Published online May 24, 2025. doi: 10.5306/wjco.v16.i5.105055
Revised: February 26, 2025
Accepted: March 26, 2025
Published online: May 24, 2025
Processing time: 129 Days and 14.2 Hours
Human papillomavirus (HPV) infection is a common sexually transmitted disease and a leading cause of cervical, other anogenital, and oropharyngeal cancers. Screening for precancerous lesions is an effective strategy for preventing HPV-related tumors. Although HPV vaccination has significantly reduced the incidence of cervical cancer, achieving universal coverage remains challenging because of cost barriers, particularly in economically disadvantaged regions. This review provides an update of HPV infection characteristics, screening methods, and strategies tailored to low-resource settings. We also discuss the global burden of HPV-related diseases, regional disparities in the implementation of screening, and future research directions. By examining the current challenges and opportunities, this review aims to inform policymakers and healthcare providers in designing effective, affordable, and scalable screening programs.
Core Tip: Human papillomavirus (HPV) is the main cause of cervical cancer, and screening for precancerous lesions is an effective measure to prevent tumors caused by HPV. Although HPV vaccination can effectively prevent the occurrence of cervical cancer, universal coverage cannot be achieved due to the cost. For economically disadvantaged areas, affordable and effective HPV cervical cancer screening strategies remain a major public health concern. Therefore, it is necessary to summarize the prevention and control methods and strategies of HPV virus screening to provide references for countries to formulate public policy research.
- Citation: Chen SH, Song YY, Gan N, Wang PT, Yan K, Wang SF, Zu YE, Peng XW. Human papillomavirus infection and screening strategies. World J Clin Oncol 2025; 16(5): 105055
- URL: https://www.wjgnet.com/2218-4333/full/v16/i5/105055.htm
- DOI: https://dx.doi.org/10.5306/wjco.v16.i5.105055
Cervical cancer remains one of the most prevalent and deadly cancers affecting women globally, particularly in low- and middle-income countries (LMICs), where it imposes a significant public health burden[1,2]. This malignant tumor originates in the cervix and is predominantly driven by persistent infection with high-risk types of human papillomavirus (HPV) such as HPV 16 and HPV 18[3]. Upon infection, viral DNA integrates into the host cell genome, disrupting normal cellular functions and leading to uncontrolled proliferation and malignant transformation[4]. Progression from HPV infection to invasive cervical cancer is typically slow, often spanning years or even decades. During this time, cervical cells undergo a series of pathological changes, evolving from normal epithelium to precancerous lesions [e.g., cervical intraepithelial neoplasia (CIN)] and eventually to invasive cancer[5]. Early stage cervical cancer is often asymptomatic; however, as the disease advances, symptoms such as irregular vaginal bleeding, postcoital bleeding, and abnormal vaginal discharge may emerge, underscoring the critical importance of early detection and intervention[1].
Cervical cancer is largely preventable through effective screening and vaccination programs. Widely used screening techniques, such as Pap smear and HPV DNA testing, have proven effective in detecting precancerous lesions and early stage cancers, enabling timely treatment and significantly reducing mortality rates[6,7]. HPV vaccination has emerged as a powerful tool for primary prevention, significantly reducing the incidence of HPV infection and associated cancers[8,9]. Despite these advances, the implementation of comprehensive screening and vaccination programs remains uneven, particularly in resource-limited settings, where access to healthcare services is often constrained by financial, logistical, and cultural barriers[9,10]. Disparities in screening coverage and vaccine accessibility persist between high- and low-income regions, highlighting the urgent need for tailored strategies to address these inequities[10,11].
In economically disadvantaged regions, the development of affordable and scalable screening strategies remains a critical public health priority in economically disadvantaged regions. Recent advancements in self-sampling technologies and point-of-care diagnostics offer promising solutions for overcoming barriers related to cost, infrastructure, and patient compliance[12,13]. These innovations enhance screening coverage, protect patient privacy and improve adherence to regular screening protocols. The integration of advanced analytical tools such as artificial intelligence and machine learning has the potential to improve the accuracy and efficiency of cervical cancer screening programs[14,15].
This review provides a comprehensive overview of current HPV screening approaches, their applications, and their implications for global cervical cancer prevention. By examining the characteristics of HPV infection, strengths and limitations of existing screening technologies, and challenges faced in resource-constrained settings, we seek to inform policymakers and healthcare providers in designing effective, affordable, and scalable screening programs. Additionally, we explored emerging trends in cervical cancer prevention, including the role of self-sampling, technological innovation, and international collaboration, to address disparities and reduce the global burden of cervical cancer. Through this analysis, we aim to contribute to ongoing efforts to achieve equitable access to cervical cancer prevention and control worldwide.
HPV is a small, double-stranded DNA virus that infects epithelial cells, leading to cellular proliferation and potential malignant transformation[16,17]. HPV is classified into high-, medium-, and low-risk types based on their oncogenic potential. High-risk HPV types, particularly HPV 16 and HPV 18, are responsible for the majority of HPV-related cancers, including cervical, anogenital, and oropharyngeal cancers[18,19]. These high-risk types integrate their viral DNA into the host genome[20,21], disrupting key tumor suppressor genes, such as p53 and Rb, which are critical for regulating cell cycle progression and apoptosis[22-25]. This integration leads to the overexpression of viral oncoproteins E6 and E7[3], which promotes uncontrolled cell proliferation, genomic instability, and malignant transformation[17,26]. In contrast, low-risk HPV types such as HPV 6 and 11 are primarily associated with benign lesions, including genital warts. These types rarely integrate into the host genome, and do not typically cause cancer. However, they can cause significant morbidity owing to the formation of recurrent or persistent lesions[27-31].
The natural history of HPV infection involves several stages[4,32]: (1) Initial infection: HPV infects the basal epithelial cells through microabrasions in the mucosal or cutaneous epithelium; (2) Viral replication: HPV replicates in the differentiated layers of the epithelium, utilizing host cell machinery; (3) Immune evasion: HPV evades immune detection by downregulating interferon responses and other immune mechanisms; and (4) Progression to cancer: In high-risk HPV infections, persistent viral integration and oncoprotein expression lead to cellular transformation and the development of precancerous lesions (e.g., CIN), which can progress to invasive cancer over years or decades.
Understanding the molecular mechanisms of HPV-induced carcinogenesis is critical for the development of targeted prevention and treatment strategies. For example, the role of E6 and E7 oncoproteins in disrupting cellular homeostasis has led to the development of therapeutic vaccines and targeted therapies aimed at inhibiting these proteins[33-35].
HPV is primarily transmitted through sexual contact or close physical interactions. To date, > 200 distinct HPV types have been identified[36]. In 2020, it was estimated that there were 604127 new cases of cervical cancer worldwide, resulting in 341831 disease-related deaths. The age-standardized incidence rate of cervical cancer is 13.3 cases per 100,000 women per year, with a corresponding mortality rate of 7.2 deaths per 100000 women per year[37]. Based on their pathogenicity and carcinogenic potential, HPV genotypes are categorized into high- and low-risk groups: (1) High-risk HPV types, including HPV 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, 68, 69, 73, and 82, are strongly associated with the development of cervical cancer and precancerous lesions[38-41]. HPV 16 and 18 are the most prevalent, accounting for 56.8%-74.8% of cervical cancer cases[42]. Additionally, high-risk HPV types can also cause vaginal, vulvar, and anal cancers[43-46]; and (2) Low-risk HPV types, including HPV 6, 11, 40, 42, 43, 44, 54, 61, and 70, primarily cause benign conditions such as genital warts (condyloma acuminatum)41. These types are generally not associated with malignant transformations such as cervical cancer. HPV 6 and 11 are the predominant types responsible for genital warts[30,31].
HPV genotyping plays a critical role in the prevention and screening of cervical cancer. This facilitates the identification of high-risk populations and provides valuable guidance for clinical diagnosis, treatment, and management strategies. By distinguishing between high- and low-risk HPV types, healthcare providers can tailor interventions to mitigate disease progression and improve patient outcomes.
Recent studies and global statistics have revealed significant variations in HPV infection rates across different regions, reflecting the complex interplay of socioeconomic, geographic, and healthcare factors. According to a meta-analysis of 194 global studies, the overall HPV infection rate among women with normal cervical cytology was approximately 11.7% (95%CI: 11.6%-11.7%)[29,30]. However, regional disparities are striking. The Caribbean exhibits the highest HPV infection rate globally, at 35.4% (95%CI: 29.0%-42.2%), followed closely by East Africa at 33.6% (95%CI: 30.2%-37.1%)[47,48]. Sub-Saharan Africa also reports a high prevalence of 24.0% (95%CI: 23.1%-25.0%), underscoring the disproportionate burden of HPV-related diseases in low-resource settings[49]. Eastern Europe shows a moderate but still elevated rate of 21.4% (95%CI: 20.1%-22.7%)[50]. In contrast, more developed regions such as North America and West Asia exhibit significantly lower HPV infection rates, reflecting better access to preventive healthcare, vaccination programs, and cervical cancer screening initiatives[51,52].
These regional disparities highlight the urgent need for targeted interventions, particularly in resource-limited areas, where HPV infection rates remain alarmingly high.
The age distribution of HPV infections provides critical insights into the dynamics of HPV transmission and persistence. HPV infection rates peak among young women under the age of 25 years, driven by high-risk sexual behaviors and limited prior exposure to the virus. This age group consistently exhibits the highest prevalence of HPV infection worldwide[53,54]. After middle age, HPV infection rates tend to decline owing to acquired immunity from prior exposure. However, in certain regions, such as Africa, a second peak in HPV infection rates is observed among older women, likely due to persistent infections or reactivation of latent HPV. A similar secondary increase, albeit smaller, has been noted in Asia and the Americas[53,55]. This bimodal distribution underscores the importance of age-specific prevention strategies, including vaccination in adolescents and regular screening in older women.
The top 10 most common HPV infection types worldwide are: HPV16 (2.8%), HPV52 (1.5%), HPV31 (1.2%), HPV53 (1.2%), HPV18 (1.1%), HPV51 (1.1%), HPV58 (1.0%), HPV39 (0.9%), HPV66 (0.9%), and HPV70 (0.8%)[16,56,57]. A meta-analysis of 115789 HPV-positive women showed that 76% of the women with low-grade cervical lesions and 85% of the women with high-grade cervical lesions were infected with HPV. In cervical lesions and cervical cancer, HPV16/18/31/33/45/52/58 were the most common types[58]. An epidemiological study of HPV infection in 1.7 million women in China revealed that the overall prevalence of HPV among Chinese women was 15.54% (95%CI: 13.83-17.24). The top five common HPV infection types in the general population were HPV16, 52, 58, 18, and 33[59]. In China, HPV16/18 causes approximately 69% of cervical cancer cases, whereas HPV types 31/33/45/52/58 account for approximately 23% of cervical cancer cases. HPV16/52/58/18 were the predominant types at all disease stages. In cervical squamous cell carcinoma, HPV16/18/31/52/58 were the most common types, with infection rates of 76.7%, 7.8%, 3.2%, 2.2%, and 2.2%, respectively[18,60-62].
