IMR Press / FBL / Volume 28 / Issue 10 / DOI: 10.31083/j.fbl2810245
Open Access Review
Advances in Etiopathological Role and Control of HPV in Cervical Cancer Oncogenesis
Show Less
1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001 Hefei, Anhui, China
2 Departement de Bacteriologie-Virologie, Université des Sciences de la Santé, BP 18231 Libreville, Gabon
3 Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009 Jiangsu, Nanjing, China
4 Department of Radiation Oncology, Cedars Sinai Hospital, Los Angeles, CA 90048, USA
5 Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, Anhui, China
6 Institute of Health and Medicine, Hefei Comprehensive National Science Center, 230007 Hefei, Anhui, China
7 Institut de Recherche en Science de la Santé, Centre National de Recherche Scientifique et Technologique (IRSS/CNRST), BP 7047 Ouagadougou 03, Burkina Faso
*Correspondence: kombe@mail.ustc.edu.cn (Arnaud John Kombe Kombe); jint@ustc.edu.cn (Tengchuan Jin); abdouazaque@gmail.com (Abdou Azaque Zouré)
Front. Biosci. (Landmark Ed) 2023, 28(10), 245; https://doi.org/10.31083/j.fbl2810245
Submitted: 15 June 2023 | Revised: 13 July 2023 | Accepted: 31 July 2023 | Published: 19 October 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The human papillomavirus (HPV) is a well-known oncovirus whose causal link in the occurrence and development of several cancers, such as cervical cancer (CC), has been well established. Indeed, numerous researches depicted the etiological role of HPV in CC pathogenesis in such a way as to develop efficient strategies, including early diagnoses and HPV vaccination, to mitigate HPV infection and CC occurrence. Despite the effectiveness of these strategies in preventing HPV infection, its persistence, and the progression to precancerous lesions and cancers, extensive work that could give a better understanding of other unknown factors favoring oncogenesis is much more needed. In this last decade, scarce or few but crucial and strategic studies have been carried out to improve and deepen our understanding of the etiopathological role of HPV in the progression towards the development of CC. In this review, we highlighted the recent findings on the pathological role of HPV in CC occurrence and the advances in novel adopted strategies to reduce HPV infection and prevent CC occurrence more effectively.

Keywords
HPV
cervical cancer
cervicovaginal microbiota
cervicovaginal metabolic profile
HPV diagnosis
HPV treatment
1. Introduction

Human papillomavirus (HPV) is a group of more than 200 small viruses characterized by an 8 kb non-enveloped circular double-stranded genomic DNA that infects human mucosal and cutaneous epithelia. Generally, it is estimated that more than 80% of the sexually active worldwide population will be infected with HPV [1]. As a sexually transmissible infection (STI), HPV transmission mainly occurs during the first sexual intercourse, regardless of race and gender. It is, therefore, a ubiquitous STI [1]. Fortunately, in around 75–90% of HPV-infected people, HPV is armless, transient, and resolves spontaneously without treatment within one or two years, with no lifestyle-threatening clinical symptoms (reviewed in [2]).

However, HPV infection persists in a few rates of the global population. Indeed, people with poor health conditions that weakened the immune system, including people with other infections such as Herpes simplex virus (HSV), Human immunodeficiency virus (HIV), Epstein-Barr virus (EBV), and other STIs (reviewed in [2]), congenital diseases (epidermodysplasia verruciformis, congenital lymphopenia) [3], organ transplantation, or under cancer-related chemotherapy [2, 4], are the most at risk for reinfection or infection persistence, leading to anogenital warts, precancerous lesions (PCLs), and subsequently to cancers in women and men. While HPV 6 and 11 are associated with benign infection and meaningless outcomes (cutaneous and genital warts), seventeen HPV types (HPV16, 18, 31, 33, 35, 39, 45, 51, 52, 53, 58, 66, 68, 73, and 82) are noteworthy to be at high oncogenic risk (HR) and mainly found in women and men with PCLs and cancers [2].

Besides the vaginal, anal, penile, and upper aero-digestive tract cancers, cervical cancer (CC) is the most common and deadliest HPV-associated cancer caused by the persistence of high-risk HPV (HR-HPV) infections. In 2020, CC was responsible for 342,000 deaths [5], a slight increase compared with the death rate in 2018 (311,365) [6, 7], and according to the latest available statistical data, HPV-associated CC would kill 443,000 women around the world by the year 2030 [2]. Most of these fatality cases occur and will continue to occur in low- and middle-income countries (reviewed in [2]). Despite the strategic means adopted so far (mandatory HPV vaccination in young girls from 9 years old and routine PCL screening) to reduce HPV infection and mitigate the occurrence of CC, the burden of CC cancer worldwide is still significant and stigmatizing. As the fourth most prevalent cancer, CC is ranked the fourth deadliest female cancer after breast cancer [6] and continues to cause significant fatalities [2]. This suggests that much effort should be put forward to deepen our understanding of the oncogenic role of HPV infection.

