Why Human Papillomavirus Acute Infections Matter. Viruses. 2017 Oct 10;9(10):293. doi: 10.3390/v9100293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schiffman M, Castle PE, Jeronimo J, Rodriguez AC, Wacholder S. Human papillomavirus and cervical cancer. Lancet. 2007 Sep 8;370(9590):890–907. doi: 10.1016/S0140-6736(07)61416-0. [DOI] [PubMed] [Google Scholar]
- 12.Woodman CB, Collins SI, Young LS. The natural history of cervical HPV infection: unresolved issues. Nat Rev Cancer. 2007 Jan;7(1):11–22. doi: 10.1038/nrc2050. [DOI] [PubMed] [Google Scholar]
- 13.Trimble CL, Piantadosi S, Gravitt P, Ronnett B, Pizer E, Elko A, Wilgus B. et al. Spontaneous regression of high-grade cervical dysplasia: effects of human papillomavirus type and HLA phenotype. Clin Cancer Res. 2005 Jul 1;11(13):4717–23. doi: 10.1158/1078-0432.CCR-04-2599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Rosales R, Rosales C. Immune therapy for human papillomaviruses-related cancers. World J Clin Oncol. 2014 Dec 10;5(5):1002–19. doi: 10.5306/wjco.v5.i5.1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Koutsky L. Epidemiology of genital human papillomavirus infection. Am J Med. 1997;102(5):3–8. doi: 10.1016/s0002-9343(97)00177-0. [DOI] [PubMed] [Google Scholar]
- 16. Bruni L. The frequency of HPV infection worldwide. https://wwwhpvworldcom/articles/the-frequency-of-hpv-infection-worldwide/. 2020.
- 17.de Sanjosé S, Diaz M, Castellsagué X, Clifford G, Bruni L, Muñoz N, Bosch FX. Worldwide prevalence and genotype distribution of cervical human papillomavirus DNA in women with normal cytology: a meta-analysis. Lancet Infect Dis. 2007 Jul;7(7):453–9. doi: 10.1016/S1473-3099(07)70158-5. [DOI] [PubMed] [Google Scholar]
- 18.Farahmand M, Moghoofei M, Dorost A, Abbasi S, Monavari SH, Kiani SJ. et al. Prevalence and genotype distribution of genital human papillomavirus infection in female sex workers in the world: a systematic review and meta-analysis. BMC Public Health. 2020;20(1):1455. doi: 10.1186/s12889-020-09570-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Roden RB, Lowy DR, Schiller JT. Papillomavirus is resistant to desiccation. J Infect Dis. 1997 Oct;176(4):1076–9. doi: 10.1086/516515. [DOI] [PubMed] [Google Scholar]
- 20.Schlecht NF, Burk RD, Nucci-Sack A, Shankar V, Peake K, Lorde-Rollins E. et al. Cervical, anal and oral HPV in an adolescent inner-city health clinic providing free vaccinations. PLoS One. 2012;7(5):e37419. doi: 10.1371/journal.pone.0037419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Manini I, Montomoli E. Epidemiology and prevention of Human Papillomavirus. Ann Ig. 2018 Jul-Aug;30(4 Supple 1):28–32. doi: 10.7416/ai.2018.2231. [DOI] [PubMed] [Google Scholar]
- 22.Howie HL, Katzenellenbogen RA, Galloway DA. Papillomavirus E6 proteins. Virology. 2009;384(2):324–34. doi: 10.1016/j.virol.2008.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wise-Draper TM, Wells SI. Papillomavirus E6 and E7 proteins and their cellular targets. Front Biosci. 2008 Jan 1;13:1003–17. doi: 10.2741/2739. [DOI] [PubMed] [Google Scholar]
- 24.Lyu Z, Feng X, Li N, Zhao W, Wei L, Chen Y. et al. Human papillomavirus in semen and the risk for male infertility: a systematic review and meta-analysis. BMC Infect Dis. 2017 Nov 9;17(1):714. doi: 10.1186/s12879-017-2812-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kenter GG, Welters MJ, Valentijn ARP, Lowik MJ, Berends-van der Meer DM, Vloon AP. et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009 Nov 5;361(19):1838–47. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
- 26.de Vos van Steenwijk PJ, van Poelgeest MI, Ramwadhdoebe TH, Löwik MJ, Berends-van der Meer DM. et al. The long-term immune response after HPV16 peptide vaccination in women with low-grade pre-malignant disorders of the uterine cervix: a placebo-controlled phase II study. Cancer Immunol Immunother. 2014 Feb;63(2):147–60. doi: 10.1007/s00262-013-1499-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.van Poelgeest MI, Welters MJ, Vermeij R, Stynenbosch LF, Loof NM, Berends-van der Meer DM. et al. Vaccination against Oncoproteins of HPV16 for Noninvasive Vulvar/Vaginal Lesions: Lesion Clearance Is Related to the Strength of the T-Cell Response. Clin Cancer Res. 2016 May 15;22(10):2342–50. doi: 10.1158/1078-0432.CCR-15-2594. [DOI] [PubMed] [Google Scholar]
- 28.Wang C, Dickie J, Sutavani RV, Pointer C, Thomas GJ, Savelyeva N. Targeting Head and Neck Cancer by Vaccination. Front Immunol. 2018 Apr 23;9:830. doi: 10.3389/fimmu.2018.00830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Spurgeon ME, Lambert PF. Human Papillomavirus and the Stroma: Bidirectional Crosstalk during the Virus Life Cycle and Carcinogenesis. Viruses. 2017 Aug 9;9(8):219. doi: 10.3390/v9080219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Durzynska J, Lesniewicz K, Poreba E. Human papillomaviruses in epigenetic regulations. Mutat Res Rev Mutat Res . 2017 Apr-Jun;772:36–50. doi: 10.1016/j.mrrev.2016.09.006. [DOI] [PubMed] [Google Scholar]
- 31.McBride AA, Warburton A. The role of integration in oncogenic progression of HPV-associated cancers. PLoS Pathog. 2017 Apr 6;13(4):e1006211. doi: 10.1371/journal.ppat.1006211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. Human papillomavirus molecular biology and disease association. Rev Med Virol. 2015 Mar;25 Suppl 1(Suppl Suppl 1):2–23. doi: 10.1002/rmv.1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. World Health O. Human papillomavirus laboratory manual. 1st ed., 2009 ed. Geneva: World Health Organization; 2010.