Dominant types: HPV16 and 18 are the most common high-risk types globally, accounting for 70%-80% of cervical cancer cases[63,64]. HPV31, 33, 45, 52, and 58 also contribute significantly to cervical cancer. However, the prevalence varies by region. Collectively, these types are considered secondary drivers of HPV-related cancers[58,65].
Region-specific infection characteristics: Sub-Saharan Africa[49,52] has highest rate globally, averaging 24%-35%, with some countries reaching up to 40%. HPV16 and 18 are predominantly seen in cervical cancer cases. HPV35 is more prevalent in this region, potentially due to unique genetic or immunological factors. Other high-risk types, HPV52 and 58, are also frequently detected. The high infection rates are associated with low vaccine coverage. The prevalence of HPV35 may be linked to genetic susceptibility in African populations.
In Asia (including China, India, and Southeast Asia)[52], infection rate averages 15%-20%, with higher rates in rural parts of China and India. HPV16 and 18 are responsible for 69%-70% of cases of cervical cancer. HPV52 and 58 are particularly common in Asian populations, particularly in younger women. HPV33 and 45 are also detected, but at lower frequencies. The high prevalence of HPV52 and 58 may be related to genetic predisposition and sexual behavior patterns in Asian populations. In regions with low vaccine coverage (e.g., India), non-vaccine-covered types remain a significant concern.
In Latin America and the Caribbean[66], infection rate averages 15%-20%, with higher rates in the Caribbean (up to 35%). HPV16 and 18 are dominant in cervical cancer cases. HPV52 and 58 are frequently detected, particularly in younger women. HPV31 and 33 are also prevalent. The high prevalence of HPV52 and 58 is similar to that observed in Asia, possibly because of shared cultural and social behaviors. In the Caribbean, high infection rates are associated with multiple sexual partners and low vaccine coverage.
In North America and Western Europe[67,68], infection rate is the lowest globally, averaging 5%-10%. HPV16 and 18 are still dominant but significantly reduced due to vaccination. HPV31 and 33 are emerging as more common types in vaccinated populations. HPV45 and 52 were detected but contributed less frequently. High vaccination and screening coverage significantly reduced HPV16 and 18 infections. Non-vaccine-covered types (e.g., HPV31 and 33) are now a growing focus of public health efforts.
In Eastern Europe and Central Asia[9,58,69], infection rate averages 21.4%, which is higher than in Western Europe and North America. HPV16 and 18 are predominant in cervical cancer cases. HPV31 and 52 are commonly detected, es
In Australia[69-72], infection rate is low, averaging 5%-8%. HPV16 and 18 were significantly reduced following widespread vaccination. HPV31 and 33 are currently the most commonly detected high-risk types. National vaccination programs and regular screening systems can effectively control HPV transmissions. The incidence of cervical cancer is nearing its elimination target.
The global burden of HPV infection remains substantial with significant regional and demographic variations. High-prevalence regions, such as sub-Saharan Africa and the Caribbean, face disproportionate challenges, while developed regions benefit from robust healthcare infrastructure and vaccination programs. Understanding age-specific distribution and dominant HPV types is crucial for tailoring prevention and intervention strategies. Continued efforts to expand vaccine coverage, improve screening programs, and address socioeconomic barriers are essential to reduce the global burden of HPV-related diseases and achieve the goal of cervical cancer elimination. Summary of global human papillomavirus infection characteristics as below (Table 1).
Sub-Saharan Africa | 24%-40% | HPV16, 18, 35, 52, 58 | High HPV35 prevalence, genetic susceptibility, and limited healthcare resources |
Asia | 15%-20% | HPV16, 18, 52, 58, 33 | High HPV52 and 58 prevalence; genetic and behavioral factors; low vaccine coverage in some areas |
Latin America & Caribbean | 15%-35% | HPV16, 18, 52, 58, 31 | High HPV52 and 58 prevalence; multiple sexual partners; low vaccine coverage in the Caribbean |
North America & Western Europe | 5%-10% | HPV16, 18, 31, 33 | High vaccination coverage and non-vaccine-covered types (e.g., HPV31, 33) are emerging as new concerns |
Eastern Europe & Central Asia | 21.40% | HPV16, 18, 31, 52 | Insufficient healthcare resources and changing sexual behaviors increase transmission risk |
Australia | 5%-8% | HPV16, 18, 31, 33 | High vaccination and screening coverage; incidence of cervical cancer near elimination |
The diagnosis of cervical cancer is a multistep process, which can be divided into screening, pathologic diagnosis, staging, and treatment. Through early screening and accurate diagnosis, the cure rate can be significantly improved and the mortality rate reduced. Medical resources and technology in different regions may affect specific implementation, but the process described above is a standard framework widely adopted worldwide[73-75]. The standard diagnostic procedure is as follows.
Screening is a critical step in the early detection of cervical cancer and its precursors such as CIN. The primary goal was to identify individuals at a high risk of developing symptoms. According to guidelines from the American Cancer Society, cervical cancer screening is recommended to start at age 25 years. In women aged 25-65 years, HPV DNA testing is preferred every 5 years. If HPV testing is unavailable, a combination of Pap smear and HPV testing can be performed every 5 years. In women aged 21-24 years, a standalone cytological test (Pap smear) is recommended every 3 years. Further evaluation is required if abnormalities are detected (e.g., atypical squamous cells, low-grade squamous intraepithelial lesions, high-grade squamous intraepithelial lesions, or a positive HPV test).
For women with abnormal screening results, additional testing was performed to confirm the presence of precancerous lesions or cancer. Colposcopy was used to magnify the cervix and identify the abnormal areas. Biopsies of the suspicious areas may be performed during this procedure. Tissue samples were collected from the cervix and examined under a microscope by a pathologist to detect signs of cancer. Common biopsy methods include the following: (1) Puncture biopsy: A small piece of tissue is removed using a sharp hollow instrument; (2) Curettage: A curette is used to collect the cells or tissue from the cervical canal; (3) Loop electrosurgical excision procedure (LEEP): A thin wire loop is used to remove cervical tissue for diagnostic or therapeutic purposes, particularly early lesions; and (4) Cone biopsy: A large, cone-shaped piece of tissue was removed from the cervix and cervical canal to diagnose and treat high-grade lesions.
Pathological diagnosis is a cornerstone for confirming the presence and nature of cervical cancer. This step involves microscopic examination of biopsy samples to determine the type and extent of the lesion. Pathological diagnosis identifies the presence of cervical cancer and classifies its type (e.g., squamous cell carcinoma or adenocarcinoma). If cervical cancer is confirmed, staging is performed to assess its extent and spread. Staging methods include imaging techniques, such as positron emission tomography–computed tomography, computed tomography, and magnetic resonance imaging.
Based on pathological diagnosis, heterogeneity of the cervical cancer microenvironment, and staging results, an individualized treatment plan can be developed. Personalized treatment strategies are formulated based on factors such as stage and type of cervical cancer, patient age, and overall health status. Common treatment modalities include surgery, radiotherapy, chemotherapy, and patient-derived xenograft model[76]. Regular follow-up is essential after treatment to monitor disease progression and evaluate the effectiveness of the intervention.
Vaccination is the most effective strategy for preventing HPV infection and associated diseases. Currently, three primary HPV vaccines have been approved worldwide; all designed to provide preventive protection against high-risk HPV infections.
Bivalent vaccine (Cervarix): Targets HPV16 and HPV18, which account for approximately 70% of cervical cancer cases. Quadrivalent vaccine (Gardasil): Targets HPV16, HPV18, HPV6, and HPV11 and offers protection against cervical cancer and reduces the incidence of genital warts. Nine-valent vaccine (Gardasil 9): Targets HPV6, 11, 16, 18, 31, 33, 45, 52, and 58, extending coverage to approximately 90% of cervical cancer cases.
These vaccines demonstrate exceptional efficacy in protecting against their targeted HPV types, with protection rates reaching 100% in certain populations. For instance, a subgroup analysis of East Asian women aged 16-26 years in the PPE population revealed that the nine-valent HPV vaccine provided 100% protection against CIN1+ lesions associated with HPV31/33/45/52/58 (95%CI: 33.5%-100%)[77]. Clinical trials have shown that the quadrivalent HPV vaccine is equally effective, offering 100% protection against CIN2+ lesions in women aged 15-26 years who are not infected with the vaccine-targeted HPV types[78,79]. Even individuals that are already infected with HPV can benefit from vaccination. A retrospective analysis of three global multicenter phase 3 clinical trials demonstrated that the nine-valent HPV vaccine significantly reduced the disease burden associated with non-infected HPV types[79,80].
Despite these advancements, current HPV vaccines do not provide complete protection against all HPV types that cause cervical cancer. Other HPV strains can also lead to cervical cancer, beyond the nine high-risk types covered by existing vaccines. Therefore, the development of vaccines that target a broader range of HPV types is imperative. Although the development and deployment of HPV vaccines have achieved remarkable success, becoming a cornerstone in the prevention of cervical cancer and other HPV-related diseases, global vaccination rates remain uneven. Substantial disparities in vaccination rates persist among countries. High-income countries tend to achieve higher coverage rates, whereas LMICs often face challenges such as resource shortages and cultural barriers that continue to pose significant challenges[9].
Expanding HPV vaccine coverage will require robust policy support and technological innovation. Key priorities include promoting male vaccination, advancing the development of novel vaccines (such as therapeutic and polyvalent vaccines), and enhancing public education. With ongoing advancements in vaccine technology and strengthened global collaboration, HPV vaccines hold the potential to further reduce the burden of HPV-related diseases and contribute to the ambitious goal of eliminating cervical cancer worldwide[81].
In the 1940s, Greek physician Papanicolaou and Traut[82] pioneered the Pap smear method, a groundbreaking technique that enables doctors to detect early cellular changes in the cervix through microscopic examination of cervical smears. The widespread implementation of this method has played a pivotal role in reducing global cervical cancer mortality rates by 50%-70%[83]. The primary advantage of the Pap smear is its low cost, which has made it accessible and effective for lowering cervical cancer mortality over the past few decades. However, this method is not without its limitations. Key disadvantages include the need for professional expertise and specialized equipment, low sensitivity and specificity, high risk of false positives and false negatives, and significant subjectivity in result interpretation. These drawbacks can lead to misdiagnosis or missed diagnosis, underscoring the importance of skilled technicians and careful evaluation[84-87]. The cost of a Pap smear typically ranges from $30 to $300 (without insurance) depending on the healthcare setting and region. Although it remains a valuable tool in cervical cancer screening, its limitations highlight the need for complementary or alternative methods, particularly in resource-limited settings[88,89].