This past decade, few but crucial, strategic, and determinant studies have been carried out to improve and deepen our understanding of the etiopathological role of HPV in CC occurrence [2]. These studies mainly focused on the beneficial role of HPV infection in the presence of other pathological or congenital disorders or how HPV infection takes advantage of weakened immunity to induce PCLs and CC. This deepened and advanced understanding of the etiopathological role of HPV infection in PCLs and CC occurrence might lead to the adoption of enhanced strategies to control infection and mitigate the occurrence of CC. In this review, we highlighted the recent findings on the pathological role of HPV in CC occurrence and the advances in novel adopted strategies to reduce HPV infection and prevent CC occurrence.

2. Unbalanced Cervicovaginal Microbiota and Metabolome: A Risk for HPV-Associated Cancers
2.1 Cervicovaginal Microbiota (CVM) in HPV Infection Persistence

It is noteworthy that HR-HPV infection may persist in certain individuals known as “people at risk”. These include people with sexually-transmissible diseases (AIDS-HIV, Chlamydia, and Gonorrhea), organ-transplant receivers, and the elderly; in short, people with an innate or acquired immune deficiency [2]. However, besides these people qualified as people at risk, it has been newly discovered that healthy women with a disrupted cervicovaginal microbiota (CVM) have a high risk for developing cervical intra-epithelial neoplasia grade 1 or 2 (CIN1/2) and invasive CC. Indeed, these studies described that the unbalanced bacterial population in the cervicovaginal area, caused by an increase or decrease of certain bacterial populations, is a major factor for HR-HPV acquisition and persistence of the infection towards high-grade lesions and cancer development [2, 8, 9].

A healthy CVM, which consists of a balanced variable population of bacteria, is dominated by Lactobacillus spp. [9, 10] and plays a crucial role in vaginal area protection against STIs and diseases [2, 8, 9]. Its disruption has been associated with several vaginal infections and diseases [8, 9]. Bacterial vaginosis (BV), for instance, which is a vaginal pathological condition, is characterized by a decreased amount of vaginal Lactobacillus spp. and an abundance of polymicrobial anaerobic bacteria, including Gardnerella vaginalis, Prevotella spp., Mobiluncus spp., Ureaplasma urealyticum, and Mycoplasma hominis [9]. Also, BV has been found to be a strong predictor of Neisseria gonorrhoeae and Chlamydia trachomatis infections [11], which are among the risk factors for HPV acquisition and infection persistence, the occurrence of CIN1/2, and malignancies, leading to the hypothesis that changes in CVM homeostasis have a crucial implication in the occurrence of cancerous lesions (CINs) [12]. Other studies have evidenced the strong causal link between genital immune mediators and the decrease in Lactobacillus spp. in cervical carcinogenesis [13, 14]. Several studies further confirmed this hypothesis (reviewed in [2]). More precisely, in their study, Usyk et al. [15] demonstrated that Lactobacillus spp. depletion in vaginal microbiota along with the abundance of strictly anaerobic bacteria, including Gardnerella vaginalis, Prevotella spp., Mobiluncus spp., Atopobium vaginae, and Mycoplasma hominis, is associated with a high risk of persistence of HR-HPV infections an occurrence of CIN2/3. Moreover, Norenhag et al. [10] confirmed that a CVM dominated by other bacterial populations than Lactobacillus spp. was associated with up to five times higher risk of contracting any HPVs and up to three times higher for HR-HPVs, which may induce invasive CC. Taken together, this demonstrates that—the abundance of—strictly anaerobic bacteria, but especially Gardnerella spp., are associated with the persistence of HR-HPVs infection and occurrence of malignancies, making “abnormal Gardnerella spp. increase” a point of control for monitoring HPV infection persistence (reviewed in [2]).

Controversially, while BV (or imbalanced CVM) has been since no long ago taught to be among the risk factors for HR-HPV infection and related carcinogenesis, Lebeau et al. [16] claimed in their recent study that it is instead the HR-HPV infection that leads to CVM alteration, making their causal relationship again questionable. Given that a healthy CVM consists of a low microbial diversity largely dominated (>90%) by a few Lactobacillus spp. (mainly L. crispatus, L. jensenii and L. iners) [9, 10, 16, 17, 18], the authors showed that, in the course of HPV infection, HR-HPV oncoproteins (E6 but mainly E7) down-regulate expression of the host innate peptides that are required for survival and sustainability of Lactobacillus spp., and antimicrobial activity on Gardnerella vaginalis in healthy vaginal mucosa [16]. Thus, inhibition of these host peptides, including defensin family (HβD1-4, HD5, HD6) and antimicrobial defensin-like family (SLPI, S100A7, Elafin, and LL-37) peptides, through the interaction of HPV E7 with NEMO, CK1, and β-TrCP of both NF-κB and Wnt/β-catenin signaling pathways, leads to BV (or dysbiosis), characterized by changes of the normally dominant lactic acid bacteria as described above [16, 17, 18] by a high diversity of bacterial population predominated by Gardnerella vaginalis and other anaerobic bacteria including Atopobium vaginae, Prevotella spp., Mobiluncus spp., Sneathia spp., Ureaplasma urealyticum, and Mycoplasma hominis [9, 16].

Overall, it should be retained that while unbalanced CVM (where Gardnerella spp. is high and Lactobacillus spp. is low) is now a well-characterized risk factor for HPV acquisition and infection persistence, HR-HPV itself participates in CVM alteration through molecular interactions, which weaken the bacterial cervicovaginal protection and enhance the occurrence of CC. Hence, an unbalanced CVM is a microbial marker that should alert clinicians to precancerous lesion risk and should be considered with high priority for treatment.