- 34.Coser J, Boeira TdR, Fonseca ASK, Ikuta N, Lunge VR. Human papillomavirus detection and typing using a nested-PCR-RFLP assay. Braz J Infect Dis. 2011;15(5):467–72. doi: 10.1016/s1413-8670(11)70229-x. [DOI] [PubMed] [Google Scholar]
- 35.Carvalho Nde O, del Castillo DM, Perone C, Januário JN, Melo VH, Brasileiro Filho G. Comparison of HPV genotyping by type-specific PCR and sequencing. Mem Inst Oswaldo Cruz. 2010 Feb;105(1):73–8. doi: 10.1590/s0074-02762010000100011. [DOI] [PubMed] [Google Scholar]
- 36.Jalouli M, Jalouli J, Ibrahim SO, Hirsch JM, Sand L. Comparison between single PCR and nested PCR in detection of human papilloma viruses in paraffin-embedded OSCC and fresh oral mucosa. In vivo. 2015;29(1):65–70. [PubMed] [Google Scholar]
- 37.Tawe L, Grover S, Narasimhamurthy M, Moyo S, Gaseitsiwe S, Kasvosve I. et al. Molecular detection of human papillomavirus (HPV) in highly fragmented DNA from cervical cancer biopsies using double-nested PCR. MethodsX. 2018;5:569–78. doi: 10.1016/j.mex.2018.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Romero-Pastrana F. Detection and typing of human papilloma virus by multiplex PCR with type-specific primers. ISRN Microbiol. 2012;2012:186915. doi: 10.5402/2012/186915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kroupis C, Vourlidis N. Human papilloma virus (HPV) molecular diagnostics. Clin Chem Lab Med. 2011 Nov;49(11):1783–99. doi: 10.1515/CCLM.2011.685. [DOI] [PubMed] [Google Scholar]
- 40.Molijn A, Kleter B, Quint W, van Doorn LJ. Molecular diagnosis of human papillomavirus (HPV) infections. J Clin Virol. 2005 Mar;32 Suppl 1:S43–51. doi: 10.1016/j.jcv.2004.12.004. [DOI] [PubMed] [Google Scholar]
- 41.Nuovo GJ. In situ detection of human papillomavirus DNA after PCR-amplification. Methods Mol Biol. 2011;688:35–46. doi: 10.1007/978-1-60761-947-5_4. [DOI] [PubMed] [Google Scholar]
- 42.Venuti A, Paolini F. HPV detection methods in head and neck cancer. Head Neck Pathol. 2012 Jul;6 Suppl 1(Suppl 1):S63–74. doi: 10.1007/s12105-012-0372-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Barcellos RB, Almeida SE, Sperhacke RD, Verza M, Rosso F, Medeiros RM. et al. Evaluation of a novel microplate colorimetric hybridization genotyping assay for human papillomavirus. J Virol Methods. 2011 Oct;177(1):38–43. doi: 10.1016/j.jviromet.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 44.Chin-Hong PV, Klausner JD. New diagnostic tests for HPV in the developed and the developing world. MLO Med Lab Obs. 2008 Nov;40(11):48, 50, 52–3. [PubMed] [Google Scholar]
- 45.Poljak M, Kocjan BJ. Commercially available assays for multiplex detection of alpha human papillomaviruses. Expert Rev Anti Infect Ther. 2010 Oct;8(10):1139–62. doi: 10.1586/eri.10.104. [DOI] [PubMed] [Google Scholar]
- 46.García-Sierra N, Martró E, Castellà E, Llatjós M, Tarrats A, Bascuñana E. et al. Evaluation of an array-based method for human papillomavirus detection and genotyping in comparison with conventional methods used in cervical cancer screening. J Clin Microbiol. 2009 Jul;47(7):2165–9. doi: 10.1128/JCM.00402-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Sun ZR, Ji YH, Zhou WQ, Zhang SL, Jiang WG, Ruan Q. Characteristics of HPV prevalence among women in Liaoning province, China. Int J Gynaecol Obstet. 2010 May;109(2):105–9. doi: 10.1016/j.ijgo.2009.11.026. [DOI] [PubMed] [Google Scholar]
- 48.Lin M, Yang LY, Li LJ, Wu JR, Peng YP, Luo ZY. Genital human papillomavirus screening by gene chip in Chinese women of Guangdong province. Aust N Z J Obstet Gynaecol. 2008 Apr;48(2):189–94. doi: 10.1111/j.1479-828X.2008.00844.x. [DOI] [PubMed] [Google Scholar]
- 49.Kim KH, Yoon MS, Na YJ, Park CS, Oh MR, Moon WC. Development and evaluation of a highly sensitive human papillomavirus genotyping DNA chip. Gynecol Oncol. 2006 Jan;100(1):38–43. doi: 10.1016/j.ygyno.2005.08.024. [DOI] [PubMed] [Google Scholar]
- 50.Klug SJ, Molijn A, Schopp B, Holz B, Iftner A, Quint W. et al. Comparison of the performance of different HPV genotyping methods for detecting genital HPV types. J Med Virol. 2008 Jul;80(7):1264–74. doi: 10.1002/jmv.21191. [DOI] [PubMed] [Google Scholar]
- 51.Erali M, Pattison DC, Wittwer CT, Petti CA. Human papillomavirus genotyping using an automated film-based chip array. J Mol Diagn. 2009 Sep;11(5):439–45. doi: 10.2353/jmoldx.2009.080154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Oh TJ, Kim CJ, Woo SK, Kim TS, Jeong DJ, Kim MS. et al. Development and clinical evaluation of a highly sensitive DNA microarray for detection and genotyping of human papillomaviruses. J Clin Microbiol. 2004 Jul;42(7):3272–80. doi: 10.1128/JCM.42.7.3272-3280.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liu SS, Leung RC, Chan KK, Cheung AN, Ngan HY. Evaluation of a newly developed GenoArray human papillomavirus (HPV) genotyping assay and comparison with the Roche Linear Array HPV genotyping assay. J Clin Microbiol. 2010;48(3):758–64. doi: 10.1128/JCM.00989-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Oh Y, Bae SM, Kim YW, Choi HS, Nam GH, Han SJ. et al. Polymerase chain reaction-based fluorescent Luminex assay to detect the presence of human papillomavirus types. Cancer Sci. 2007 Apr;98(4):549–54. doi: 10.1111/j.1349-7006.2007.00427.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jiang HL, Zhu HH, Zhou LF, Chen F, Chen Z. Genotyping of human papillomavirus in cervical lesions by L1 consensus PCR and the Luminex xMAP system. J Med Microbiol. 2006 Jun;55(Pt 6):715–720. doi: 10.1099/jmm.0.46493-0. [DOI] [PubMed] [Google Scholar]
- 56.Schmitt M, Dondog B, Waterboer T, Pawlita M, Tommasino M, Gheit T. Abundance of multiple high-risk human papillomavirus (HPV) infections found in cervical cells analyzed by use of an ultrasensitive HPV genotyping assay. J Clin Microbiol. 2010;48(1):143–9. doi: 10.1128/JCM.00991-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Schmitt M, Dondog B, Waterboer T, Pawlita M. Homogeneous amplification of genital human alpha papillomaviruses by PCR using novel broad-spectrum GP5+ and GP6+ primers. J Clin Microbiol. 2008;46(3):1050–9. doi: 10.1128/JCM.02227-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Schmitt M, Bravo IG, Snijders PJ, Gissmann L, Pawlita M, Waterboer T. Bead-based multiplex genotyping of human papillomaviruses. J Clin Microbiol. 2006;44(2):504–12. doi: 10.1128/JCM.44.2.504-512.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Park KS, Kim JY, Ki CS, Lee NY. Comparison of the digene HPV genotyping LQ test and the PANArray HPV genotyping chip for detection of high-risk or probable high-risk human papillomavirus genotypes. Ann Lab Med. 2014 Jul;34(4):279–85. doi: 10.3343/alm.2014.34.4.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Geraets DT, Lenselink CH, Bekkers RL, van Doorn LJ, Quint WG, Melchers WJ. Universal human papillomavirus genotyping by the digene HPV Genotyping RH and LQ Tests. J Clin Virol. 2011 Apr;50(4):276–80. doi: 10.1016/j.jcv.2010.12.011. [DOI] [PubMed] [Google Scholar]
- 61.Roberts I, Ng G, Foster N, Stanley M, Herdman MT, Pett MR. et al. Critical evaluation of HPV16 gene copy number quantification by SYBR green PCR. BMC Biotechnol. 2008;8:57. doi: 10.1186/1472-6750-8-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mateos ML, Chacón de Antonio J, Rodríguez-Domínguez M, Sanz I, Rubio MD. Evaluación de un sistema de PCR a tiempo real (cobas 4800) para la detección separada de los genotipos 16 y 18 y otros genotipos de alto riesgo del virus del papiloma humano en la prevención del cáncer cervical [Evaluation of a prototype real-time PCR assay for the separate detection of human papilloma virus genotypes 16 and 18 and other high risk human papillomavirus in cervical cancer screening] Enferm Infecc Microbiol Clin. 2011 Jun-Jul;29(6):411–4. doi: 10.1016/j.eimc.2011.01.007. Spanish. [DOI] [PubMed] [Google Scholar]
- 63.Kocjan BJ, Seme K, Poljak M. Comparison of the Abbott RealTime High Risk HPV test and INNO-LiPA HPV Genotyping Extra test for the detection of human papillomaviruses in formalin-fixed, paraffin-embedded cervical cancer specimens. J Virol Methods. 2011 Jul;175(1):117–9. doi: 10.1016/j.jviromet.2011.04.006. [DOI] [PubMed] [Google Scholar]
- 64.Martínez SB, Palomares JC, Artura A, Parra M, Cabezas JL, Romo JM. et al. Comparison of the Cobas 4800 Human Papillomavirus test against a combination of the Amplicor Human Papillomavirus and the Linear Array tests for detection of HPV types 16 and 18 in cervical samples. J Virol Methods. 2012 Mar;180(1-2):7–10. doi: 10.1016/j.jviromet.2011.12.002. [DOI] [PubMed] [Google Scholar]
- 65.Poljak M, Ostrbenk A. The Abbott RealTime High Risk HPV test is a clinically validated human papillomavirus assay for triage in the referral population and use in primary cervical cancer screening in women 30 years and older: a review of validation studies. Acta Dermatovenerol Alpina, Pannonica, et Adriatica. 2013;22(2):43–7. [PubMed] [Google Scholar]
- 66.Heideman DA, Hesselink AT, Berkhof J, van Kemenade F, Melchers WJ, Daalmeijer NF. et al. Clinical validation of the cobas 4800 HPV test for cervical screening purposes. J Clin Microbiol. 2011;49(11):3983–5. doi: 10.1128/JCM.05552-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cuzick J, Mayrand MH, Ronco G, Snijders P, Wardle J. Chapter 10: New dimensions in cervical cancer screening. Vaccine. 2006;24 Suppl 3:S3/90–7. doi: 10.1016/j.vaccine.2006.05.122. [DOI] [PubMed] [Google Scholar]
- 68.Lie AK, Kristensen G. Human papillomavirus E6/E7 mRNA testing as a predictive marker for cervical carcinoma. Expert Rev Mol Diagn. 2008 Jul;8(4):405–15. doi: 10.1586/14737159.8.4.405. [DOI] [PubMed] [Google Scholar]
- 69.Villa LL, Denny L. CHAPTER 7 Methods for detection of HPV infection and its clinical utility. Int J Gynaecol Obstet. 2006 Nov;94 Suppl 1:S71–S80. doi: 10.1016/S0020-7292(07)60013-7. [DOI] [PubMed] [Google Scholar]
- 70.Hwang SJ, Shroyer KR. Biomarkers of cervical dysplasia and carcinoma. J Oncol. 2012;2012:507286. doi: 10.1155/2012/507286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dockter J, Schroder A, Eaton B, Wang A, Sikhamsay N, Morales L. et al. Analytical characterization of the APTIMA HPV Assay the official publication of the Pan American Society for Clinical Virology. J Clin Virol. 2009;45 Suppl 1:S39–47. doi: 10.1016/S1386-6532(09)70007-1. [DOI] [PubMed] [Google Scholar]
- 72.Player AN, Shen LP, Kenny D, Antao VP, Kolberg JA. Single-copy gene detection using branched DNA (bDNA) in situ hybridization. J Histochem Cytochem. 2001 May;49(5):603–12. doi: 10.1177/002215540104900507. [DOI] [PubMed] [Google Scholar]
- 73. Wang YF. Signal Amplification Techniques: bDNA, Hybrid Capture. In: Tang YW, Stratton CW, editors. Advanced Techniques in Diagnostic Microbiology. Boston, MA: Springer US; 2006. p. 228-42.