In the 1990s, visual inspection with acetic acid/iodine staining (VIA/VILI) became widely adopted in developing countries for cervical cancer screening. This technique involves application of a chemical solution to the cervix and direct observation of the epithelial reaction with the naked eye. Its key advantages include independence from specialized equipment, ease of operation, and low cost, making it particularly suitable for resource-limited settings. However, it has notable drawbacks, such as a high false-positive rate and significant susceptibility to operator subjectivity[90-92]. It costs $10–$50 (without insurance)[88].
Liquid-based cytology assay (TCT) is an advanced version of the traditional Pap smear technique. Emerging in the late 20th century as one of a new generation of cytological methods, it was first approved for the United States market in 1996[87]. In this procedure, doctors use a specialized brush to collect the shed cervical cells, which are then rinsed into a small bottle containing a cell preservation solution. The samples undergo centrifugation, slide preparation, and staining, allowing high-quality images to be observed under a microscope. Compared with traditional Pap smears, TCT offers higher sensitivity, reduces the risk of sample contamination, and significantly lowers the false-negative rate. These advantages have led to their rapid adoption in the developed regions worldwide. However, this method is expensive and requires specialized laboratories[93,94]. It costs $100–$400 (without insurance)[95,96].
In the 1970s, German scientist zur Hausen[97] made a groundbreaking discovery by identifying the association between cervical cancer and HPV infection. This pivotal finding laid the foundation for further research on the role of HPV in cervical carcinogenesis. By the late 1990s, with advances in the understanding of the relationship between HPV and cervical cancer, HPV testing began to be incorporated into cervical cancer screening programs. In 1999, the United States FDA approved the HC2 test, which became the gold standard for HPV testing. This marked a significant shift in cervical cancer screening, transitioning from reliance on cytology alone to an integrated approach that combines cytology with nucleic acid testing[22,98]. In the early 21st century, HPV testing emerged as the preferred method for cervical cancer screening owing to its high sensitivity and strong negative predictive value[99]. This method involves sequencing to detect the type of HPV infection, thereby enabling the identification of high-risk HPV types associated with cervical cancer[100]. The key advantages of HPV testing include its ability to extend the screening interval (e.g., every 5 years), reducing the frequency of testing, and conserving healthcare resources[101]. However, this method is not without its limitations. It is expensive and there is a potential risk of overdiagnosis in low-risk populations, which could lead to unnecessary follow-up procedures[102]. It costs $50-$200 (without insurance)[103-108].
Combined screening, which integrates cytology and HPV testing, is the primary method used in regions with sufficient healthcare resources[85]. This approach leverages the strengths of both tests, thereby significantly improving accuracy and efficiency. For instance, one study demonstrated that combining cervical TCT with HPV-DNA testing achieved a sensitivity of 99.8%, which was markedly higher than HPV-DNA testing alone (72.0%) or TCT testing alone (75.4%)[100]. Owing to the high accuracy of combined testing, the screening interval can be appropriately extended. The American Cancer Society 2020 guidelines recommend primary HPV screening or combined cytology and HPV testing every 5 years[75,109,110]. Extending the screening interval reduces the frequency of testing, lowers costs, and conserves medical resources[95,96,111-117]. Although combined testing is expensive and requires more resources and technical support, its high sensitivity and extended screening intervals help reduce missed diagnoses, ultimately lowering follow-up treatment costs and health risks for patients[118]. It costs $150-$600 (without insurance).
HPV self-sampling allows women to collect samples at home for HPV testing, eliminating the need to visit a hospital for sample collection by a physician. This method is particularly beneficial for women in remote areas, regions with limited transportation, and areas with constrained medical resources, as it can significantly enhance the coverage of cervical cancer screening. For women who may feel shy or embarrassed about undergoing cervical sampling at a healthcare facility, self-sampling offers a private and convenient alternative that can be completed within the comfort of their home environment[41]. This approach not only protects privacy but also improves patient compliance by reducing discomfort and stigma. Additionally, self-sampling is simple and cost-effective, reducing both the time and financial burden associated with hospital visits, thereby encouraging more women to participate in regular cervical cancer screening[29]. Self-sampling also aligns with global health goals, contributing to the World Health Organization target of achieving 70% cervical cancer screening coverage by 2030. However, there are some limitations to consider[101]. The quality of self-collected samples may not always be as consistent or reliable as those collected by trained healthcare professionals, which could impact the test accuracy[119]. To mitigate this issue, robust detection technologies and clear instructions for proper sample collection are essential. It costs $50-$200, depending on the testing method and geographic location[95].
Cone biopsy involves removal of cervical tissue for pathological diagnosis[120]. Its primary advantage is that it can simultaneously serve both diagnostic and therapeutic purposes, particularly in high-grade lesions[121]. However, the procedure is invasive, costly, and may lead to complications such as bleeding or infection[122]. It costs $500-$2000 (without insurance)[95].
LEEP utilizes a thin wire loop to remove cervical tissue. Its main advantage is its dual functionality, as it can be used for both diagnosing and treating early-stage lesions[123]. The disadvantages include relatively high costs and potential for minor complications[122]. It costs $300-$1000 (without insurance)[95]. The costs associated with each screening method are summarized in the table below (Table 2).
Pap smear | $30-$300 |
VIA/VILI | $10-$50 |
TCT | $100-$400 |
HPV DNA testing | $50-$200 |
Combined screening | $150-$600 |
Self-sampling | $50-$200 |
LEEP | $300-$1000 |
Cone biopsy | $500-$2000 |
Pap Smear | $30-$300 |
VIA/VILI | $10-$50 |
TCT | $100-$400 |
High-income countries, with their robust healthcare infrastructure and financial resources, predominantly adopt advanced screening tools such as HPV DNA testing or combined screening (cytology and HPV testing)[101]. These methods are favored because of their high sensitivity, specificity, and ability to extend the screening interval, thereby reducing the frequency of testing while maintaining effectiveness[96]. The United States introduced HPV testing in 2012, and HPV DNA testing is now the primary recommended method for cervical cancer screening[124]. Women are advised to undergo HPV testing every 5 years starting at age 30 years. If HPV testing is unavailable, combined screening (cytology + HPV testing) is an alternative option[74]. The United Kingdom adopted HPV testing as the preferred screening method in 2019. Women are recommended to undergo HPV testing every 5 years starting at age 25 years[125]. Australia introduced HPV testing as the preferred screening method in 2017[81]. Women are advised to undergo HPV testing every 5 years starting at age 25 years. These countries leverage their economic and medical resources to implement large-scale HPV DNA testing programs, ensuring high screening coverage and early detection of cervical cancer. The extended screening intervals (e.g., every 5 years) also reduce healthcare costs and improve efficiency without compromising accuracy[95].
Middle-income countries often face economic constraints, leading them to rely on low-cost traditional screening methods such as cytology or VIA/VILI. However, in certain regions, there is a gradual shift toward introducing HPV testing where feasible[126]. In China[127], currently, there is no nationwide systematic screening program. However, some regions have implemented localized screening initiatives. For example, in Changsha city, HPV testing is used as the primary screening method, supplemented by cytological examination55. In Brazil[128,129], women aged 30-64 years are recommended to undergo cytological examinations every 3 years. HPV testing is offered as an additional measure for high-risk groups. Self-sampling has also been piloted in select areas[130]. Middle-income countries generally favor traditional screening tools (e.g., cytology or VIA/VILI) due to their affordability and simplicity. Where economic conditions permit, HPV testing is gradually being introduced to improve screening accuracy and coverage. Significant regional disparities exist, with some areas advancing screening technologies through pilot projects or targeted initiatives. These efforts aim to bridge gaps in healthcare access and enhance early detection of cervical cancer.
Low-income countries face significant economic and resource constraints, leading them to rely primarily on low-cost screening tools such as visual inspection with VIA/VILI. While HPV testing is considered the ideal method for cervical cancer screening, its practical application remains limited in these regions due to financial, technological, and infrastructural barriers[67,131-136]. In Kenya, HPV testing is recommended as the preferred screening method. However, due to economic and resource limitations, acetic acid staining for gross visual observation (VIA) remains the primary screening approach[137]. In Uganda, HPV testing is recommended, but VIA/VILI is still widely used in practice due to its affordability and simplicity[138]. In low-income countries, VIA/VILI is favored because it is inexpensive, easy to perform, and does not require advanced equipment or laboratory facilities. However, this method has significant drawbacks, including low sensitivity and a high risk of false-positive and false-negative results, which can compromise the effectiveness of screening programs. The implementation of HPV testing is hindered by challenges such as the lack of necessary equipment, trained personnel, and sustainable funding. Despite these limitations, international organizations are playing a crucial role in promoting and supporting the adoption of HPV testing in these regions.
Economic resources: High-income countries can afford large-scale implementation of advanced screening methods like HPV DNA testing, ensuring high coverage and accuracy[95]. In contrast, LMICs face significant financial constraints, limiting their ability to adopt costly technologies and restricting them to simpler, less effective methods such as VIA/VILI[139].
Medical infrastructure: The availability of equipment, laboratory facilities, and trained healthcare professionals plays a crucial role in determining the feasibility of specific screening methods. Regions with well-established medical in
Policy support: Government funding and international collaborations are essential for scaling up cervical cancer screening programs. Financial backing from global health organizations can help bridge gaps in resource-limited settings, enabling the adoption of more accurate and cost-effective screening technologies[140].
Innovations such as self-sampling techniques and portable HPV testing devices are emerging as game-changers for improving screening coverage in resource-limited areas. These methods address barriers like accessibility, affordability, and privacy concerns, making it easier for women to participate in screening programs.
Partnerships between governments, nonprofit organizations, and international organizations are critical for expanding access to advanced screening technologies. By pooling resources and expertise, these collaborations can help overcome economic and infrastructural challenges in low- and middle-income regions[140]. Addressing disparities in cervical cancer screening requires a multifaceted approach that considers economic resources, medical infrastructure, and policy support. Leveraging technological advancements and fostering international cooperation will be key to reducing the global burden of cervical cancer, particularly in underserved populations.