2.2 HPV Infection and Severity Induce Typical Cervicovaginal Metabolic Profiles (CVMPs)

Additionally, to the negative effect of HPV infection on CVM (and/or vis-versa) leading to HPV-associated disorders, the cervicovaginal metabolic profile (CVMP) might constitute another point of control for monitoring HPV infection risk and persistence of HR-HPV infection, but preferably for monitoring cervicovaginal cancers, because as metabolic diseases, cancers target and modify metabolome composition at the local environment [19, 20]. Studies in women with CC showed unique and typical CVMPs defined by a specific composition from amino acids to nucleic acids in CC tissues, which was completely absent from cancer-free/healthy women [21]. In the context of HPV-related CC, this typical CVM change is indirectly induced by HR-HPV infection through inhibition of host innate peptide [16] and cytokine [22] production that subsequently alters bacterial dynamics and antimicrobial activity in the cervicovaginal microenvironment and causes BV as described hereinbefore. For instance, changes from a healthy Lactobacillus spp.-dominated CVM towards an increased Gardnerella spp.-dominated anaerobic and microaerophilic poly-bacterial communities are strongly associated with increased amino acid catabolites and polyamines [23, 24]. More precisely, in a study profiling the cervicovaginal metabolism in HPV-infected women, with PCLs, CIN1/2/3, and CC, it was reported that for each woman and each specific grade of HPV-infection severity, the CVMP was typical and specifically different with a huge variation in vaginal pH and genital inflammation (Fig. 1) [25]. The CVMP in HPV-positive women with PCLs and CIN1/2/3 showed a decrease of amino acids metabolites and depletion of dipeptides characterized by HPV-positive women with CIN3. However, in HPV-positive women diagnosed with CC, the CVMP profile is characterized by increased xenobiotics and lipids from multiple macromolecule sources [25]. Hence, any increase of xenobiotics (mainly anti-inflammatory and analgesic drugs) and lipids (mainly 3-hydroxybutyrate, eicosenoate, and oleate/vaccinate) in an HPV-infected woman should be a suspicion of cancer development [25, 26], because many others studies evidenced that high level of 3-hydroxybutyrate, eicosenoate, and oleate/vaccinate augment are associated with tumor development and growth [27, 28], specifically CC [26, 29].

Fig. 1.

Effect of human papillomavirus (HPV) on cervicovaginal microbiota and metabolome profile and implication in carcinogenesis. The background color circled on the female’s body indicates the health state of the cervicovaginal microbiota: healthy and balanced cytomegalovirus (CMV) (colorless), slightly unbalanced cervicovaginal microbiota (CVM) (yellow), moderately unbalanced CVM (pink), severely unbalanced CVM (red). The color of the arrow bar (yellow [acid] to blue [basic]) indicates the vaginal pH. CIN1/2/3: cervical intra-epithelial neoplasia grade 1, 2 or 3.

3. Immunological Insight of Congenital Disorder in HPV Infection Persistence

Immunodeficiency is one of the main causes of HR-HPV infection persistence, leading to invasive CCs. Two main types of immunodeficiency are known: acquired and congenital immunodeficiency. Acquired or secondary immunodeficiency (AID or SID), mainly due to HIV infection or immunosuppressive treatments, is the most attributable risk factor for HR-HPV infection, infection persistence, and cancer occurrence [2]. Congenital or primary immunodeficiency (CID or PID) is due to inborn errors of immunity caused by mutations in genes involved in immune host defense and immune regulation [30]. The causal link of CID with HPV infection susceptibility was speculative. It was previously discovered that patients without AID but with severe HPV infection leading to malignancies were suffering from CID. Interestingly, many studies recently focused on the etiological role of CID in HPV susceptibility and reported that CID is involved in the persistence of HR-HPV infection in individuals without AID. This supports that CID, like AID, is a risk factor for HPV acquisition, persistence, and HPV-associated disease development [31, 32, 33, 34, 35].

To understand the involvement of CID in susceptibility to HPV acquisition and the persistence of the infection towards CC, findings from immunological studies have contributed quite a lot. It was reported that in either CID or AID, or both cases of patients, the cellular immune response was too low, with a low count of CD4+ and CD8+ T-cells, which is the main characteristic of lymphopenia states. Also, it is known that HIV specifically targets CD4+ T-cells and kills them. The immunological profile in CID and AID patients infected with HR-HPV, but not low-risk HPV, is comparable and characterized by a decreased T-cell response. These results suggested that T-cells, especially CD4+ T-cells, play a central role in the clearance of HPV and prevention of cancer occurrence [31, 32, 33, 34, 35]. A study by Nicholls et al. [36] reported that infiltration of CD4+, CD8+ T-cells, and dendritic cells in dogs infected with canine oral papillomavirus induces regression of papillomavirus-associated cancerous lesions, with CD4+ T-cells providing the most efficient healing effect. This study corroborates and confirms previous studies conducted by Iwatsuki et al. [37] and Coleman et al. [38], reporting that T-cells infiltration (more CD4+ and low CD8+ T-cells) are responsible for HPV6 and 11- associated warts regression.