- 74.Shen Y, Gong J, He Y, Cheng G, Okunieff P, Li X. Quantivirus(R) HPV E6/E7 RNA 30 assay (bDNA) is as sensitive, but less specific than Hybrid Capture 2 test. J Virol Methods. 2013;187(2):288–93. doi: 10.1016/j.jviromet.2012.11.024. [DOI] [PubMed] [Google Scholar]
- 75.Lörincz AT. Hybrid Capture method for detection of human papillomavirus DNA in clinical specimens: a tool for clinical management of equivocal Pap smears and for population screening. J Obstet Gynaecol Res. 1996 Dec;22(6):629–36. doi: 10.1111/j.1447-0756.1996.tb01081.x. [DOI] [PubMed] [Google Scholar]
- 76.Hubbard RA. Human papillomavirus testing methods. Arch Pathol Lab Med. 2003 Aug;127(8):940–5. doi: 10.5858/2003-127-940-HPTM. [DOI] [PubMed] [Google Scholar]
- 77.Chaudhary AK, Pandya S, Mehrotra R, Bharti AC, Singh M, Singh M. Comparative study between the Hybrid Capture II test and PCR based assay for the detection of human papillomavirus DNA in oral submucous fibrosis and oral squamous cell carcinoma. Virol J. 2010 Sep 23;7:253. doi: 10.1186/1743-422X-7-253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Smeets SJ, Hesselink AT, Speel EJ, Haesevoets A, Snijders PJ, Pawlita M. et al. A novel algorithm for reliable detection of human papillomavirus in paraffin embedded head and neck cancer specimen. Int J Cancer. 2007 Dec 1;121(11):2465–72. doi: 10.1002/ijc.22980. [DOI] [PubMed] [Google Scholar]
- 79.Badaracco G, Venuti A, Morello R, Muller A, Marcante ML. Human papillomavirus in head and neck carcinomas: prevalence, physical status and relationship with clinical/pathological parameters. Anticancer Res. 2000 Mar-Apr;20(2B):1301–5. [PubMed] [Google Scholar]
- 80.Venuti A, Manni V, Morello R, De Marco F, Marzetti F, Marcante ML. Physical state and expression of human papillomavirus in laryngeal carcinoma and surrounding normal mucosa. J Med Virol. 2000 Apr;60(4):396–402. doi: 10.1002/(sici)1096-9071(200004)60:4<396::aid-jmv6>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
- 81.Yeudall WA, Campo MS. Human papillomavirus DNA in biopsies of oral tissues. J Gen Virol. 1991 Jan;72(Pt 1):173–6. doi: 10.1099/0022-1317-72-1-173. [DOI] [PubMed] [Google Scholar]
- 82.Mayrand MH, Coutlée F, Hankins C, Lapointe N, Forest P, de Ladurantaye M. et al. Detection of human papillomavirus type 16 DNA in consecutive genital samples does not always represent persistent infection as determined by molecular variant analysis. J Clin Microbiol. 2000 Sep;38(9):3388–93. doi: 10.1128/jcm.38.9.3388-3393.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.de Sanjosé S, Palefsky J. Cervical and anal HPV infections in HIV positive women and men. Virus Res. 2002 Nov;89(2):201–11. doi: 10.1016/s0168-1702(02)00189-2. [DOI] [PubMed] [Google Scholar]
- 84.Mesher D, Stanford E, White J, Findlow J, Warrington R, Das S. et al. HPV Serology Testing Confirms High HPV Immunisation Coverage in England. PloS One. 2016;11(3):e0150107. doi: 10.1371/journal.pone.0150107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dillner J. The serological response to papillomaviruses. Semin Cancer Biol. 1999 Dec;9(6):423–30. doi: 10.1006/scbi.1999.0146. [DOI] [PubMed] [Google Scholar]
- 86.Chung MY, Kim YW, Bae SM, Kwon EH, Chaturvedi PK, Battogtokh G. et al. Development of a bead-based multiplex genotyping method for diagnostic characterization of HPV infection. PloS One. 2012;7(2):e32259. doi: 10.1371/journal.pone.0032259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Day PM, Thompson CD, Buck CB, Pang YY, Lowy DR, Schiller JT. Neutralization of human papillomavirus with monoclonal antibodies reveals different mechanisms of inhibition. J Virol. 2007 Aug;81(16):8784–92. doi: 10.1128/JVI.00552-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Day PM, Pang YY, Kines RC, Thompson CD, Lowy DR, Schiller JT. A human papillomavirus (HPV) in vitro neutralization assay that recapitulates the in vitro process of infection provides a sensitive measure of HPV L2 infection-inhibiting antibodies. Clin Vaccine Immunol. 2012 Jul;19(7):1075–82. doi: 10.1128/CVI.00139-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Draper E, Bissett SL, Howell-Jones R, Edwards D, Munslow G, Soldan K. et al. Neutralization of non-vaccine human papillomavirus pseudoviruses from the A7 and A9 species groups by bivalent HPV vaccine sera. Vaccine. 2011;29(47):8585–90. doi: 10.1016/j.vaccine.2011.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Li Y, Liu J, Gong L, Sun X, Long W. Combining HPV DNA load with p16/Ki-67 staining to detect cervical precancerous lesions and predict the progression of CIN1-2 lesions. Virol J. 2019 Oct 16;16(1):117. doi: 10.1186/s12985-019-1225-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Yu L, Fei L, Liu X, Pi X, Wang L, Chen S. Application of p16/Ki-67 dual-staining cytology in cervical cancers. J Cancer. 2019 Jun 2;10(12):2654–2660. doi: 10.7150/jca.32743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Yu L, Wang L, Zhong J, Chen S. Diagnostic value of p16INK4A, Ki-67, and human papillomavirus L1 capsid protein immunochemical staining on cell blocks from residual liquid-based gynecologic cytology specimens. Cancer Cytopathol. 2010 Feb 25;118(1):47–55. doi: 10.1002/cncy.20061. [DOI] [PubMed] [Google Scholar]
- 93.Wang HR, Li YC, Guo HQ, Yu LL, Wu Z, Yin J. et al. A cocktail of p16INK4a and Ki-67, p16INK4a and minichromosome maintenance protein 2 as triage tests for human papillomavirus primary cervical cancer screening. Oncotarget. 2017 Aug 3;8(48):83890–83899. doi: 10.18632/oncotarget.19870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bello JO, Nieva LO, Paredes AC, Gonzalez AM, Zavaleta LR, Lizano M. Regulation of the Wnt/beta-Catenin Signaling Pathway by Human Papillomavirus E6 and E7 Oncoproteins. Viruses. 2015;7(8):4734–55. doi: 10.3390/v7082842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Talora C, Sgroi DC, Crum CP, Dotto GP. Specific down-modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV-E6/E7 expression and late steps of malignant transformation. Genes Dev. 2002 Sep 1;16(17):2252–63. doi: 10.1101/gad.988902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bossler F, Hoppe-Seyler K, Hoppe-Seyler F. PI3K/AKT/mTOR Signaling Regulates the Virus/Host Cell Crosstalk in HPV-Positive Cervical Cancer Cells. Int J Mol Sci. 2019 May 3;20(9):2188. doi: 10.3390/ijms20092188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hoppe-Seyler K, Bossler F, Braun JA, Herrmann AL, Hoppe-Seyler F. The HPV E6/E7 Oncogenes: Key Factors for Viral Carcinogenesis and Therapeutic Targets. Trends Microbiol. 2018 Feb;26(2):158–168. doi: 10.1016/j.tim.2017.07.007. [DOI] [PubMed] [Google Scholar]