The choice of an effective screening strategy should consider the following factors: (1) Medical resources: Availability of professional equipment, trained personnel, and laboratory facilities. High-income regions can support advanced methods like HPV DNA testing, while low-resource areas may prioritize simpler, cost-effective techniques like VIA/VILI; (2) Economic conditions: Affordability of screening programs is critical. Low-cost methods are essential in economically disadvantaged regions, while wealthier nations can invest in more expensive but accurate technologies; (3) Demographic characteristics: considerations such as the age distribution, health status, and cultural preferences of the target population play a vital role. For instance, self-sampling may be more acceptable in populations where privacy concerns or stigma around cervical cancer screening exist; and (4) Policy support: Governmental and international financial backing is crucial for implementing large-scale screening programs. Partnerships with global health organizations can help bridge gaps in resource-limited areas.
By optimizing cervical cancer screening strategies based on available resources, economic conditions, and demographic needs, it is possible to significantly enhance early detection rates and reduce the global burden of cervical cancer. Tailored approaches that integrate innovative technologies, such as self-sampling and portable HPV tests, hold great promise for improving outcomes, particularly in underserved regions.
Cervical cancer, a paradigmatic manifestation of HPV-associated diseases, remains a critical global health challenge marked by pronounced regional disparities and inequities in technological access. Through comprehensive evaluation of HPV epidemiology, advancements in screening modalities, and vaccine implementation landscapes, this study highlights the geographically heterogeneous distribution of high-risk HPV genotypes-exemplified by the predominance of HPV35 in African populations and HPV52/58 in Asian cohorts-as well as age-stratified infection dynamics, characterized by peak prevalence in young women (< 25 years) and emergent reinfection risks among older age groups. Current screening paradigms demonstrate pronounced resource dependency: high-income nations utilize HPV DNA testing and co-testing (cytology + HPV detection) to achieve precision prevention, whereas low- and middle-income regions, constrained by economic and infrastructural limitations, predominantly rely on visual inspection with VIA/VILI or conventional cytology, resulting in stark disparities in screening efficacy and population coverage.
Although existing prophylactic vaccines (bivalent, quadrivalent, and nonavalent) prevent approximately 90% of cervical cancers, their global uptake remains inequitable, with coverage rates in resource-limited settings lagging significantly behind high-income regions. Persistent challenges-including incomplete protection against all oncogenic subtypes, cultural barriers to vaccination programs, and cost-prohibitive pricing-underscore the urgent need for next-generation broad-spectrum vaccines and optimized immunization strategies tailored to diverse sociocultural contexts.
To overcome these barriers, multi-faceted approaches are imperative: Technological innovations such as point-of-care HPV diagnostics, artificial intelligence-driven cytological analysis, and refined self-sampling methodologies must be prioritized to enhance accessibility; international collaborations should focus on resource redistribution and technology transfer to under-resourced regions; and region-specific screening protocols must be developed, integrating local epi
1. | Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5690] [Cited by in RCA: 6017] [Article Influence: 6017.0] [Reference Citation Analysis (1)] |
2. | Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 53206] [Cited by in RCA: 55248] [Article Influence: 7892.6] [Reference Citation Analysis (125)] |
3. | zur Hausen H. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer. 2002;2:342-350. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2728] [Cited by in RCA: 2757] [Article Influence: 119.9] [Reference Citation Analysis (0)] |
4. | Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley MA. The biology and life-cycle of human papillomaviruses. Vaccine. 2012;30 Suppl 5:F55-F70. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 928] [Cited by in RCA: 913] [Article Influence: 70.2] [Reference Citation Analysis (0)] |
5. | Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, Bray F. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Glob Health. 2020;8:e191-e203. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1944] [Cited by in RCA: 2216] [Article Influence: 443.2] [Reference Citation Analysis (0)] |
6. | de Thurah L, Bonde J, Lam JUH, Rebolj M. Not all HPV nucleic acid tests are equal: only those calibrated to detect high grade lesions matter for cervical screening: Response to 'Concordant testing results between various human papillomavirus assays in primary cervical cancer screening: systematic review' by de Thurah, Bonde, Uyen, Lam and Rebolj. Published 27 May, 2017. Clin Microbiol Infect. 2018;24:438-439. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 4] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
7. | World Health Organization. WHO guideline for screening and treatment of cervical pre-cancer lesions for cervical cancer prevention, second edition. 2021. Available from: https://www.who.int/publications/i/item/9789240030824. |
8. | Drolet M, Bénard É, Pérez N, Brisson M; HPV Vaccination Impact Study Group. Population-level impact and herd effects following the introduction of human papillomavirus vaccination programmes: updated systematic review and meta-analysis. Lancet. 2019;394:497-509. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 481] [Cited by in RCA: 625] [Article Influence: 104.2] [Reference Citation Analysis (0)] |
9. | Bruni L, Diaz M, Barrionuevo-Rosas L, Herrero R, Bray F, Bosch FX, de Sanjosé S, Castellsagué X. Global estimates of human papillomavirus vaccination coverage by region and income level: a pooled analysis. Lancet Glob Health. 2016;4:e453-e463. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 545] [Cited by in RCA: 563] [Article Influence: 62.6] [Reference Citation Analysis (0)] |
10. | Brisson M, Kim JJ, Canfell K, Drolet M, Gingras G, Burger EA, Martin D, Simms KT, Bénard É, Boily MC, Sy S, Regan C, Keane A, Caruana M, Nguyen DTN, Smith MA, Laprise JF, Jit M, Alary M, Bray F, Fidarova E, Elsheikh F, Bloem PJN, Broutet N, Hutubessy R. Impact of HPV vaccination and cervical screening on cervical cancer elimination: a comparative modelling analysis in 78 low-income and lower-middle-income countries. Lancet. 2020;395:575-590. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 293] [Cited by in RCA: 465] [Article Influence: 93.0] [Reference Citation Analysis (0)] |
11. | Spayne J, Hesketh T. Estimate of global human papillomavirus vaccination coverage: analysis of country-level indicators. BMJ Open. 2021;11:e052016. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 65] [Article Influence: 16.3] [Reference Citation Analysis (0)] |
12. | Zhang J, Dong Z, Xu L, Han X, Sheng Z, Chen W, Zheng J, Lai D, Shen F. An Injection Molded SlipChip with Self-Sampling for Integrated Point-of-Care Testing of Human Papilloma Virus. Adv Sci (Weinh). 2024;11:e2406367. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
13. | Einstein MH, Zhou N, Gabor L, Sahasrabuddhe VV. Primary Human Papillomavirus Testing and Other New Technologies for Cervical Cancer Screening. Obstet Gynecol. 2023;142:1036-1043. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
14. | Meza Ramirez CA, Greenop M, Almoshawah YA, Martin Hirsch PL, Rehman IU. Advancing cervical cancer diagnosis and screening with spectroscopy and machine learning. Expert Rev Mol Diagn. 2023;23:375-390. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
15. | Fu L, Xia W, Shi W, Cao GX, Ruan YT, Zhao XY, Liu M, Niu SM, Li F, Gao X. Deep learning based cervical screening by the cross-modal integration of colposcopy, cytology, and HPV test. Int J Med Inform. 2022;159:104675. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 13] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
16. | Brianti P, De Flammineis E, Mercuri SR. Review of HPV-related diseases and cancers. New Microbiol. 2017;40:80-85. [PubMed] |
17. | Moody CA, Laimins LA. Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer. 2010;10:550-560. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1120] [Cited by in RCA: 1196] [Article Influence: 79.7] [Reference Citation Analysis (0)] |
18. | Chen W, Zhang X, Molijn A, Jenkins D, Shi JF, Quint W, Schmidt JE, Wang P, Liu YL, Li LK, Shi H, Liu JH, Xie X, Niyazi M, Yang P, Wei LH, Li LY, Li J, Liu JF, Zhou Q, Hong Y, Li L, Li Q, Zhou HL, Bian ML, Chen J, Qiao YL, Smith JS. Human papillomavirus type-distribution in cervical cancer in China: the importance of HPV 16 and 18. Cancer Causes Control. 2009;20:1705-1713. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 71] [Cited by in RCA: 81] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
19. | Cho EH, Park MS, Woo HY, Park H, Kwon MJ. Evaluation of clinical usefulness of HPV-16 and HPV-18 genotyping for cervical cancer screening. J Gynecol Oncol. 2024;35:e72. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
20. | McBride AA, White EA. HPV Integration Can Drive the Formation of Virus-Host Extrachromosomal DNA in Tumors. Cancer Discov. 2023;13:814-816. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Reference Citation Analysis (0)] |
21. | Zhou L, Qiu Q, Zhou Q, Li J, Yu M, Li K, Xu L, Ke X, Xu H, Lu B, Wang H, Lu W, Liu P, Lu Y. Long-read sequencing unveils high-resolution HPV integration and its oncogenic progression in cervical cancer. Nat Commun. 2022;13:2563. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 59] [Article Influence: 19.7] [Reference Citation Analysis (0)] |
22. | Schiffman M, Castle PE, Jeronimo J, Rodriguez AC, Wacholder S. Human papillomavirus and cervical cancer. Lancet. 2007;370:890-907. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1829] [Cited by in RCA: 1863] [Article Influence: 103.5] [Reference Citation Analysis (0)] |
23. | Zhang Y, Liu Y, Xing X, Liu H, Guan W. Genome-Wide Analysis of p53 Targets Reveals SCN2A as a Novel Player in p53-Induced Cell Arrest in HPV-Positive Cells. Viruses. 2024;16. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
24. | Marur S, D'Souza G, Westra WH, Forastiere AA. HPV-associated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol. 2010;11:781-789. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1340] [Cited by in RCA: 1332] [Article Influence: 88.8] [Reference Citation Analysis (0)] |