Taken together, this highlights the protective role of CD8+ and preferentially CD4+ T-cells in the clearance of HPV infection, especially HR-HPVs, and their importance in preventing CC development. In other words, lymphocyte depletion caused by CID (as well as AID) constitutes a risk factor for HPV infection, persistence, and CC development.

4. Molecular Implication of HPV/HIV Co-Infection in CC Occurrence

The HPV/HIV co-infections are associated with an increased risk of HR-HPV persistence, acquisition of opportunistic infections, and development of genital malignancies [39]. The effect of HPV and HIV co-infection has been established, and co-infected HPV/HIV women are known to have reduced HPV clearance rates and an increased risk for persistent HR-HPV infection compared with HIV- negative women [40]. By promoting HPV-related carcinogenesis [39, 41], HIV positivity remained strongly associated with HR-HPV infection and HPV-associated cancer occurrence [40].

HIV-infected people have a suppressed immunity resulting in a high prevalence of HIV/AIDS-associated malignancies such as HPV-associated cancers, as described in paragraph 2. Besides the immunological point of view, other molecular mechanisms in which HPV and HIV are engaged have been suggested. However, the molecular mechanism behind these phenomena remains largely unclear [42].

Besides the lack of or the low titer of CD8+/CD4+ T-cell response due to HIV infection, host molecular genetic variations and HPV/HIV co-infection have an impact and play a crucial role in CC occurrence and progression [43, 44]. Some studies reported that people with a certain predisposition to human leukocyte antigen (HLA) molecules might be susceptible to HPV infection and persistence. Indeed, the persistence of oncogenic HPV in some women with specific HLA II alleles is strongly associated with the development of CC. For instance, the HLA II DRB1*13:01, DQB1*03:01, DQB1*03:19, and DQB1*06:02 alleles can modulate cervical carcinogenesis in HPV/HIV-1 co-infected women [45].

CC has also been associated with DNA genomic instability and DNA repair mechanisms. In HIV/AIDS, other factors than immunodeficiency, such as oncogenic DNA viruses and the HIV-1 protein Tat, have been associated with malignancies, as they significantly increase the incidence of carcinoma associated with DNA repair pathway dysregulation. It has been found that Tat-induced DNA repair deficiencies may play a significant role in the development of HIV-associated malignancies [46]. Similarly, based on this observation, Barillari et al. [47] could demonstrate that the extracellular Tat protein plays a significant role in the development of HPV-associated cancers, including CC. The prolonged interaction of viral envelope gp120 and transactivator proteins Tat with cervical induces an epithelial-mesenchymal transition mediated by SMAD2 phosphorylation and activation of transcription factors Slug, Snail, Twist1, and ZEB1. This interaction of gp120 and Tat proteins reduces E-cadherin expression and activates vimentin and N-cadherin expression [48]. The HIV Tat protein is incorporated into HIV-1 virions as a potent transcriptional transactivator of virus gene expression, which is essential for virus infectivity, replication, and transmission [49]. The HIV Tat protein transactivates the HPV long control region, increasing the expression of the oncogenes E6 and E7. One of the molecular interactions between HIV and HPV has been recently proposed [50]. HIV-1 Tat protein up-regulates HPV E6 and E7 gene expressions, which are mainly involved in cervical carcinogenesis and, therefore, the development of tumor cells.

Furthermore, the HIV-1 Tat protein induces cell cycle proliferation through the expression of cell cycle regulator genes [51], meaning that Tat promotes metastasis of cervical carcinoma during carcinogenesis mechanisms. Precisely, Tat protein is taken up by human uterine cervical carcinoma cells, and this affects (up-regulates) the expression of HPV oncoproteins (E6 or E7) and down-regulate cellular proteins (p16 and/or p53), which are key to cervical carcinoma development or progression. It has been demonstrated that HIV-1 Tat protein is taken up by human uterine cervical carcinoma cells and directly followed by an increase in the expression of the E6 protein of HPV, which is associated with the high incidence and clinical aggressiveness of uterine cervical carcinoma observed in HPV/HIV co-infected women [47].

5. New Regulations for Screening and Treatment of Cervical Pre-Cancer Lesions and CC Prevention

As previously reviewed [2], the two main strategies to fight against HPV-associated precancerous lesions and CC occurrence are vaccination and early diagnosis/screening, followed by appropriate treatment. However, because vaccines are prophylactics and only limited to preventing from getting infected by HPVs, preventing the persistence of HPV infection may slightly or not be overcome by HPV vaccines and not can it guarantee cancer occurrence, as the infection is already present. To a lesser extent, vaccines can prevent new infections after a primary transient infection, autoinfection, or sur-infection and limit the risk of developing CC. There is, unfortunately, no available direct treatment to eradicate HPV infection to date. Thus, for overall better prevention, HPV vaccines should be affordable, evenly, and equally available to all populations, specifically to people and areas of high prevalence, and implemented at an early age [52]. Yet, in developing countries, where the HPV-associated burden is tremendous (accounting for more than 80% of the global CC burden [5, 6, 53]), accessibility to prophylactic treatments is limited because of cost, cold chain transportation challenges, and poor health care.