- 98.Taghizadeh E, Jahangiri S, Rostami D, Taheri F, Renani PG, Taghizadeh H. et al. Roles of E6 and E7 Human Papillomavirus Proteins in Molecular Pathogenesis of Cervical Cancer. Curr Protein Pept Sci. 2019;20(9):926–934. doi: 10.2174/1389203720666190618101441. [DOI] [PubMed] [Google Scholar]
- 99.Zhang B, Spandau DF, Roman A. E5 protein of human papillomavirus type 16 protects human foreskin keratinocytes from UV B-irradiation-induced apoptosis. Virol J. 2002;76(1):220–31. doi: 10.1128/JVI.76.1.220-231.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wasson CW, Morgan EL, Muller M, Ross RL, Hartley M, Roberts S. et al. Human papillomavirus type 18 E5 oncogene supports cell cycle progression and impairs epithelial differentiation by modulating growth factor receptor signalling during the virus life cycle. Oncotarget. 2017;8(61):103581–600. doi: 10.18632/oncotarget.21658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Venuti A, Paolini F, Nasir L, Corteggio A, Roperto S, Campo MS. et al. Papillomavirus E5: the smallest oncoprotein with many functions. Mol Cancer. 2011 Nov 11;10:140. doi: 10.1186/1476-4598-10-140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Altavilla G, Staffieri A, Busatto G, Canesso A, Giacomelli L, Marioni G. Expression of p53, p16INK4A, pRb, p21WAF1/CIP1, p27KIP1, cyclin D1, Ki-67 and HPV DNA in sinonasal endophytic Schneiderian (inverted) papilloma. Acta Otolaryngol. 2009 Nov;129(11):1242–9. doi: 10.3109/00016480802620647. [DOI] [PubMed] [Google Scholar]
- 103.Wang X, Wang HK, Li Y, Hafner M, Banerjee NS, Tang S. et al. microRNAs are biomarkers of oncogenic human papillomavirus infections. Proc Natl Acad Sci U S A. 2014 Mar 18;111(11):4262–7. doi: 10.1073/pnas.1401430111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zamani S, Sohrabi A, Hosseini SM, Rahnamaye-Farzami M, Akbari A. Deregulation of miR-21 and miR-29a in Cervical Cancer Related to HPV Infection. Microrna. 2019;8(2):110–115. doi: 10.2174/2211536607666181017124349. [DOI] [PubMed] [Google Scholar]
- 105.Bumrungthai S, Ekalaksananan T, Evans MF, Chopjitt P, Tangsiriwatthana T, Patarapadungkit N. et al. Up-Regulation of miR-21 Is Associated with Cervicitis and Human Papillomavirus Infection in Cervical Tissues. PloS One. 2015;10(5):e0127109. doi: 10.1371/journal.pone.0127109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Myklebust MP, Bruland O, Fluge Ø, Skarstein A, Balteskard L, Dahl O. MicroRNA-15b is induced with E2F-controlled genes in HPV-related cancer. Br J Cancer. 2011 Nov 22;105(11):1719–25. doi: 10.1038/bjc.2011.457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lajer CB, Garnæs E, Friis-Hansen L, Norrild B, Therkildsen MH, Glud M. et al. The role of miRNAs in human papilloma virus (HPV)-associated cancers: bridging between HPV-related head and neck cancer and cervical cancer. Br J Cancer. 2017 Aug 22;117(5):e2. doi: 10.1038/bjc.2017.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Coimbra EC, DA Conceição Gomes Leitão M, Júnior MR, DE Oliveira TH, DA Costa Silva Neto J, DE Freitas AC. Expression Profile of MicroRNA-203 and its ΔNp63 Target in Cervical Carcinogenesis: Prospects for Cervical Cancer Screening. Anticancer Res. 2016 Aug;36(8):3939–46. [PubMed] [Google Scholar]
- 109.Park S, Kim J, Eom K, Oh S, Kim S, Kim G. et al. microRNA-944 overexpression is a biomarker for poor prognosis of advanced cervical cancer. BMC Cancer. 2019;19(1):419. doi: 10.1186/s12885-019-5620-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Fang H, Shuang D, Yi Z, Sheng H, Liu Y. Up-regulated microRNA-155 expression is associated with poor prognosis in cervical cancer patients. Biomed Pharmacother. 2016;83:64–9. doi: 10.1016/j.biopha.2016.06.006. [DOI] [PubMed] [Google Scholar]
- 111.Wang F, Li Y, Zhou J, Xu J, Peng C, Ye F. et al. miR-375 is down-regulated in squamous cervical cancer and inhibits cell migration and invasion via targeting transcription factor SP1. Am J Pathol. 2011;179(5):2580–8. doi: 10.1016/j.ajpath.2011.07.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Jayamohan S, Kannan M, Moorthy RK, Rajasekaran N, Jung HS, Shin YK. et al. Dysregulation of miR-375/AEG-1 Axis by Human Papillomavirus 16/18-E6/E7 Promotes Cellular Proliferation, Migration, and Invasion in Cervical Cancer. Front Oncol. 2019 Sep 9;9:847. doi: 10.3389/fonc.2019.00847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Stanley M. Pathology and epidemiology of HPV infection in females. Gynecol Oncol. 2010 May;117(2 Suppl):S5–10. doi: 10.1016/j.ygyno.2010.01.024. [DOI] [PubMed] [Google Scholar]
- 114.Garland SM, Steben M, Sings HL, James M, Lu S, Railkar R. et al. Natural history of genital warts: analysis of the placebo arm of 2 randomized phase III trials of a quadrivalent human papillomavirus (types 6, 11, 16, and 18) Vaccine. 2009;199(6):805–14. doi: 10.1086/597071. [DOI] [PubMed] [Google Scholar]
- 115.Hasanzadeh M, Rejali M, Mehramiz M, Akbari M, Mousavi Seresht L, Yazdandoost Y. et al. The interaction of high and low-risk human papillomavirus genotypes increases the risk of developing genital warts: A population-based cohort study. J Cell Biochem. 2019;120(8):12870–4. doi: 10.1002/jcb.28557. [DOI] [PubMed] [Google Scholar]
- 116.Mougin C, Dalstein V, Prétet JL, Gay C, Schaal JP, Riethmuller D. Epidémiologie des infections cervicales à papillomavirus Acquisitions récentes [Epidemiology of cervical papillomavirus infections Recent knowledge] Presse Med. 2001 Jun 9;30(20):1017–23. French. [PubMed] [Google Scholar]
- 117.Batista CS, Atallah AN, Saconato H, da Silva EM. 5-FU for genital warts in non-immunocompromised individuals. Cochrane Database Syst Rev. 2010 Apr 14;2010(4):CD006562. doi: 10.1002/14651858.CD006562.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Blomberg M, Friis S, Munk C, Bautz A, Kjaer SK. Genital warts and risk of cancer: a Danish study of nearly 50 000 patients with genital warts. J Infect Dis. 2012 May 15;205(10):1544–53. doi: 10.1093/infdis/jis228. [DOI] [PubMed] [Google Scholar]
- 119. 119 Baykal C, Yazganoğlu KD. Skin Appendage Tumors. In: Clinical Atlas of Skin Tumors. Springer, Berlin, Heidelberg. 2014.