25. | Wiest T, Schwarz E, Enders C, Flechtenmacher C, Bosch FX. Involvement of intact HPV16 E6/E7 gene expression in head and neck cancers with unaltered p53 status and perturbed pRb cell cycle control. Oncogene. 2002;21:1510-1517. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 269] [Cited by in RCA: 261] [Article Influence: 11.3] [Reference Citation Analysis (0)] |
26. | Scarth JA, Patterson MR, Morgan EL, Macdonald A. The human papillomavirus oncoproteins: a review of the host pathways targeted on the road to transformation. J Gen Virol. 2021;102. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 83] [Cited by in RCA: 109] [Article Influence: 27.3] [Reference Citation Analysis (0)] |
27. | Ates D, Sahin EN, Katipoglu K, Usubutun A. A Low-Risk HPV-Associated Well-Differentiated Squamous Cell Carcinoma of the Cervix with Low-Grade Squamous Intraepithelial Lesion Morphology: Clinical and Pathologic Diagnostic Difficulties and Review of the Literature. Turk Patoloji Derg. 2024;40:196-201. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
28. | Gillis JL, Grennan T, Grewal R, Ogilvie G, Gaspar M, Grace D, Lofters A, Raboud JM, Saarela O, MacPherson P, Rosenes R, Salit IE, Burchell AN; OHTN Cohort Study Team. Low human papillomavirus (HPV) knowledge related to low risk perception among men living with HIV: Implications for HPV-associated disease prevention. Prev Med. 2020;141:106274. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 4] [Article Influence: 0.8] [Reference Citation Analysis (0)] |
29. | Pretsch PK, Spees LP, Brewer NT, Hudgens MG, Sanusi B, Rohner E, Miller E, Jackson SL, Barclay L, Carter A, Wheeler SB, Smith JS. Effect of HPV self-collection kits on cervical cancer screening uptake among under-screened women from low-income US backgrounds (MBMT-3): a phase 3, open-label, randomised controlled trial. Lancet Public Health. 2023;8:e411-e421. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 30] [Cited by in RCA: 23] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
30. | Egawa N, Doorbar J. The low-risk papillomaviruses. Virus Res. 2017;231:119-127. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 133] [Cited by in RCA: 155] [Article Influence: 17.2] [Reference Citation Analysis (0)] |
31. | Guimerà N, Lloveras B, Lindeman J, Alemany L, van de Sandt M, Alejo M, Hernandez-Suarez G, Bravo IG, Molijn A, Jenkins D, Cubilla A, Muñoz N, de Sanjose S, Bosch FX, Quint W; RIS HPV TTHPV VVAPO study groups. The occasional role of low-risk human papillomaviruses 6, 11, 42, 44, and 70 in anogenital carcinoma defined by laser capture microdissection/PCR methodology: results from a global study. Am J Surg Pathol. 2013;37:1299-1310. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 70] [Cited by in RCA: 75] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
32. | Schiffman M, Doorbar J, Wentzensen N, de Sanjosé S, Fakhry C, Monk BJ, Stanley MA, Franceschi S. Carcinogenic human papillomavirus infection. Nat Rev Dis Primers. 2016;2:16086. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 568] [Cited by in RCA: 593] [Article Influence: 65.9] [Reference Citation Analysis (0)] |
33. | Xiao X, Cao Y, Bi K, Wang W, Yang J, Wang J, Li Y, Li C, Guan R, Zhang Y, Wang J, Song S, Zhu L, Shi H. The Triaging Effect of the Human Papillomavirus 16/18 E7 Oncoprotein Assay in HPV 16/18-Positive Patients for High-Grade Cervical Intraepithelial Neoplasia Screening: A Cross-Sectional Study. J Womens Health (Larchmt). 2023;32:1136-1141. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
34. | Bertagnin C, Messa L, Pavan M, Celegato M, Sturlese M, Mercorelli B, Moro S, Loregian A. A small molecule targeting the interaction between human papillomavirus E7 oncoprotein and cellular phosphatase PTPN14 exerts antitumoral activity in cervical cancer cells. Cancer Lett. 2023;571:216331. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
35. | Hidayatullah A, Putra WE, Sustiprijatno S, Rifa'i M, Widiastuti D, Heikal MF, Permatasari GW. Concatenation of molecular docking and dynamics simulation of human papillomavirus type 16 E7 oncoprotein targeted ligands: In quest of cervical cancer's treatment. An Acad Bras Cienc. 2023;95:e20220633. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
36. | Lizano M, Berumen J, García-Carrancá A. HPV-related carcinogenesis: basic concepts, viral types and variants. Arch Med Res. 2009;40:428-434. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 45] [Cited by in RCA: 45] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
37. | Singh D, Vignat J, Lorenzoni V, Eslahi M, Ginsburg O, Lauby-Secretan B, Arbyn M, Basu P, Bray F, Vaccarella S. Global estimates of incidence and mortality of cervical cancer in 2020: a baseline analysis of the WHO Global Cervical Cancer Elimination Initiative. Lancet Glob Health. 2023;11:e197-e206. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 409] [Cited by in RCA: 597] [Article Influence: 298.5] [Reference Citation Analysis (0)] |
38. | Ntova CK, Kottaridi C, Chranioti A, Spathis A, Kassanos D, Paraskevaidis E, Karakitsos P. Genetic variability and phylogeny of high risk HPV type 16, 18, 31, 33 and 45 L1 gene in Greek women. Int J Mol Sci. 2012;13:1-17. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 19] [Cited by in RCA: 19] [Article Influence: 1.4] [Reference Citation Analysis (0)] |
39. | Bertuccio MP, Spataro P, Caruso C, Picerno I. Detection of human papillomavirus E6/E7 mRNA in women with high-risk HPV types 16, 18, 31, 33 and 45 which are associated with the development of human cervical cancer. Eur J Gynaecol Oncol. 2011;32:62-64. [PubMed] |
40. | Raiol T, Wyant PS, de Amorim RM, Cerqueira DM, Milanezi Nv, Brígido Mde M, Sichero L, Martins CR. Genetic variability and phylogeny of the high-risk HPV-31, -33, -35, -52, and -58 in central Brazil. J Med Virol. 2009;81:685-692. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 40] [Cited by in RCA: 41] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
41. | Winer RL, Lin J, Anderson ML, Tiro JA, Green BB, Gao H, Meenan RT, Hansen K, Sparks A, Buist DSM. Strategies to Increase Cervical Cancer Screening With Mailed Human Papillomavirus Self-Sampling Kits: A Randomized Clinical Trial. JAMA. 2023;330:1971-1981. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 10] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
42. | Hammer A, Rositch A, Qeadan F, Gravitt PE, Blaakaer J. Age-specific prevalence of HPV16/18 genotypes in cervical cancer: A systematic review and meta-analysis. Int J Cancer. 2016;138:2795-2803. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 45] [Cited by in RCA: 46] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
43. | Tsukamoto K, Yamashita A, Maeki M, Tokeshi M, Imai H, Fukao A, Fujiwara T, Okudera K, Mizuki N, Okuda K, Shimada M. Enhanced Broad-Spectrum Efficacy of an L2-Based mRNA Vaccine Targeting HPV Types 6, 11, 16, 18, with Cross-Protection Against Multiple Additional High-Risk Types. Vaccines (Basel). 2024;12. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
44. | Gavioli CFB, Festa Neto C, Tyring SK, Silva LLDC, Oliveira WRP. High-risk mucosal HPV types associated with squamous cell carcinoma on the nose tip in an immunocompetent young man. An Bras Dermatol. 2018;93:716-718. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 3] [Article Influence: 0.4] [Reference Citation Analysis (0)] |
45. | Mortaki D, Tsitsopoulos E, Louizou E, Tsiambas E, Peschos D, Sioulas V, Galanos A, Tagka A, Gregoriou S, Stratigos A, Rigopoulos D, Nicolaidou E. Prevalence of Cervico-vaginal High-risk HPV Types and Other Sexually Transmitted Pathogens in Anogenital Warts Patients. Anticancer Res. 2020;40:2219-2223. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 1] [Article Influence: 0.2] [Reference Citation Analysis (0)] |
46. | De Marco F, Houissa-Kchouk F, Khelifa R, Marcante ML. High-risk HPV types in Tunisia. A pilot study reveals an unexpectedly high prevalence of types 58 and 82 and lack of HPV 18 among female prostitutes. J Med Virol. 2006;78:950-953. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 18] [Article Influence: 0.9] [Reference Citation Analysis (0)] |
47. | Geraets DT, Grünberg AW, van der Helm JJ, Schim van der Loeff MF, Quint KD, Sabajo LO, de Vries HJ. Cross-sectional study of genital carcinogenic HPV infections in Paramaribo, Suriname: prevalence and determinants in an ethnically diverse population of women in a pre-vaccination era. Sex Transm Infect. 2014;90:627-633. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 10] [Article Influence: 0.9] [Reference Citation Analysis (0)] |
48. | Ndemela LM, Ottoman OM, Chitemo HD, Minja CA, Rambau PF, Kidenya BR. Epidemiological distribution of high-risk human papillomavirus genotypes and associated factors among patients with esophageal carcinoma at Bugando medical center in Mwanza, Tanzania. BMC Cancer. 2024;24:932. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
49. | De Vuyst H, Alemany L, Lacey C, Chibwesha CJ, Sahasrabuddhe V, Banura C, Denny L, Parham GP. The burden of human papillomavirus infections and related diseases in sub-saharan Africa. Vaccine. 2013;31 Suppl 5:F32-F46. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 134] [Cited by in RCA: 171] [Article Influence: 15.5] [Reference Citation Analysis (0)] |
50. | Berza N, Zodzika J, Kivite-Urtane A, Baltzer N, Curkste A, Pole I, Nygård M, Pärna K, Stankunas M, Tisler A, Uuskula A. Understanding the high-risk human papillomavirus prevalence and associated factors in the European country with a high incidence of cervical cancer. Eur J Public Health. 2024;34:826-832. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
51. | Vinodhini K, Shanmughapriya S, Das BC, Natarajaseenivasan K. Prevalence and risk factors of HPV infection among women from various provinces of the world. Arch Gynecol Obstet. 2012;285:771-777. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 61] [Cited by in RCA: 55] [Article Influence: 3.9] [Reference Citation Analysis (0)] |
52. | Okoye JO, Chukwukelu CF, Okekpa SI, Ogenyi SI, Onyekachi-Umah IN, Ngokere AA. Racial Disparities Associated with the Prevalence of Vaccine and Non-Vaccine HPV Types and Multiple HPV Infections between Asia and Africa: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev. 2021;22:2729-2741. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 16] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