The millennium objective targeted by the WHO is “to vaccinate 90% of eligible girls from a very young age against HPV, to screen 70% of eligible women at least twice in their lifetimes, and to effectively treat 90% of those with a positive screening test or a cervical lesion, including palliative care when needed”, in order to accelerate the elimination of CC as a public health problem, by 2030 ([54], https://www.who.int/publications/i/item/9789240014107). Therefore, reinforcing the currently implemented vaccination programs or designing alternative HPV vaccines with cost-effective development strategies for affordable products to control HPV infection and prevent CC occurrence are needed. Besides, diagnosing HPV infection and screening for cervical precancerous lesions are recommended to prevent CC occurrence.

5.1 Molecular Basis for the Development of Next-Generation HPV Prophylactics

Cervical cancer is a preventable disease as it lies on infection by HPV. The currently available HPV prophylaxis, including Cevarix, Gardasil, and Gardasil-9, are made from one of the main HPV proteins involved in the first steps of the infection, the major capsid L1 protein, obtained in the form of virus-like particles [55, 56]. Targeting this protein blocks further HPV interactions with host cells to initiate the early steps and prevents infection progression, which explains the proven efficacy of these three vaccines [52, 57] and long-term preventive immunity [58, 59, 60]. However, these vaccines have limitations, including limited protection against some but not all HR-HPVs.

Screening the molecular and intracellular trafficking of the HPV infection steps provides molecular insights for developing alternative drugs against HPV infections and CC (reviewed in [61]). Specifically, three main molecular target sites in the early steps of HPV infection, including (i) the heparan sulfate binding site, (ii) the HPV cellular internalization site, and (iii) the intracellular trafficking molecules, are promising in preventing HPV entry into cells and replication [61]. Of interest, targeting these therapeutic sites could allow the development of broad-spectrum prophylactics against all HPV types and, to a lesser extent, other viruses, as the antiviral mechanism lies on inhibiting the viral attachment to the cell surface or preventing the interactions between viral particles and intracellular components, by either binding to viral particles or interacting with intra- or extracellular receptors [61].

Specifically, heparan sulfate proteoglycans (HSPGs) are required cell receptors for the initial attachment of HPV and many other viruses, including coronaviruses [62, 63], herpes simplex viruses 1 and 2 (HSV-1/2), Dengue virus (DENV), echoviruses 5 and 6, enterovirus, human cytomegalovirus (HCMV), HIV, polyomaviruses [64], and hepatitis viruses [64, 65]. Thus, potential inhibitors that target the HSPG-depending early attachment might be promising against broad-spectrum viral infections [61, 64]. Interestingly, several molecules mimicking or interacting with HSPGs, including heparin, dendrimers, and carrageenan, have proven to be effective against HPV, HSV-1/2, HCMV, SARS-CoV-2, and DENV [61, 62, 64]. Hence, deeper in vivo studies to assess the protective efficacy and safety of these molecules are highly encouraged.

Likewise, during infection, HPVs use similar downstream intracellular trafficking with other viruses, which may constitute therapeutic targets against a broad range of viruses [61]. Several reports describe the promising use of molecules to prevent infection by HPVs and the occurrence of CC. Zhang et al. [66] designed a short peptide that can bind to retromer (an intracellular virus interacting protein that binds to HPV L2 protein [67]) and potentially compete with HPV16 as an antiviral strategy. They demonstrated that the synthetic peptide can enter cells, sequester retromer by occupying the HPV-16 L2 retromer-binding site and inhibit HPV infection [66]. Such a drug could prevent the occurrence of CC.

Moreover, endoplasmic reticulum (ER) chaperones have been identified as common proteins involved and required in intracellular viral trafficking, from entry and viral replication to the assembly and release of virions. To develop broad-spectrum antiviral therapeutics, several studies have proposed potential inhibitors of ER chaperones that can be used against HPV, HIV, hepatitis viruses, SARS-CoV-2, Zika virus, DENV, Ebola virus, HCMV, and many others [68]. ER chaperone inhibitors, including AR-12 (OSU-03012), EGCG (polyphenol), PU-WS13, Migitol, Indonlizidine derivatives, Juniferdin, PACMA 31, 16F16, Isoquecetin, Bacitracin, Origamicin, LOC14, and Tizoxanide, have proven to block the progression of viral infection [68]. Similarly, myosin motors are proteins that bind to actin filaments to generate force, mobility, and shape for cell structure and dynamics [69]. They play a central role in intracellular trafficking, exo- and endocytosis, and transcription. Thus, viruses, including HPVs, use or take advantage of these proteins of the cytoskeleton, which are necessary for HPV infection, specifically for viral internalization, packaging, and release [70, 71]. Therefore, the specific interactions between HPVs and myosin motors serve as a basis for developing antiviral drugs that could compromise such interactions. Overall, these studies might help develop broad-spectrum antiviral prophylactics that target common intracellular virus trafficking.