- 120.Hogewoning CJ, Bleeker MC, van den Brule AJ, Voorhorst FJ, Snijders PJ, Berkhof J. et al. Condom use promotes regression of cervical intraepithelial neoplasia and clearance of human papillomavirus: a randomized clinical trial. Int J Cancer. 2003 Dec 10;107(5):811–6. doi: 10.1002/ijc.11474. [DOI] [PubMed] [Google Scholar]
- 121.Ferenczy A. Epidemiology and clinical pathophysiology of condylomata acuminata. Am J Obstet Gynecol. 1995 Apr;172(4 Pt 2):1331–9. doi: 10.1016/0002-9378(95)90399-2. [DOI] [PubMed] [Google Scholar]
- 122.Workowski KA, Berman SM. Sexually transmitted diseases treatment guidelines. 2006;55(11):38–42. [PubMed] [Google Scholar]
- 123.Nathan M, Hickey N, Mayuranathan L, Vowler SL, Singh N. Treatment of anal human papillomavirus-associated disease: a long term outcome study. Int J STD AIDS. 2008 Jul;19(7):445–9. doi: 10.1258/ijsa.2007.007290. [DOI] [PubMed] [Google Scholar]
- 124.Berry JM, Jay N, Cranston RD, Darragh TM, Holly EA, Welton ML. et al. Progression of anal high‐grade squamous intraepithelial lesions to invasive anal cancer among HIV‐infected men who have sex with men. Int J Cancer. 2014;134(5):1147–55. doi: 10.1002/ijc.28431. [DOI] [PubMed] [Google Scholar]
- 125.Fathi R, Tsoukas MM. Genital warts and other HPV infections: Established and novel therapies. Clin Dermatol. 2014;32(2):299–306. doi: 10.1016/j.clindermatol.2013.08.014. [DOI] [PubMed] [Google Scholar]
- 126.Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reprod Biol Endocrinol. 2015;13(1):37. doi: 10.1186/s12958-015-0032-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Krausz C. Male infertility: pathogenesis and clinical diagnosis. Best Pract Res Clin Endocrinol Metab. 2011;25(2):271–85. doi: 10.1016/j.beem.2010.08.006. [DOI] [PubMed] [Google Scholar]
- 128.Nasseri S, Monavari SH, Keyvani H, Nikkhoo B, Vahabpour Roudsari R, Khazeni M. The prevalence of Human Papilloma Virus (HPV) infection in the oligospermic and azoospermic men. Med J Islam Repub Iran. 2015;29(1):1001–6. [PMC free article] [PubMed] [Google Scholar]
- 129.Kyrgiou M, Athanasiou A, Paraskevaidi M, Mitra A, Kalliala I, Martin-Hirsch P. et al. Adverse obstetric outcomes after local treatment for cervical preinvasive and early invasive disease according to cone depth: systematic review and meta-analysis. BMJ. 2016;354:i3633. doi: 10.1136/bmj.i3633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zuo Z, Goel S, Carter JE. Association of cervical cytology and HPV DNA status during pregnancy with placental abnormalities and preterm birth. Am J Clin Pathol. 2011;136(2):260–5. doi: 10.1309/AJCP93JMIUEKRPIW. [DOI] [PubMed] [Google Scholar]
- 131.Luttmer R, Dijkstra MG, Snijders PJ, Hompes PG, Pronk DT, Hubeek I. et al. Presence of human papillomavirus in semen in relation to semen quality. Hum Reprod. 2016;31(2):280–6. doi: 10.1093/humrep/dev317. [DOI] [PubMed] [Google Scholar]
- 132.Jeršovienė V, Gudlevičienė Ž, Rimienė J, Butkauskas D. Human Papillomavirus and Infertility. Medicina (Kaunas) 2019;55(7):377. doi: 10.3390/medicina55070377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Wierzbicka M, Józefiak A, Szydłowski J, Marszałek A, Stankiewicz C, Hassman-Poznańska E. et al. Recommendations for the diagnosis of human papilloma virus (HPV) high and low risk in the prevention and treatment of diseases of the oral cavity, pharynx and larynx Guide of experts PTORL and KIDL. BMC Infect Dis. 2013;67(3):113–34. doi: 10.1016/j.otpol.2013.01.003. [DOI] [PubMed] [Google Scholar]
- 134.Yang A, Jeang J, Cheng K, Cheng T, Yang B, Wu TC. et al. Current state in the development of candidate therapeutic HPV vaccines. Expert Rev Vaccines. 2016;15(8):989–1007. doi: 10.1586/14760584.2016.1157477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Laurent JS, Luckett R, Feldman S. HPV vaccination and the effects on rates of HPV-related cancers. Curr Probl Cancer. 2018;42(5):493–506. doi: 10.1016/j.currproblcancer.2018.06.004. [DOI] [PubMed] [Google Scholar]
- 136.Dillner J, Arbyn M, Unger E, Dillner LJC, Immunology E. Monitoring of human papillomavirus vaccination. Clin Exp Immunol. 2011;163(1):17–25. doi: 10.1111/j.1365-2249.2010.04268.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kleinsorge F, Schmidmayr M. Humane Papillomavirusinfektion bei Frauen. Gynäkol Onkol. 2018;57(12):1445–51. doi: 10.1007/s00120-018-0795-z. [DOI] [PubMed] [Google Scholar]
- 138.Yang A, Farmer E, Wu TC, Hung CF. Perspectives for therapeutic HPV vaccine development. J Biomed Sci. 2016;23(1):75. doi: 10.1186/s12929-016-0293-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Steben M, Thompson MT, Rodier C, Mallette N, Racovitan V, DeAngelis F. et al. A review of the impact and effectiveness of the quadrivalent human papillomavirus vaccine: 10 years of clinical experience in Canada. J Obstet Gynaecol Can. 2018;40(12):1635–45. doi: 10.1016/j.jogc.2018.05.024. [DOI] [PubMed] [Google Scholar]
- 140.Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow SN, Apter D. et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009;374(9686):301–14. doi: 10.1016/S0140-6736(09)61248-4. [DOI] [PubMed] [Google Scholar]
- 141.Clifford GM, Rana RK, Franceschi S, Smith JS, Gough G, Pimenta JM. et al. Human papillomavirus genotype distribution in low-grade cervical lesions: comparison by geographic region and with cervical cancer. Cancer Epidemiol Biomarkers Prev. 2005;14(5):1157–64. doi: 10.1158/1055-9965.EPI-04-0812. [DOI] [PubMed] [Google Scholar]
- 142.Smith JS, Lindsay L, Hoots B, Keys J, Franceschi S, Winer R. et al. Human papillomavirus type distribution in invasive cervical cancer and high‐grade cervical lesions: a meta‐analysis update. Int J Cancer. 2007;121(3):621–32. doi: 10.1002/ijc.22527. [DOI] [PubMed] [Google Scholar]
- 143.Ikeda SI, Hineno A, Ozawa K, Kinoshita TJIr. Suspected adverse effects after human papillomavirus vaccination: a temporal relationship. Drug Saf. 2018;66(6):723–5. doi: 10.1007/s12026-018-9063-y. [DOI] [PubMed] [Google Scholar]
- 144. WHO. https://www.who.int/immunization/hpv/deliver/en/. 2020.
- 145.De Vuyst H, Clifford GM, Nascimento MC, Madeleine MM, Franceschi S. Prevalence and type distribution of human papillomavirus in carcinoma and intraepithelial neoplasia of the vulva, vagina and anus: a meta‐analysis. Int J Cancer. 2009;124(7):1626–36. doi: 10.1002/ijc.24116. [DOI] [PubMed] [Google Scholar]
- 146.Hernandez BY, Wilkens LR, Zhu X, Thompson P, McDuffie K, Shvetsov YB. et al. Transmission of human papillomavirus in heterosexual couples. Emerg Infect Dis. 2008;14(6):888. doi: 10.3201/eid1406.070616.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Baldwin JI, Baldwin JD. Heterosexual anal intercourse: an understudied, high-risk sexual behavior. Arch Sex Behav. 2000;29(4):357–73. doi: 10.1023/a:1001918504344. [DOI] [PubMed] [Google Scholar]
- 148.Zhen S, Li X, Biochemistry Biochemistry. Oncogenic human papillomavirus: application of CRISPR/Cas9 therapeutic strategies for cervical cancer. Cell Physiol Biochem. 2017;44(6):2455–66. doi: 10.1159/000486168. [DOI] [PubMed] [Google Scholar]
- 149.Šarenac T, Mikov M. Cervical cancer, different treatments and importance of bile acids as therapeutic agents in this disease. Front Pharmacol. 2019;10 doi: 10.3389/fphar.2019.00484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Barbosa MS, Vass WC, Lowy DR, Schiller JT. In vitro biological activities of the E6 and E7 genes vary among human papillomaviruses of different oncogenic potential. J Virol. 1991;65(1):292–8. doi: 10.1128/jvi.65.1.292-298.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Halbert C, Demers G, Galloway D. The E7 gene of human papillomavirus type 16 is sufficient for immortalization of human epithelial cells. J Virol. 1991;65(1):473–8. doi: 10.1128/jvi.65.1.473-478.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Lacey CJN, Guimera N, Garland SM. Chapter 10 - Low-risk Human Papillomavirus: Genital Warts, Cancer and Respiratory Papillomatosis. In: Jenkins D, Bosch FX, editors. Human Papillomavirus: Academic Press; 2020. p. 165-78.