53. | Smith JS, Melendy A, Rana RK, Pimenta JM. Age-specific prevalence of infection with human papillomavirus in females: a global review. J Adolesc Health. 2008;43:S5-25, S25.e1. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 134] [Cited by in RCA: 200] [Article Influence: 11.8] [Reference Citation Analysis (0)] |
54. | Agarwal R, King JB, Gopalani SV, Senkomago V. Cervical cancer incidence and trends among women aged 15-29 years by county-level economic status and rurality - United States, 2007-2020. Cancer Epidemiol. 2025;94:102730. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
55. | Zu YE, Wang SF, Peng XX, Wen YC, Shen XX, Wang XL, Liao WB, Jia D, Liu JY, Peng XW. New cheaper human papilloma virus mass screening strategy reduces cervical cancer incidence in Changsha city: A clinical trial. World J Clin Oncol. 2024;15:1491-1500. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
56. | Li N, Franceschi S, Howell-Jones R, Snijders PJ, Clifford GM. Human papillomavirus type distribution in 30,848 invasive cervical cancers worldwide: Variation by geographical region, histological type and year of publication. Int J Cancer. 2011;128:927-935. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 694] [Cited by in RCA: 757] [Article Influence: 54.1] [Reference Citation Analysis (0)] |
57. | Zhao FH, Zhu FC, Chen W, Li J, Hu YM, Hong Y, Zhang YJ, Pan QJ, Zhu JH, Zhang X, Chen Y, Tang H, Zhang H, Durand C, Datta SK, Struyf F, Bi D; HPV-039 study group. Baseline prevalence and type distribution of human papillomavirus in healthy Chinese women aged 18-25 years enrolled in a clinical trial. Int J Cancer. 2014;135:2604-2611. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 17] [Cited by in RCA: 21] [Article Influence: 1.9] [Reference Citation Analysis (0)] |
58. | Guan P, Howell-Jones R, Li N, Bruni L, de Sanjosé S, Franceschi S, Clifford GM. Human papillomavirus types in 115,789 HPV-positive women: a meta-analysis from cervical infection to cancer. Int J Cancer. 2012;131:2349-2359. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 558] [Cited by in RCA: 680] [Article Influence: 52.3] [Reference Citation Analysis (0)] |
59. | Zhu B, Liu Y, Zuo T, Cui X, Li M, Zhang J, Yu H, Piao H. The prevalence, trends, and geographical distribution of human papillomavirus infection in China: The pooled analysis of 1.7 million women. Cancer Med. 2019;8:5373-5385. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 22] [Cited by in RCA: 47] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
60. | Tang X, Jones TE, Jiang W, Austin M, He Y, Li L, Tong L, Wang C, Yang K, Yin R, Zhao C. Extended human papillomavirus genotype distribution in cervical intraepithelial neoplasia and cancer: Analysis of 40 352 cases from a large academic gynecologic center in China. J Med Virol. 2023;95:e28302. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 16] [Reference Citation Analysis (0)] |
61. | Zhao S, Zhao X, Hu S, Lu J, Duan X, Zhang X, Chen F, Zhao F. Distribution of high-risk human papillomavirus genotype prevalence and attribution to cervical precancerous lesions in rural North China. Chin J Cancer Res. 2019;31:663-672. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 7] [Cited by in RCA: 11] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
62. | Wang Z, Li Z, Li J, Wang C, Wang W, Hao M, Zhao C. Prevalence and Distribution of HPV Genotypes in 1387 Women with Cervical Intraepithelial Neoplasia 2/3 in Shanxi Province, China. J Cancer. 2018;9:2802-2806. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 17] [Cited by in RCA: 13] [Article Influence: 1.9] [Reference Citation Analysis (0)] |
63. | Ramakrishnan S, Partricia S, Mathan G. Overview of high-risk HPV's 16 and 18 infected cervical cancer: pathogenesis to prevention. Biomed Pharmacother. 2015;70:103-110. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 35] [Cited by in RCA: 39] [Article Influence: 3.9] [Reference Citation Analysis (0)] |
64. | Obeid DA, Almatrrouk SA, Alfageeh MB, Al-Ahdal MNA, Alhamlan FS. Human papillomavirus epidemiology in populations with normal or abnormal cervical cytology or cervical cancer in the Middle East and North Africa: A systematic review and meta-analysis. J Infect Public Health. 2020;13:1304-1313. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 16] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
65. | Arbyn M, Tommasino M, Depuydt C, Dillner J. Are 20 human papillomavirus types causing cervical cancer? J Pathol. 2014;234:431-435. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 154] [Cited by in RCA: 171] [Article Influence: 17.1] [Reference Citation Analysis (0)] |
66. | Ciapponi A, Bardach A, Glujovsky D, Gibbons L, Picconi MA. Type-specific HPV prevalence in cervical cancer and high-grade lesions in Latin America and the Caribbean: systematic review and meta-analysis. PLoS One. 2011;6:e25493. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 69] [Cited by in RCA: 71] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
67. | de Martel C, Plummer M, Vignat J, Franceschi S. Worldwide burden of cancer attributable to HPV by site, country and HPV type. Int J Cancer. 2017;141:664-670. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1331] [Cited by in RCA: 1466] [Article Influence: 183.3] [Reference Citation Analysis (0)] |
68. | De Vuyst H, Clifford G, Li N, Franceschi S. HPV infection in Europe. Eur J Cancer. 2009;45:2632-2639. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 122] [Cited by in RCA: 109] [Article Influence: 6.8] [Reference Citation Analysis (0)] |
69. | Clifford GM, Gallus S, Herrero R, Muñoz N, Snijders PJ, Vaccarella S, Anh PT, Ferreccio C, Hieu NT, Matos E, Molano M, Rajkumar R, Ronco G, de Sanjosé S, Shin HR, Sukvirach S, Thomas JO, Tunsakul S, Meijer CJ, Franceschi S; IARC HPV Prevalence Surveys Study Group. Worldwide distribution of human papillomavirus types in cytologically normal women in the International Agency for Research on Cancer HPV prevalence surveys: a pooled analysis. Lancet. 2005;366:991-998. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 722] [Cited by in RCA: 692] [Article Influence: 34.6] [Reference Citation Analysis (0)] |
70. | Irving SA, Groom HC, Stokley S, McNeil MM, Gee J, Smith N, Naleway AL. Human Papillomavirus Vaccine Coverage and Prevalence of Missed Opportunities for Vaccination in an Integrated Healthcare System. Acad Pediatr. 2018;18:S85-S92. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 14] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
71. | Tabrizi SN, Brotherton JM, Kaldor JM, Skinner SR, Liu B, Bateson D, McNamee K, Garefalakis M, Phillips S, Cummins E, Malloy M, Garland SM. Assessment of herd immunity and cross-protection after a human papillomavirus vaccination programme in Australia: a repeat cross-sectional study. Lancet Infect Dis. 2014;14:958-966. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 213] [Cited by in RCA: 216] [Article Influence: 19.6] [Reference Citation Analysis (0)] |
72. | Machalek DA, Garland SM, Brotherton JML, Bateson D, McNamee K, Stewart M, Rachel Skinner S, Liu B, Cornall AM, Kaldor JM, Tabrizi SN. Very Low Prevalence of Vaccine Human Papillomavirus Types Among 18- to 35-Year Old Australian Women 9 Years Following Implementation of Vaccination. J Infect Dis. 2018;217:1590-1600. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 87] [Cited by in RCA: 98] [Article Influence: 16.3] [Reference Citation Analysis (0)] |
73. | Fuzzell LN, Perkins RB, Christy SM, Lake PW, Vadaparampil ST. Cervical cancer screening in the United States: Challenges and potential solutions for underscreened groups. Prev Med. 2021;144:106400. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 37] [Cited by in RCA: 97] [Article Influence: 24.3] [Reference Citation Analysis (0)] |
74. | Fontham ETH, Wolf AMD, Church TR, Etzioni R, Flowers CR, Herzig A, Guerra CE, Oeffinger KC, Shih YT, Walter LC, Kim JJ, Andrews KS, DeSantis CE, Fedewa SA, Manassaram-Baptiste D, Saslow D, Wender RC, Smith RA. Cervical cancer screening for individuals at average risk: 2020 guideline update from the American Cancer Society. CA Cancer J Clin. 2020;70:321-346. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 293] [Cited by in RCA: 521] [Article Influence: 104.2] [Reference Citation Analysis (0)] |
75. | Saslow D, Andrews KS, Manassaram-Baptiste D, Smith RA, Fontham ETH; American Cancer Society Guideline Development Group. Human papillomavirus vaccination 2020 guideline update: American Cancer Society guideline adaptation. CA Cancer J Clin. 2020;70:274-280. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 60] [Cited by in RCA: 105] [Article Influence: 21.0] [Reference Citation Analysis (0)] |
76. | Gu A, Li J, Li MY, Liu Y. Patient-derived xenograft model in cancer: establishment and applications. MedComm (2020). 2025;6:e70059. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Reference Citation Analysis (0)] |
77. | Garland SM, Kjaer SK, Muñoz N, Block SL, Brown DR, DiNubile MJ, Lindsay BR, Kuter BJ, Perez G, Dominiak-Felden G, Saah AJ, Drury R, Das R, Velicer C. Impact and Effectiveness of the Quadrivalent Human Papillomavirus Vaccine: A Systematic Review of 10 Years of Real-world Experience. Clin Infect Dis. 2016;63:519-527. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 285] [Cited by in RCA: 339] [Article Influence: 37.7] [Reference Citation Analysis (0)] |
78. | Huh WK, Joura EA, Giuliano AR, Iversen OE, de Andrade RP, Ault KA, Bartholomew D, Cestero RM, Fedrizzi EN, Hirschberg AL, Mayrand MH, Ruiz-Sternberg AM, Stapleton JT, Wiley DJ, Ferenczy A, Kurman R, Ronnett BM, Stoler MH, Cuzick J, Garland SM, Kjaer SK, Bautista OM, Haupt R, Moeller E, Ritter M, Roberts CC, Shields C, Luxembourg A. Final efficacy, immunogenicity, and safety analyses of a nine-valent human papillomavirus vaccine in women aged 16-26 years: a randomised, double-blind trial. Lancet. 2017;390:2143-2159. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 321] [Cited by in RCA: 288] [Article Influence: 36.0] [Reference Citation Analysis (0)] |
79. | Wu Q, Qian M, Welby S, Guignard A, Rosillon D, Gopala K, Xu Y, Liu K, He Y, Jiang N, Tan Q, Xie J, Zhu T, Wang Q, Pan Y, Zeng R, Yang J, Zhao X, Zhou M, Navarro-Torné A, Yu H, Borys D. Prospective, multi-center post-marketing surveillance cohort study to monitor the safety of the human papillomavirus-16/18 AS04-adjuvanted vaccine in Chinese girls and women aged 9 to 45 years, 2018-2020. Hum Vaccin Immunother. 2023;19:2283912. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
80. | Joura EA, Giuliano AR, Iversen OE, Bouchard C, Mao C, Mehlsen J, Moreira ED Jr, Ngan Y, Petersen LK, Lazcano-Ponce E, Pitisuttithum P, Restrepo JA, Stuart G, Woelber L, Yang YC, Cuzick J, Garland SM, Huh W, Kjaer SK, Bautista OM, Chan IS, Chen J, Gesser R, Moeller E, Ritter M, Vuocolo S, Luxembourg A; Broad Spectrum HPV Vaccine Study. A 9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med. 2015;372:711-723. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 992] [Cited by in RCA: 929] [Article Influence: 92.9] [Reference Citation Analysis (0)] |