5.2 Diagnostic Approaches for CC Screening and Next-Generation Tests

For effective CC prevention, sexually active women are recommended to undergo cervicovaginal screening to identify those who are at risk of cervical pre-cancer (CIN1, 2, or 3). Currently, several diagnostic tests are available on the market. The well-known of these precancerous lesion screening methods include the Papanicolaou test, also known as the Pap smear or smear test, a cervicovaginal cytological test designed to detect abnormal cervical cells [72, 73, 74, 75]. Although wildly considered an effective method for detecting and preventing CC, the Pap smear test sensitivity is low (ranging from 30% to 87%) [72, 73, 76, 77, 78]. Hence, it was highly recommended that the Pap smear must be performed yearly for a better follow-up [73]. Besides, it was also proposed that the Pap smear may be replaced by liquid-based cytology (LBC) (which is slightly more sensitive than the Pap smear [72]) or associated with colposcopy-directed biopsy [73] to reduce the rate of undiagnosed cervical dysplasia. Therefore, it has been finally adopted that colposcopy-directed biopsy should be done in cases of macroscopic cervical architectural changes, abnormal findings, or positivity of the Pap smear cytology result [54, 73, 79]. Appropriate treatment is then determined after a biopsy of suspicious lesions for histological diagnosis. This approach has been proven effective in developed countries where cytology-based CC screening programs are well-implemented and strictly followed, yielding a five-time reduce fatalities from CC [80]. This observation is unfortunately not the case in developing countries [81], where access to health care and availability of standard care facilities are still major public health concerns. This implies that governments from low- and middle-income countries should make available financial resources to set up or strengthen screening programs, to reduce the CC-related death rate.

In the last decade, researchers have developed improved diagnostic/screening tests in order to reach the WHO’s goal by 2030. These screening tests include visual inspection with acetic acid (VIA) and molecular tests, specifically HR-HPV DNA detection-based tests by PCR [54], which are more suitable to use in any laboratory setting (Table 1, Ref. [54, 72, 74, 75, 82, 83, 84, 85]). More interestingly, it was recently reported that newer cervicovaginal screening tests had been made available for diagnosing HR-HPV infection and precancerous lesions, among which molecular tests, specifically directed to the detection of HPV mRNA, HPV-E6/E7, and HPV-DNA methylation. Besides that, more conclusive screening tests performed on cytological samples (p16/Ki-67 dual staining) and highly technology-based advanced visual inspection screening tests based on artificial intelligence or machine learning platforms (automated visual evaluation of digital images) have been launched (Table 1) [54, 82, 83, 84, 85, 86].

Table 1.Diagnostic tests for HPV infection and precancerous lesion screening.
Screening tests Screening principle/method Ref.
Current tests Pap smear Visual detection of epithelial cells (cytological) abnormalities/metastasis and dysplasia in a cervix smear sample directly onto a microscope slide [74, 75]
Liquid-based cytology (LBC) Visual detection and semi-automated quantitation of epithelial cells (cytological) abnormalities/meta-, and dysplasia in a cervix smear sample, pre-treated in physiologic liquid [72]
Dual staining cytology to identify p16 and Ki-67 Automated staining and semi-quantitative evaluation of slides/samples from the cervico-epithelial cells [85]
Visual inspection with acetic acid or with Lugol’s iodine (VIA/VILI) Visual (necked eye) detection of cervical epithelial cells with acetic acid or with Lugol’s iodine (VIA/VILI) [54]
Automated visual evaluation of digital image Intelligence artificial or deep learning-based cytological image evaluation [83]
Nucleic acid amplification tests (NAAT) Detection of high-risk HPV (HR-HPV) DNA or mRNA through nucleic acid amplification (PCR) [84]
Tests under evaluation (future tests) HPV-DNA methylation PCR-based quantitative site-specific CpG methylation using next-generation sequencing (NGS) approach [82]
Protein biomarkers Detection of HR-HPV antibodies or oncoproteins [54]
5.3 Approaches to Cervical Cancer Screening and Treatment

There are two regulation approaches for screening and treatment of precancerous lesions and preventing CC occurrence, categorized into seven possible combinations or algorithms, which include (i) the screen-and-treat approach, for which the treatment is only based on a positive primary screening test (2 algorithms), and (ii) the screen-triage-and-treat approach for which the adequate treatment is based on and follows a positive primary test followed by a positive secondary test [54] (Fig. 2).

Fig. 2.

The seven algorithms considered for screening and treatment of cervical pre-cancer lesions and cervical cancer (CC) prevention. HPV, human papillomavirus; VIA, visual inspection with acetic acid; Cyt., cytology; +, positive; -, negative; Susp, suspicion of positive cytology test.

Specifically, in the screen-and-treat approach, women positive for HR-HPV and precancerous lesions after the primary screening test are directly eligible for the appropriate treatment. Secondary screening is not needed in this approach to start the treatment. Precancerous cell ablation and excision are the main treatments for women to prevent CC development. Once the primary screening is positive, the appropriate treatment is done on either the same visit of the screening test (single-visit approach) or another day (multiple-visit approach) [2, 54].

In a screen-triage-and-treat approach, women who are positive for HR-HPV and precancerous lesions after the primary screening test undergo a secondary screening test. If this later is confirmed positive, the appropriate treatment is determined by prime colposcopy with biopsy and histopathological examination. However, if the secondary screening test is negative, only an appropriate follow-up evaluation is required [2, 54].

6. Conclusions

Cervical cancer is the fourth deadliest cancer in women, and HPV, the leading viral cause of CC, is the most common transmissible virus in the world. Persistence of the infection mainly occurs in case of infection with HR-HPVs, and this constitutes the primary risk factor for leading CC. Besides, host factors, notably immunodeficiency due to poor health conditions, including viral infections [2], congenital diseases [3], organ transplantation, and cancer-related chemotherapy [2, 4], constitute the second risk for infection persistence, responsible for PCLs, and CC.