- 153.Meites E, Szilagyi PG, Chesson HW, Unger ER, Romero JR, Markowitz LE. Human papillomavirus vaccination for adults: Updated recommendations of the Advisory Committee on Immunization Practices. 2019;19(11):3202–6. doi: 10.15585/mmwr.mm6832a3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Rodriguez AM, Do TQN, Goodman M, Schmeler KM, Kaul S, Kuo YF. Human papillomavirus vaccine interventions in the US: A systematic review and meta-analysis. Am J Prev Med. 2019 Apr;56(4):591–602. doi: 10.1016/j.amepre.2018.10.033. [DOI] [PubMed] [Google Scholar]
- 155.Di Tucci C, Schiavi MC, Faiano P, D’Oria O, Prata G, Sciuga V. et al. Therapeutic vaccines and immune checkpoints inhibition options for gynecological cancers. Crit Rev Oncol Hematol. 2018;128:30–42. doi: 10.1016/j.critrevonc.2018.05.011. [DOI] [PubMed] [Google Scholar]
- 156. https://www.hpvcentre.net/. 2020.
- 157. Clifford G, Franceschi S, Diaz M, Munoz N, Villa L. Chapter 3: HPV type-distribution in women with and without cervical neoplastic diseases. Vaccine 24. S3/26–34, https://doi. org/10.1016/j. vaccine. 2006.05 026; 2006. [DOI] [PubMed]
- 158.Alkatout I, Schubert M, Garbrecht N, Weigel MT, Jonat W, Mundhenke C. et al. Vulvar cancer: epidemiology, clinical presentation, and management options. Int J Womens Health. 2015;7:305. doi: 10.2147/IJWH.S68979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Rajaram S, Maheshwari A, Srivastava AJBP, Obstetrics RC. Staging for vaginal cancer. Best Pract Res Clin Obstet Gynaecol. 2015;29(6):822–32. doi: 10.1016/j.bpobgyn.2015.01.006. [DOI] [PubMed] [Google Scholar]
- 160.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
- 161.Watson M, Saraiya M, Wu X. Update of HPV-associated female genital cancers in the United States, 1999–2004. J Womens Health (Larchmt) 2009;18(11):1731–8. doi: 10.1089/jwh.2009.1570. [DOI] [PubMed] [Google Scholar]
- 162.Insinga RP, Liaw KL, Johnson LG, Madeleine MM. A systematic review of the prevalence and attribution of human papillomavirus types among cervical, vaginal, and vulvar precancers and cancers in the United States. Cancer Epidemiol Biomarkers Prev. 2008;17(7):1611–22. doi: 10.1158/1055-9965.EPI-07-2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Uronis HE, Bendell JC. Anal cancer: an overview. Oncologist. 2007;12(5):524–34. doi: 10.1634/theoncologist.12-5-524. [DOI] [PubMed] [Google Scholar]
- 164.Sobhani I, Vuagnat A, Walker F, Vissuzaine C, Mirin B, Hervatin F. et al. Prevalence of high-grade dysplasia and cancer in the anal canal in human papillomavirus–infected individuals. Gastroenterology. 2001;120(4):857–66. doi: 10.1053/gast.2001.22446. [DOI] [PubMed] [Google Scholar]
- 165. Cancer Control Strategy for Poland 2015-2024. 10 June 2014. Avaliable Online at: http://www.walkazrakiem.pl/sites/default/ files/library/files/polish_cancer_control_strategy_0.pdf.
- 166. https://www.cdc.gov/cancer/hpv/statistics/anal.htm.
- 167.Boda D, Docea AO, Calina D, Ilie MA, Caruntu C, Zurac S. et al. Human papilloma virus: Apprehending the link with carcinogenesis and unveiling new research avenues. Int J Oncol. 2018;52(3):637–55. doi: 10.3892/ijo.2018.4256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DMJIjoc. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917. doi: 10.1002/ijc.25516. [DOI] [PubMed] [Google Scholar]
- 169.Ludmir EB, Stephens SJ, Palta M, Willett CG, Czito BG. Human papillomavirus tumor infection in esophageal squamous cell carcinoma. J Gastrointest Oncol. 2015;6(3):287. doi: 10.3978/j.issn.2078-6891.2015.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Liu H, Li J, Diao M, Cai Z, Yang J, Zeng Y. Statistical analysis of human papillomavirus in a subset of upper aerodigestive tract tumors. J Med Virol. 2013;85(10):1775–85. doi: 10.1002/jmv.23662. [DOI] [PubMed] [Google Scholar]
- 171.Enzinger PC, Mayer R. Esophageal cancer. N Engl J Med. 2003;349(23):2241–52. doi: 10.1056/NEJMra035010. [DOI] [PubMed] [Google Scholar]
- 172.Wang X, Tian X, Liu F, Zhao Y, Sun M, Chen D. et al. Detection of HPV DNA in esophageal cancer specimens from different regions and ethnic groups: a descriptive study. BMC Cancer. 2010;10(1):19. doi: 10.1186/1471-2407-10-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet‐Tieulent J, Jemal A. Global cancer statistics. CA Cancer J Clin. 2012 2015;65(2):87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
- 174.Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64(2):104–17. doi: 10.3322/caac.21220. [DOI] [PubMed] [Google Scholar]
- 175.Lorenzon L, Mazzetta F, Pilozzi E, Uggeri G, Torrisi MR, Ferri M. et al. Human papillomavirus does not have a causal role in colorectal carcinogenesis. World J Gastroenterol. 2015;21(1):342. doi: 10.3748/wjg.v21.i1.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Bodaghi S, Yamanegi K, Xiao S-, Da Costa M, Palefsky JM, Zheng ZM. Colorectal papillomavirus infection in patients with colorectal cancer. Clin Cancer Res. 2005;11(8):2862–7. doi: 10.1158/1078-0432.CCR-04-1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH. et al. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 2016;66(4):271–89. doi: 10.3322/caac.21349. [DOI] [PubMed] [Google Scholar]
- 178.Quinn M, Babb PJBi. Patterns and trends in prostate cancer incidence, survival, prevalence and mortality Part I: international comparisons. BJU Int. 2002;90(2):162–73. doi: 10.1046/j.1464-410x.2002.2822.x. [DOI] [PubMed] [Google Scholar]
- 179.Grönberg H. Prostate cancer epidemiology. Lancet. 2003;361(9360):859–64. doi: 10.1016/S0140-6736(03)12713-4. [DOI] [PubMed] [Google Scholar]
- 180.Bae JM. Human papillomavirus 16 infection as a potential risk factor for prostate cancer: an adaptive meta-analysis. Epidemiol Health. 2015;37 doi: 10.4178/epih/e2015005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Hsing AW, Tsao L, Devesa SS. International trends and patterns of prostate cancer incidence and mortality. Int J Cancer. 2000;85(1):60–7. doi: 10.1002/(sici)1097-0215(20000101)85:1<60::aid-ijc11>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 182.Thuener JE. Urologic Malignancies. Prim Care. 2019;46(2):275–85. doi: 10.1016/j.pop.2019.02.009. [DOI] [PubMed] [Google Scholar]
- 183.Cai T, Mazzoli S, Meacci F, Nesi G, Geppetti P, Malossini G. et al. Human papillomavirus and non-muscle invasive urothelial bladder cancer: potential relationship from a pilot study. Urol Int. 2011;25(2):485–9. doi: 10.3892/or.2010.1083. [DOI] [PubMed] [Google Scholar]
- 184.Khaled HM, Bahnassi AA, Zekri A-RN, Kassem HA, Mokhtar N, editors editors. Correlation between p53 mutations and HPV in bilharzial bladder cancer Urologic Oncology: Seminars and Original Investigations. Urol Oncol. 2003;21(5):334–341. doi: 10.1016/s1078-1439(03)00014-0. [DOI] [PubMed] [Google Scholar]
- 185.Mvula M, Iwasaka T, Iguchi A, Nakamura S, Masaki Z, Sugimori H. Do human papillomaviruses have a role in the pathogenesis of bladder carcinoma? J Urol. 1996;155(2):471–4. [PubMed] [Google Scholar]