81. | Hall MT, Simms KT, Lew JB, Smith MA, Brotherton JM, Saville M, Frazer IH, Canfell K. The projected timeframe until cervical cancer elimination in Australia: a modelling study. Lancet Public Health. 2019;4:e19-e27. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 177] [Cited by in RCA: 194] [Article Influence: 32.3] [Reference Citation Analysis (0)] |
82. | Papanicolaou GN, Traut HF. The diagnostic value of vaginal smears in carcinoma of the uterus. 1941. Arch Pathol Lab Med. 1997;121:211-224. [PubMed] |
83. | Schiffman M, Castle PE. The promise of global cervical-cancer prevention. N Engl J Med. 2005;353:2101-2104. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 158] [Cited by in RCA: 153] [Article Influence: 7.7] [Reference Citation Analysis (0)] |
84. | Nanda K, McCrory DC, Myers ER, Bastian LA, Hasselblad V, Hickey JD, Matchar DB. Accuracy of the Papanicolaou test in screening for and follow-up of cervical cytologic abnormalities: a systematic review. Ann Intern Med. 2000;132:810-819. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 689] [Cited by in RCA: 643] [Article Influence: 25.7] [Reference Citation Analysis (0)] |
85. | Arbyn M, Anttila A, Jordan J, Ronco G, Schenck U, Segnan N, Wiener H, Herbert A, von Karsa L. European Guidelines for Quality Assurance in Cervical Cancer Screening. Second edition--summary document. Ann Oncol. 2010;21:448-458. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 314] [Cited by in RCA: 344] [Article Influence: 22.9] [Reference Citation Analysis (0)] |
86. | Jordan J, Arbyn M, Martin-Hirsch P, Schenck U, Baldauf JJ, Da Silva D, Anttila A, Nieminen P, Prendiville W. European guidelines for quality assurance in cervical cancer screening: recommendations for clinical management of abnormal cervical cytology, part 1. Cytopathology. 2008;19:342-354. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 112] [Cited by in RCA: 119] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
87. | Davey E, Barratt A, Irwig L, Chan SF, Macaskill P, Mannes P, Saville AM. Effect of study design and quality on unsatisfactory rates, cytology classifications, and accuracy in liquid-based versus conventional cervical cytology: a systematic review. Lancet. 2006;367:122-132. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 183] [Cited by in RCA: 155] [Article Influence: 8.2] [Reference Citation Analysis (0)] |
88. | Goldie SJ, Gaffikin L, Goldhaber-Fiebert JD, Gordillo-Tobar A, Levin C, Mahé C, Wright TC; Alliance for Cervical Cancer Prevention Cost Working Group. Cost-effectiveness of cervical-cancer screening in five developing countries. N Engl J Med. 2005;353:2158-2168. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 428] [Cited by in RCA: 423] [Article Influence: 21.2] [Reference Citation Analysis (0)] |
89. | Ibrahim A, Aro AR, Rasch V, Pukkala E. Cervical cancer screening in primary health care setting in Sudan: a comparative study of visual inspection with acetic acid and Pap smear. Int J Womens Health. 2012;4:67-73. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 14] [Cited by in RCA: 15] [Article Influence: 1.2] [Reference Citation Analysis (0)] |
90. | Arbyn M, Sankaranarayanan R, Muwonge R, Keita N, Dolo A, Mbalawa CG, Nouhou H, Sakande B, Wesley R, Somanathan T, Sharma A, Shastri S, Basu P. Pooled analysis of the accuracy of five cervical cancer screening tests assessed in eleven studies in Africa and India. Int J Cancer. 2008;123:153-160. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 200] [Cited by in RCA: 197] [Article Influence: 11.6] [Reference Citation Analysis (0)] |
91. | Denny L, Quinn M, Sankaranarayanan R. Chapter 8: Screening for cervical cancer in developing countries. Vaccine. 2006;24 Suppl 3:S3/71-S3/77. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 156] [Cited by in RCA: 189] [Article Influence: 11.1] [Reference Citation Analysis (0)] |
92. | Sauvaget C, Fayette JM, Muwonge R, Wesley R, Sankaranarayanan R. Accuracy of visual inspection with acetic acid for cervical cancer screening. Int J Gynaecol Obstet. 2011;113:14-24. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 136] [Cited by in RCA: 160] [Article Influence: 11.4] [Reference Citation Analysis (0)] |
93. | Delga A, Goffin F, Kridelka F, Marée R, Lambert C, Delvenne P. Evaluation of CellSolutions BestPrep® automated thin-layer liquid-based cytology Papanicolaou slide preparation and BestCyte® cell sorter imaging system. Acta Cytol. 2014;58:469-477. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 9] [Cited by in RCA: 13] [Article Influence: 1.2] [Reference Citation Analysis (0)] |
94. | Siebers AG, Klinkhamer PJ, Grefte JM, Massuger LF, Vedder JE, Beijers-Broos A, Bulten J, Arbyn M. Comparison of liquid-based cytology with conventional cytology for detection of cervical cancer precursors: a randomized controlled trial. JAMA. 2009;302:1757-1764. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 160] [Cited by in RCA: 142] [Article Influence: 8.9] [Reference Citation Analysis (0)] |
95. | Goldhaber-Fiebert JD, Stout NK, Salomon JA, Kuntz KM, Goldie SJ. Cost-effectiveness of cervical cancer screening with human papillomavirus DNA testing and HPV-16,18 vaccination. J Natl Cancer Inst. 2008;100:308-320. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 145] [Cited by in RCA: 153] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
96. | Kim JJ, Burger EA, Sy S, Campos NG. Optimal Cervical Cancer Screening in Women Vaccinated Against Human Papillomavirus. J Natl Cancer Inst. 2017;109. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 54] [Cited by in RCA: 62] [Article Influence: 6.9] [Reference Citation Analysis (0)] |
97. | zur Hausen H. Condylomata acuminata and human genital cancer. Cancer Res. 1976;36:794. [PubMed] |
98. | Clavel C, Masure M, Bory JP, Putaud I, Mangeonjean C, Lorenzato M, Gabriel R, Quereux C, Birembaut P. Hybrid Capture II-based human papillomavirus detection, a sensitive test to detect in routine high-grade cervical lesions: a preliminary study on 1518 women. Br J Cancer. 1999;80:1306-1311. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 156] [Cited by in RCA: 164] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
99. | Ronco G, Dillner J, Elfström KM, Tunesi S, Snijders PJ, Arbyn M, Kitchener H, Segnan N, Gilham C, Giorgi-Rossi P, Berkhof J, Peto J, Meijer CJ; International HPV screening working group. Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet. 2014;383:524-532. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1041] [Cited by in RCA: 1183] [Article Influence: 107.5] [Reference Citation Analysis (0)] |
100. | Schiffman M, Wentzensen N, Wacholder S, Kinney W, Gage JC, Castle PE. Human papillomavirus testing in the prevention of cervical cancer. J Natl Cancer Inst. 2011;103:368-383. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 442] [Cited by in RCA: 496] [Article Influence: 35.4] [Reference Citation Analysis (0)] |
101. | Arbyn M, Smith SB, Temin S, Sultana F, Castle P; Collaboration on Self-Sampling and HPV Testing. Detecting cervical precancer and reaching underscreened women by using HPV testing on self samples: updated meta-analyses. BMJ. 2018;363:k4823. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 305] [Cited by in RCA: 467] [Article Influence: 66.7] [Reference Citation Analysis (0)] |
102. | Castle PE, Stoler MH, Wright TC Jr, Sharma A, Wright TL, Behrens CM. Performance of carcinogenic human papillomavirus (HPV) testing and HPV16 or HPV18 genotyping for cervical cancer screening of women aged 25 years and older: a subanalysis of the ATHENA study. Lancet Oncol. 2011;12:880-890. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 359] [Cited by in RCA: 377] [Article Influence: 26.9] [Reference Citation Analysis (0)] |
103. | Hariprasad R, Bagepally BS, Kumar S, Pradhan S, Gurung D, Tamang H, Sharma A, Bhatnagar T. Cost-utility analysis of primary HPV testing through home-based self-sampling in comparison to visual inspection using acetic acid for cervical cancer screening in East district, Sikkim, India, 2023. PLoS One. 2024;19:e0300556. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
104. | Ferrandino RM, Barlow J, Gold B, Kraft D, Roof SA, Teng MS, Khan MN, Berger MH, Misiukiewicz KJ, Sindhu KK, Bakst RL, Posner MR, Westra WH, Genden EM, Chai RL. Diagnostic Accuracy of Circulating Tumor HPV DNA Testing in Patients With a Lateral Neck Mass. JAMA Otolaryngol Head Neck Surg. 2024;150:1089-1096. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
105. | Forson W, Bhatia R, Currie H, Elasifer H, Connor L, Wilson A, Cuschieri K. High-risk HPV mRNA testing on self-samples offered to those who do not attend for organised cervical screening - real world data from the Dumfries and Galloway region in Scotland. J Clin Virol. 2024;175:105734. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
106. | Marchi F, Campagnari V, Filauro M, Iandelli A, Piazza C, Peretti G. Improving oropharyngeal cancer diagnosis and management: A call for accurate p16/HPV DNA testing in Southern Europe. Head Neck. 2024;46:3146-3147. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
107. | Teixeira JC, Vale DB, Campos CS, Polegatto I, Bragança JF, Discacciati MG, Zeferino LC. Transition from opportunistic cytological to organized screening program with DNA-HPV testing detected prevalent cervical cancers 10 years in advance. Sci Rep. 2024;14:20761. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
108. | Glass-Riveros E, Baumann K, Craemer K, Geller S, Nava Frenier M, McDonald J, Holt HK. The Acceptability and Feasibility of Self-Collected HPV Testing for Cervical Cancer Screening Among Black and Latinx Women in Chicago: Perspectives from the Community. Womens Health Rep (New Rochelle). 2024;5:735-743. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
109. | Saslow D, Andrews KS, Manassaram-Baptiste D, Loomer L, Lam KE, Fisher-Borne M, Smith RA, Fontham ET; American Cancer Society Guideline Development Group. Human papillomavirus vaccination guideline update: American Cancer Society guideline endorsement. CA Cancer J Clin. 2016;66:375-385. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 51] [Cited by in RCA: 48] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