Recent studies on the etiopathological role of HPV infection in CC occurrence demonstrated that the persistence of HR-HPV infection might also be more prominent in case of disruption of CVM, characterized by the abundance of Gardnerella spp. and scarcity of Lactobacillus spp. In fact, HPV infection promotes its own persistence by inducing CVM disruption through molecular inhibition of the host innate peptides required for the survival and sustainability of Lactobacillus spp. and antimicrobial activity on Gardnerella vaginalis in the healthy vaginal mucosa. Moreover, the HPV-associated disrupted cervicovaginal microenvironment is characterized by a typical metabolome profile specific for a given HPV infection severity or cervicovaginal inflammation. Furthermore, HIV-HPV co-infection and primary lymphopenia constitute other crucial factors for the persistence of HR-HPV infection and the occurrence of CC.

Given the causal role of HR-HPV infection in the occurrence of CC, the fighting strategies lie in diagnosing and treating precancerous intraepithelial lesions based on new regulations. This includes that every woman from 20 years old should undergo a cervicovaginal screening to identify those who are at risk of cervical pre-cancer and should be received an adequate treatment determined by one of the seven screening-treatment algorithms adopted Fig. 2. To end, while diagnosing HPV and cervical cytology have reduced incidence of CC, government and international decision-makers should provide resource lines to implement HPV vaccination programs rather than only screening programs around the world, but especially in low-and middle-income countries, where the HPV associated burden is blatant [2]. Furthermore, given the limitations of the current HPV vaccines, the development of broad-spectrum antiviral drugs and vaccines is highly encouraged.

Author Contributions

AJKK conceptualized and conceived the main idea, extracted the data, wrote the original draft, and formatted the manuscript for submission. SZA, GAB and FANB helped in designing the work, contributed in the main idea, extensively reviewed and edited the final version of the manuscript before submission. TJ and AAZ conceptualized the main idea and provided resources. All authors contributed to editorial changes in the manuscript. All the authors read and approved the final version of the manuscript for publication.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We would like to thanks our respective funders for their support.