- 186.Pashos CL, Botteman MF, Laskin BL, Redaelli A. Bladder cancer: epidemiology, diagnosis, and management. Cancer Pract. 2002;10(6):311–22. doi: 10.1046/j.1523-5394.2002.106011.x. [DOI] [PubMed] [Google Scholar]
- 187.Bryant P, Davies P, Wilson D. Detection of human papillomavirus DNA in cancer of the urinary bladder by in situ hybridisation. Br J Urol. 1991;68(1):49–52. doi: 10.1111/j.1464-410x.1991.tb15256.x. [DOI] [PubMed] [Google Scholar]
- 188.Purdue MP, Devesa SS, Sigurdson AJ, McGlynn KA. International patterns and trends in testis cancer incidence. Int J Cancer. 2005;115(5):822–7. doi: 10.1002/ijc.20931. [DOI] [PubMed] [Google Scholar]
- 189.Yousif L, Hammer GP, Blettner M, Zeeb H. Testicular cancer and viral infections: A systematic literature review and meta‐analysis. J Med Virol. 2013;85(12):2165–75. doi: 10.1002/jmv.23704. [DOI] [PubMed] [Google Scholar]
- 190.Strickler H, Schiffman M, Shah K, Rabkin C, Schiller J, Wacholder S. et al. A survey of human papillomavirus 16 antibodies in patients with epithelial cancers. Cancer Epidemiol. 1998;7(4):305–13. doi: 10.1097/00008469-199808000-00006. [DOI] [PubMed] [Google Scholar]
- 191.Kondoh G, Murata Y, Aozasa K, Yutsudo M, Hakura A. Very high incidence of germ cell tumorigenesis (seminomagenesis) in human papillomavirus type 16 transgenic mice. J Virol. 1991;65(6):3335–9. doi: 10.1128/jvi.65.6.3335-3339.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Bleeker M, Heideman D, Snijders P, Horenblas S, Dillner J, Meijer C. Penile cancer: epidemiology, pathogenesis and prevention. World J Urol. 2009;27(2):141. doi: 10.1007/s00345-008-0302-z. [DOI] [PubMed] [Google Scholar]
- 193.McCance DJ, Kopan R, Fuchs E, Laimins LAJPotNAoS. Human papillomavirus type 16 alters human epithelial cell differentiation in vitro. Proc Natl Acad Sci U S A. 1988;85(19):7169–73. doi: 10.1073/pnas.85.19.7169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Hakenberg OW, Compérat EM, Minhas S, Necchi A, Protzel C, Watkin N. EAU guidelines on penile cancer: 2014 update. Eur Urol. 2015;67(1):142–50. doi: 10.1016/j.eururo.2014.10.017. [DOI] [PubMed] [Google Scholar]
- 195.Marur S, Forastiere AA, editors editors. Head and neck cancer: changing epidemiology, diagnosis, and treatment Mayo Clinic Proceedings. Mayo Clin Proc. 2008 Apr;83(4):489–501. doi: 10.4065/83.4.489. [DOI] [PubMed] [Google Scholar]
- 196.Gillison ML, Broutian T, Pickard RK, Tong ZY, Xiao W, Kahle L. et al. Prevalence of oral HPV infection in the United States, 2009-2010. JAMA. 2012;307(7):693–703. doi: 10.1001/jama.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Curado MP, Hashibe M. Recent changes in the epidemiology of head and neck cancer. Curr Opin Oncol. 2009;21(3):194–200. doi: 10.1097/CCO.0b013e32832a68ca. [DOI] [PubMed] [Google Scholar]
- 198. Karia PS. Epidemiology and outcomes of cutaneous squamous cell carcinoma. High-Risk Cutaneous Squamous Cell Carcinoma: Springer; 2016. p. 3-28.
- 199.Karia PS, Han J, Schmults CD. Cutaneous squamous cell carcinoma: estimated incidence of disease, nodal metastasis, and deaths from disease in the United States, 2012. J Am Acad Dermatol. 2013;68(6):957–66. doi: 10.1016/j.jaad.2012.11.037. [DOI] [PubMed] [Google Scholar]
- 200.Gewirtzman A, Bartlett B, Tyring S. Epidermodysplasia verruciformis and human papilloma virus. Curr Opin Infect Dis. 2008;21(2):141–6. doi: 10.1097/QCO.0b013e3282f486c9. [DOI] [PubMed] [Google Scholar]
- 201.Ramoz N, Rueda LA, Bouadjar B, Montoya LS, Orth G, Favre M. Mutations in two adjacent novel genes are associated with epidermodysplasia verruciformis. Nat Genet. 2002;32(4):579–81. doi: 10.1038/ng1044. [DOI] [PubMed] [Google Scholar]
- 202.Satterwhite CL, Torrone E, Meites E, Dunne EF, Mahajan R, Ocfemia MCB. et al. Sexually transmitted infections among US women and men: prevalence and incidence estimates, 2008. Sex Transm Dis. 2013;40(3):187–93. doi: 10.1097/OLQ.0b013e318286bb53. [DOI] [PubMed] [Google Scholar]
- 203.Moscicki AB. Impact of HPV infection in adolescent populations. J Adolesc Health. 2005;37(6):S3–S9. doi: 10.1016/j.jadohealth.2005.09.011. [DOI] [PubMed] [Google Scholar]
- 204.Wright Jr TC, Massad LS, Dunton CJ, Spitzer M, Wilkinson EJ, Solomon D. et al. 2006 consensus guidelines for the management of women with cervical intraepithelial neoplasia or adenocarcinoma in situ. Am J Obstet Gynecol. 2007;197(4):340–5. doi: 10.1016/j.ajog.2007.07.050. [DOI] [PubMed] [Google Scholar]
- 205.Tainio K, Athanasiou A, Tikkinen KA, Aaltonen R, Cárdenas J, Glazer-Livson S. et al. Clinical course of untreated cervical intraepithelial neoplasia grade 2 under active surveillance: systematic review and meta-analysis. BMJ. 2018;360:k499. doi: 10.1136/bmj.k499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Grce M, Mravak-Stipetić M. Human papillomavirus–associated diseases. Clin Dermatol. 2014;32(2):253–8. doi: 10.1016/j.clindermatol.2013.10.006. [DOI] [PubMed] [Google Scholar]
- 207.Bruni L, Diaz M, Castellsagué M, Ferrer E, Bosch FX, de Sanjosé S. Cervical human papillomavirus prevalence in 5 continents: meta-analysis of 1 million women with normal cytological findings. J Infect Dis. 2010;202(12):1789–99. doi: 10.1086/657321. [DOI] [PubMed] [Google Scholar]
- 208.Skapa P, Robová H, Rob L, Zámečník J. Review of precancerous vulvar lesions. Cesk Patol. 2012;48(1):15–21. [PubMed] [Google Scholar]
- 209.Rakislova N, Saco A, Sierra A, del Pino M, Ordi J. Role of human papillomavirus in vulvar cancer. Adv Anat Pathol. 2017;24(4):201–14. doi: 10.1097/PAP.0000000000000155. [DOI] [PubMed] [Google Scholar]
- 210.Nelson EL, Bogliatto F, Stockdale CKJCo, gynecology gynecology. Vulvar intraepithelial neoplasia (VIN) and condylomata. Clin Obstet Gynecol. 2015;58(3):512–25. doi: 10.1097/GRF.0000000000000132. [DOI] [PubMed] [Google Scholar]
- 211.Miles B, Safran HP, Monk B. Therapeutic options for treatment of human papillomavirus-associated cancers-novel immunologic vaccines: Gynecol Oncol Res Pract. ADXS11–001. 2017;4(1):10. doi: 10.1186/s40661-017-0047-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Lytwyn A, Salit IE, Raboud J, Chapman W, Darragh T, Winkler B. et al. Interobserver agreement in the interpretation of anal intraepithelial neoplasia. Cancer. 2005;103(7):1447–56. doi: 10.1002/cncr.20927. [DOI] [PubMed] [Google Scholar]
- 213.Levi FJEJoC. Cancer incidence in five continents, vol VI: A Review by Fabio Levi. Eur J Cancer. 1993;29(16):2315–9. [Google Scholar]
- 214.Ouhoummane N, Steben M, Coutlee F, Vuong T, Forest P, Rodier C. et al. Squamous anal cancer: patient characteristics and HPV type distribution. Cancer Epidemiol. 2013;37(6):807–12. doi: 10.1016/j.canep.2013.09.015. [DOI] [PubMed] [Google Scholar]
- 215.Martin D, Balermpas P, Winkelmann R, Rödel F, Rödel C, Fokas E. Anal squamous cell carcinoma–State of the art management and future perspectives. Cancer Treat Rev. 2018;65:11–21. doi: 10.1016/j.ctrv.2018.02.001. [DOI] [PubMed] [Google Scholar]