110. | Saslow D, Castle PE, Cox JT, Davey DD, Einstein MH, Ferris DG, Goldie SJ, Harper DM, Kinney W, Moscicki AB, Noller KL, Wheeler CM, Ades T, Andrews KS, Doroshenk MK, Kahn KG, Schmidt C, Shafey O, Smith RA, Partridge EE; Gynecologic Cancer Advisory Group, Garcia F. American Cancer Society Guideline for human papillomavirus (HPV) vaccine use to prevent cervical cancer and its precursors. CA Cancer J Clin. 2007;57:7-28. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 327] [Cited by in RCA: 301] [Article Influence: 16.7] [Reference Citation Analysis (0)] |
111. | Bi XY, Pan ZW, Liu SL, Chen XM, Li J, Bu QF, Zuo ML, Wang J, Sun WP, Zhang Y, Liu W, Wang F, Li CZ. A feasibility study of DNA ploidy analysis, HPV, and TCT for screening of cervical cancer: A retrospective study. Medicine (Baltimore). 2024;103:e40038. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
112. | Xiao M, Zhang Z, Yu X, Fan Y, Chen X. Value of TCT combined with HPV and CA125 in early cervical cancer screening in a medical examination population. Cell Mol Biol (Noisy-le-grand). 2022;68:160-164. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
113. | Li H, Li L, Sun J, Dong S, Li H. Value of TCT combined with serum CA153 and CA50 in early diagnosis of cervical cancer and precancerous lesions. Pak J Med Sci. 2022;38:1471-1476. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
114. | Li T, Lai Y, Yuan J. The diagnostic accuracy of TCT + HPV-DNA for cervical cancer: systematic review and meta-analysis. Ann Transl Med. 2022;10:761. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
115. | Liu Y, Zhang L, Zhao G, Che L, Zhang H, Fang J. The clinical research of Thinprep Cytology Test (TCT) combined with HPV-DNA detection in screening cervical cancer. Cell Mol Biol (Noisy-le-grand). 2017;63:92-95. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 15] [Article Influence: 1.9] [Reference Citation Analysis (0)] |
116. | Liang H, Fu M, Zhou J, Song L. Evaluation of 3D-CPA, HR-HPV, and TCT joint detection on cervical disease screening. Oncol Lett. 2016;12:887-892. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 9] [Cited by in RCA: 11] [Article Influence: 1.2] [Reference Citation Analysis (0)] |
117. | Sun XF, Gu YQ, Wang AC, Wang J, Xie JL. [Value assessment of high-risk HPV test and TCT in the screening of cervical carcinoma]. Zhonghua Shiyan He Linchuangbing Duxue Zazhi. 2013;27:273-276. |
118. | Kulasingam SL, Havrilesky LJ, Ghebre R, Myers ER. Screening for cervical cancer: a modeling study for the US Preventive Services Task Force. J Low Genit Tract Dis. 2013;17:193-202. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 37] [Cited by in RCA: 41] [Article Influence: 3.4] [Reference Citation Analysis (0)] |
119. | Polman NJ, Ebisch RMF, Heideman DAM, Melchers WJG, Bekkers RLM, Molijn AC, Meijer CJLM, Quint WGV, Snijders PJF, Massuger LFAG, van Kemenade FJ, Berkhof J. Performance of human papillomavirus testing on self-collected versus clinician-collected samples for the detection of cervical intraepithelial neoplasia of grade 2 or worse: a randomised, paired screen-positive, non-inferiority trial. Lancet Oncol. 2019;20:229-238. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 76] [Cited by in RCA: 134] [Article Influence: 22.3] [Reference Citation Analysis (0)] |
120. | Massad LS, Einstein MH, Huh WK, Katki HA, Kinney WK, Schiffman M, Solomon D, Wentzensen N, Lawson HW; 2012 ASCCP Consensus Guidelines Conference. 2012 updated consensus guidelines for the management of abnormal cervical cancer screening tests and cancer precursors. J Low Genit Tract Dis. 2013;17:S1-S27. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 478] [Cited by in RCA: 499] [Article Influence: 41.6] [Reference Citation Analysis (0)] |
121. | McCredie MR, Sharples KJ, Paul C, Baranyai J, Medley G, Jones RW, Skegg DC. Natural history of cervical neoplasia and risk of invasive cancer in women with cervical intraepithelial neoplasia 3: a retrospective cohort study. Lancet Oncol. 2008;9:425-434. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 702] [Cited by in RCA: 746] [Article Influence: 43.9] [Reference Citation Analysis (0)] |
122. | Kyrgiou M, Athanasiou A, Kalliala IEJ, Paraskevaidi M, Mitra A, Martin-Hirsch PP, Arbyn M, Bennett P, Paraskevaidis E. Obstetric outcomes after conservative treatment for cervical intraepithelial lesions and early invasive disease. Cochrane Database Syst Rev. 2017;11:CD012847. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 56] [Cited by in RCA: 89] [Article Influence: 11.1] [Reference Citation Analysis (0)] |
123. | D'Alessandro P, Arduino B, Borgo M, Saccone G, Venturella R, Di Cello A, Zullo F. Loop Electrosurgical Excision Procedure versus Cryotherapy in the Treatment of Cervical Intraepithelial Neoplasia: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Gynecol Minim Invasive Ther. 2018;7:145-151. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 15] [Cited by in RCA: 22] [Article Influence: 3.1] [Reference Citation Analysis (0)] |
124. | Saslow D, Solomon D, Lawson HW, Killackey M, Kulasingam SL, Cain J, Garcia FA, Moriarty AT, Waxman AG, Wilbur DC, Wentzensen N, Downs LS Jr, Spitzer M, Moscicki AB, Franco EL, Stoler MH, Schiffman M, Castle PE, Myers ER; ACS-ASCCP-ASCP Cervical Cancer Guideline Committee. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. CA Cancer J Clin. 2012;62:147-172. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 854] [Cited by in RCA: 800] [Article Influence: 61.5] [Reference Citation Analysis (0)] |
125. | Choi S, Ismail A, Pappas-Gogos G, Boussios S. HPV and Cervical Cancer: A Review of Epidemiology and Screening Uptake in the UK. Pathogens. 2023;12. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 59] [Reference Citation Analysis (0)] |
126. | Smith MA, Gertig D, Hall M, Simms K, Lew JB, Malloy M, Saville M, Canfell K. Transitioning from cytology-based screening to HPV-based screening at longer intervals: implications for resource use. BMC Health Serv Res. 2016;16:147. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 26] [Cited by in RCA: 31] [Article Influence: 3.4] [Reference Citation Analysis (0)] |
127. | Qiao YL, Sellors JW, Eder PS, Bao YP, Lim JM, Zhao FH, Weigl B, Zhang WH, Peck RB, Li L, Chen F, Pan QJ, Lorincz AT. A new HPV-DNA test for cervical-cancer screening in developing regions: a cross-sectional study of clinical accuracy in rural China. Lancet Oncol. 2008;9:929-936. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 334] [Cited by in RCA: 339] [Article Influence: 19.9] [Reference Citation Analysis (0)] |
128. | Lorenzi AT, Syrjänen KJ, Longatto-Filho A. Human papillomavirus (HPV) screening and cervical cancer burden. A Brazilian perspective. Virol J. 2015;12:112. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 14] [Article Influence: 1.4] [Reference Citation Analysis (0)] |
129. | Zeferino LC, Bastos JB, Vale DBAPD, Zanine RM, Melo YLMF, Primo WQSP, Corrêa FM, Val ICCD, Russomano F. Guidelines for HPV-DNA Testing for Cervical Cancer Screening in Brazil. Rev Bras Ginecol Obstet. 2018;40:360-368. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 13] [Article Influence: 1.9] [Reference Citation Analysis (0)] |
130. | Lichtenfels M, Lorenzi NPC, Tacla M, Yokochi K, Frustockl F, Silva CA, Silva ALD, Termini L, Farias CB. A New Brazilian Device for Cervical Cancer Screening: Acceptability and Accuracy of Self-sampling. Rev Bras Ginecol Obstet. 2023;45:235-241. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
131. | Zhang L, Mosquera I, Lucas E, Rol ML, Carvalho AL, Basu P; CanScreen5 collaborators. CanScreen5, a global repository for breast, cervical and colorectal cancer screening programs. Nat Med. 2023;29:1135-1145. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 27] [Article Influence: 13.5] [Reference Citation Analysis (0)] |
132. | Minamitani M, Tatemichi M, Mukai T, Katano A, Ohira S, Nakagawa K. Adherence to national guidelines for colorectal, breast, and cervical cancer screenings in Japanese workplaces: a survey-based classification of enterprises' practices into "overscreening," "underscreening," and "guideline-adherence screening". BMC Public Health. 2024;24:2223. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
133. | Wang W, Arcà E, Sinha A, Hartl K, Houwing N, Kothari S. Cervical cancer screening guidelines and screening practices in 11 countries: A systematic literature review. Prev Med Rep. 2022;28:101813. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 33] [Reference Citation Analysis (0)] |
134. | Head KJ, Johnson NL, Scott SF, Zimet GD. Communicating Cervical Cancer Screening Results in Light of New Guidelines: Clinical Practices at Federally Qualified Health Centers. Health Commun. 2020;35:815-821. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 2] [Article Influence: 0.4] [Reference Citation Analysis (0)] |
135. | Castle P, Feldman S, Perkins RB. The Next Generation of Cervical Cancer Screening: Should Guidelines Focus on Best Practices for the Future or Current Screening Capacity? J Low Genit Tract Dis. 2018;22:91-96. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
136. | Salloum RG. Capsule Commentary on Haas et al., Provider Attitudes and Screening Practices Following Changes in Breast and Cervical Cancer Screening Guidelines. J Gen Intern Med. 2016;31:103. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
137. | Orang'o EO, Were E, Rode O, Muthoka K, Byczkowski M, Sartor H, Vanden Broeck D, Schmidt D, Reuschenbach M, von Knebel Doeberitz M, Bussmann H. Novel concepts in cervical cancer screening: a comparison of VIA, HPV DNA test and p16(INK4a)/Ki-67 dual stain cytology in Western Kenya. Infect Agent Cancer. 2020;15:57. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 10] [Cited by in RCA: 8] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
138. | Corrado G, Mazzara C, Certelli C, Nsubuga JB, Zanetto F, Schurfeld K, Dell'Antonio G, Orti CD, Savarese A, Andriani MT. Screening for cervical cancer in Africa: A proposal of a different combination of VIA test and cervical smear in Uganda. Int J Gynaecol Obstet. 2021;152:68-71. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
139. | Toliman PJ, Kaldor JM, Tabrizi SN, Vallely AJ. Innovative approaches to cervical cancer screening in low- and middle-income countries. Climacteric. 2018;21:235-238. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 16] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
140. | Ginsburg O, Bray F, Coleman MP, Vanderpuye V, Eniu A, Kotha SR, Sarker M, Huong TT, Allemani C, Dvaladze A, Gralow J, Yeates K, Taylor C, Oomman N, Krishnan S, Sullivan R, Kombe D, Blas MM, Parham G, Kassami N, Conteh L. The global burden of women's cancers: a grand challenge in global health. Lancet. 2017;389:847-860. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 572] [Cited by in RCA: 668] [Article Influence: 83.5] [Reference Citation Analysis (0)] |