- 216.Bucchi D, Stracci F, Buonora N, Masanotti G. Human papillomavirus and gastrointestinal cancer: A review. World J Gastroenterol. 2016;22(33):7415. doi: 10.3748/wjg.v22.i33.7415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Voeten D, Goedegebuure R, Heineman D, Daams F. Non-metastatic oesophageal cancer: diagnosis and treatment. Ned Tijdschr Geneeskd. 2019;163 [PubMed] [Google Scholar]
- 218.Salepci T, Yazici H, Dane F, Topuz E, Dalay N, Onat H. et al. Detection of human papillomavirus DNA by polymerase chain reaction and southern blot hybridization in colorectal cancer patients. J BUON. 2009;14(3):495–9. [PubMed] [Google Scholar]
- 219.Navarro M, Nicolas A, Ferrandez A, Lanas A. Colorectal cancer population screening programs worldwide in 2016: An update. World J Gastroenterol. 2017;23(20):3632. doi: 10.3748/wjg.v23.i20.3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kuipers EJ, Grady WM, Lieberman D, Seufferlein T, Sung JJ, Boelens PG. et al. Colorectal cancer. Nat Rev Dis Primers. 2015;1:15065. doi: 10.1038/nrdp.2015.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Tolstov Y, Hadaschik B, Pahernik S, Hohenfellner M, Duensing S, editors. Human papillomaviruses in urological malignancies: a critical assessment. In Urologic Oncology: Seminars and Original Investigations 2014 Jan 1 (Vol. 32, No. 1, pp. 46-e19). Elsevier. [DOI] [PubMed]
- 222.Jimenez-Pacheco A, Exposito-Ruiz M, Arrabal-Polo MA, Lopez-Luque A. Meta-analysis of studies analyzing the role of human papillomavirus in the development of bladder carcinoma. Korean J Urol. 2012;53(4):240–7. doi: 10.4111/kju.2012.53.4.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Chou R, Dana T. Screening adults for bladder cancer: a review of the evidence for the US preventive services task force. Ann Intern Med. 2010;153(7):461–8. doi: 10.7326/0003-4819-153-7-201010050-00009. [DOI] [PubMed] [Google Scholar]
- 224.Kamat AM. Bladder cancer: 2017 and beyond. Indian J Urol. 2017;33(2):104–5. doi: 10.4103/0970-1591.203419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Bochner B, Hansel D, Efstathiou J, Konety B, Lee C, McKiernan J, et al. Urinary bladder. 2017:757.
- 226.Garolla A, Pizzol D, Bertoldo A, Ghezzi M, Carraro U, Ferlin A. et al. Testicular cancer and HPV semen infection. Front Endocrinol (Lausanne) 2012;3:172. doi: 10.3389/fendo.2012.00172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Ilic D, Misso ML. Screening for testicular cancer. Cochrane Database Syst Rev. 2011 Feb 16;(2):CD007853. doi: 10.1002/14651858.CD007853.pub2. [DOI] [PubMed] [Google Scholar]
- 228.Dieckmann KP, Frey U, Lock G. Contemporary diagnostic work-up of testicular germ cell tumours. Nat Rev Urol. 2013;10(12):703. doi: 10.1038/nrurol.2013.254. [DOI] [PubMed] [Google Scholar]
- 229.Cheng L, Albers P, Berney DM, Feldman DR, Daugaard G, Gilligan T. et al. Testicular cancer. Nat Rev Dis Primers. 2018;4(1):1–24. doi: 10.1038/s41572-018-0029-0. [DOI] [PubMed] [Google Scholar]
- 230.Farhadi A, Behzad‐Behbahani A, Geramizadeh B, Sekawi Z, Rahsaz M, Sharifzadeh S. High‐risk human papillomavirus infection in different histological subtypes of renal cell carcinoma. J Med Virol. 2014;86(7):1134–44. doi: 10.1002/jmv.23945. [DOI] [PubMed] [Google Scholar]
- 231.Rotola A, Monini P, di Luca D, Savioli A, Simone R, Secchiero P. et al. Presence and physical state of HPV DNA in prostate and urinary‐tract tissues. Int J Cancer. 1992;52(3):359–65. doi: 10.1002/ijc.2910520306. [DOI] [PubMed] [Google Scholar]
- 232.Khoury JD, Tannir NM, Williams MD, Chen Y, Yao H, Zhang J. et al. Landscape of DNA virus associations across human malignant cancers: analysis of 3,775 cases using RNA-Seq. J Virol. 2013;87(16):8916–26. doi: 10.1128/JVI.00340-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Herts BR, Silverman SG, Hindman NM, Uzzo RG, Hartman RP, Israel GM. et al. Management of the incidental renal mass on CT: a white paper of the ACR Incidental Findings Committee. J Am Coll Radiol. 2018;15(2):264–73. doi: 10.1016/j.jacr.2017.04.028. [DOI] [PubMed] [Google Scholar]
- 234.Giuliano AR, Lee JH, Fulp W, Villa LL, Lazcano E, Papenfuss MR. et al. Incidence and clearance of genital human papillomavirus infection in men (HIM): a cohort study. Lancet. 2011;377(9769):932–40. doi: 10.1016/S0140-6736(10)62342-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Sievert KD, Dräger DL, Köhn FM, Milerski S, Protzel C, Hakenberg O. Peniskarzinom: Diagnose und Staging. Der Urologe. 2018;57(4):418–22. doi: 10.1007/s00120-018-0602-x. [DOI] [PubMed] [Google Scholar]
- 236.Schlenker B, Schneede P. The Role of Human Papilloma Virus in Penile Cancer Prevention and New Therapeutic Agents. Eur Urol Focus. 2019;5(1):42–5. doi: 10.1016/j.euf.2018.09.010. [DOI] [PubMed] [Google Scholar]
- 237. Johnston MJ, Nigam R. Recent advances in the management of penile cancer. F1000Research 2019, 8(F1000 Faculty Rev):558 Last updated: 26 APR 2019;8.
- 238.Spiess PE, Dhillon J, Baumgarten AS, Johnstone PA, Giuliano AR. Pathophysiological basis of human papillomavirus in penile cancer: Key to prevention and delivery of more effective therapies. CA Cancer J Clin. 2016;66(6):481–95. doi: 10.3322/caac.21354. [DOI] [PubMed] [Google Scholar]
- 239.Crispen PL, Mydlo JH. Penile intraepithelial neoplasia and other premalignant lesions of the penis. Urol Clin North Am. 2010;37(3):335–42. doi: 10.1016/j.ucl.2010.04.003. [DOI] [PubMed] [Google Scholar]
- 240.Dayyani F, Etzel CJ, Liu M, Ho CH, Lippman SM, Tsao AS. 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(1):15. doi: 10.1186/1758-3284-2-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Chen AM, Felix C, Wang PC, Hsu S, Basehart V, Garst J. et al. Reduced-dose radiotherapy for human papillomavirus-associated squamous-cell carcinoma of the oropharynx: a single-arm, phase 2 study. Lancet Oncol. 2017;18(6):803–11. doi: 10.1016/S1470-2045(17)30246-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Wagner S, Reder H, Sharma S, Würdemann N, Wittekindt C, Klußmann J. Das HPV-getriebene Oropharynxkarzinom–Inzidenz, Trends, Diagnose und Therapie. Laryngorhinootologie. 2018;57(12):1457–63. doi: 10.1007/s00120-018-0810-4. [DOI] [PubMed] [Google Scholar]
- 243.Kim KY, Lewis Jr JS, Chen ZJT. Current status of clinical testing for human papillomavirus in oropharyngeal squamous cell carcinoma. J Pathol Clin Res October. 2018;4:213–226. doi: 10.1002/cjp2.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Kobayashi K, Hisamatsu K, Suzui N, Hara A, Tomita H, Miyazaki T. A review of HPV-related head and neck cancer. J Clin Med. 2018;7(9):241. doi: 10.3390/jcm7090241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Kabir S, Schmults CD, Ruiz ES. A review of cutaneous squamous cell carcinoma epidemiology, diagnosis, and management. Int J Cancer Manag. 2018;11(1) [Google Scholar]
- 246.Kaliterna V, Barisic Z. Genital human papillomavirus infections. Front Biosci (Landmark Ed) 2018;6:8. doi: 10.2741/4662. [DOI] [PubMed] [Google Scholar]