Funding

AJKK is supported by the Chinese National Postdoctorate Subvention. TJ is supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (Grant No. XDB29030104), the National Natural Science Fund (Grant No.: 82272301), the Fundamental Research Funds for the Central Universities (WK9100000001). AAZ is supported by Institut de Recherche en Science de la Santé, Ouagadougou, BF.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Chesson HW, Dunne EF, Hariri S, Markowitz LE. The estimated lifetime probability of acquiring human papillomavirus in the United States. Sexually Transmitted Diseases. 2014; 41: 660–664.
[2]
Kombe Kombe AJ, Li B, Zahid A, Mengist HM, Bounda GA, Zhou Y, et al. Epidemiology and Burden of Human Papillomavirus and Related Diseases, Molecular Pathogenesis, and Vaccine Evaluation. Frontiers in Public Health. 2021; 8: 552028.
[3]
Birkhäuser M. Climacteric depression and anxiety. Therapeutische Umschau. Revue Therapeutique. 2021; 78: 427–434. (In German)
[4]
Tamarelle J, Thiébaut ACM, de Barbeyrac B, Bébéar C, Ravel J, Delarocque-Astagneau E. The vaginal microbiota and its association with human papillomavirus, Chlamydia trachomatis, Neisseria gonorrhoeae and Mycoplasma genitalium infections: a systematic review and meta-analysis. Clinical Microbiology and Infection: the Official Publication of the European Society of Clinical Microbiology and Infectious Diseases. 2019; 25: 35–47.
[5]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer Journal for Clinicians. 2021; 71: 209–249.
[6]
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: a Cancer Journal for Clinicians. 2018; 68: 394–424.
[7]
Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, et al. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. The Lancet. Global Health. 2020; 8: e191–e203.
[8]
López-Filloy M, Cortez FJ, Gheit T, Cruz Y Cruz O, Cruz-Talonia F, Chávez-Torres M, et al. Altered Vaginal Microbiota Composition Correlates With Human Papillomavirus and Mucosal Immune Responses in Women With Symptomatic Cervical Ectopy. Frontiers in Cellular and Infection Microbiology. 2022; 12: 884272.
[9]
Wei B, Chen Y, Lu T, Cao W, Tang Z, Yang H. Correlation between vaginal microbiota and different progression stages of cervical cancer. Genetics and Molecular Biology. 2022; 45: e20200450.
[10]
Norenhag J, Du J, Olovsson M, Verstraelen H, Engstrand L, Brusselaers N. The vaginal microbiota, human papillomavirus and cervical dysplasia: a systematic review and network meta-analysis. BJOG: an International Journal of Obstetrics and Gynaecology. 2020; 127: 171–180.
[11]
Wiesenfeld HC, Hillier SL, Krohn MA, Landers DV, Sweet RL. Bacterial vaginosis is a strong predictor of Neisseria gonorrhoeae and Chlamydia trachomatis infection. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America. 2003; 36: 663–668.
[12]
Mitra A, MacIntyre DA, Marchesi JR, Lee YS, Bennett PR, Kyrgiou M. The vaginal microbiota, human papillomavirus infection and cervical intraepithelial neoplasia: what do we know and where are we going next? Microbiome. 2016; 4: 58.
[13]
Łaniewski P, Barnes D, Goulder A, Cui H, Roe DJ, Chase DM, et al. Linking cervicovaginal immune signatures, HPV and microbiota composition in cervical carcinogenesis in non-Hispanic and Hispanic women. Scientific Reports. 2018; 8: 7593.
[14]
Audirac-Chalifour A, Torres-Poveda K, Bahena-Román M, Téllez-Sosa J, Martínez-Barnetche J, Cortina-Ceballos B, et al. Cervical Microbiome and Cytokine Profile at Various Stages of Cervical Cancer: A Pilot Study. PLoS ONE. 2016; 11: e0153274.
[15]
Usyk M, Zolnik CP, Castle PE, Porras C, Herrero R, Gradissimo A, et al. Cervicovaginal microbiome and natural history of HPV in a longitudinal study. PLoS Pathogens. 2020; 16: e1008376.
[16]
Lebeau A, Bruyere D, Roncarati P, Peixoto P, Hervouet E, Cobraiville G, et al. HPV infection alters vaginal microbiome through down-regulating host mucosal innate peptides used by Lactobacilli as amino acid sources. Nature Communications. 2022; 13: 1076.
[17]
Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486: 207–214.
[18]
Zhou X, Bent SJ, Schneider MG, Davis CC, Islam MR, Forney LJ. Characterization of vaginal microbial communities in adult healthy women using cultivation-independent methods. Microbiology (Reading, England). 2004; 150: 2565–2573.
[19]
Wishart DS, Mandal R, Stanislaus A, Ramirez-Gaona M. Cancer Metabolomics and the Human Metabolome Database. Metabolites. 2016; 6: 10.
[20]
Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: Still emerging. Cell Metabolism. 2022; 34: 355–377.
[21]
Sitter B, Bathen T, Hagen B, Arentz C, Skjeldestad FE, Gribbestad IS. Cervical cancer tissue characterized by high-resolution magic angle spinning MR spectroscopy. Magma (New York, N.Y.). 2004; 16: 174–181.
[22]
Karim R, Tummers B, Meyers C, Biryukov JL, Alam S, Backendorf C, et al. Human papillomavirus (HPV) upregulates the cellular deubiquitinase UCHL1 to suppress the keratinocyte’s innate immune response. PLoS Pathogens. 2013; 9: e1003384.
[23]
Srinivasan S, Morgan MT, Fiedler TL, Djukovic D, Hoffman NG, Raftery D, et al. Metabolic signatures of bacterial vaginosis. mBio. 2015; 6: e00204–e00215.
[24]
Liu Z, Roche PA. Macropinocytosis in phagocytes: regulation of MHC class-II-restricted antigen presentation in dendritic cells. Frontiers in Physiology. 2015; 6: 1.
[25]
Ilhan ZE, Łaniewski P, Thomas N, Roe DJ, Chase DM, Herbst-Kralovetz MM. Deciphering the complex interplay between microbiota, HPV, inflammation and cancer through cervicovaginal metabolic profiling. EBioMedicine. 2019; 44: 675–690.
[26]
Zietkowski D, Davidson RL, Eykyn TR, De Silva SS, Desouza NM, Payne GS. Detection of cancer in cervical tissue biopsies using mobile lipid resonances measured with diffusion-weighted (1)H magnetic resonance spectroscopy. NMR in Biomedicine. 2010; 23: 382–390.
[27]
Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, Chiavarina B, et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle (Georgetown, Tex.). 2010; 9: 3506–3514.
[28]
Turkoglu O, Zeb A, Graham S, Szyperski T, Szender JB, Odunsi K, et al. Metabolomics of biomarker discovery in ovarian cancer: a systematic review of the current literature. Metabolomics: Official Journal of the Metabolomic Society. 2016; 12: 60.
[29]
Yang K, Xia B, Wang W, Cheng J, Yin M, Xie H, et al. A Comprehensive Analysis of Metabolomics and Transcriptomics in Cervical Cancer. Scientific Reports. 2017; 7: 43353.
[30]
Castagnoli R, Delmonte OM, Notarangelo LD. Congenital and acquired defects of immunity: An ever-evolving story. Pediatric Allergy and Immunology: Official Publication of the European Society of Pediatric Allergy and Immunology. 2022; 33: 61–64.
[31]
Gormley RH, Kovarik CL. Human papillomavirus-related genital disease in the immunocompromised host: Part I. Journal of the American Academy of Dermatology. 2012; 66: 867.e1–882.
[32]
Gormley RH, Kovarik CL. Human papillomavirus-related genital disease in the immunocompromised host: Part II. Journal of the American Academy of Dermatology. 2012; 66: 883.e1–900.
[33]
Moerman M, Danielides VG, Nousia CS, Van Wanzeele F, Forsyth R, Vermeersch H. Recurrent focal epithelial hyperplasia due to HPV13 in an HIV-positive patient. Dermatology (Basel, Switzerland). 2001; 203: 339–341.
[34]
Tschachler E, Bergstresser PR, Stingl G. HIV-related skin diseases. Lancet (London, England). 1996; 348: 659–663.
[35]
Wieland U, Kreuter A, Pfister H. Human papillomavirus and immunosuppression. Current Problems in Dermatology. 2014; 45: 154–165.
[36]
Nicholls PK, Moore PF, Anderson DM, Moore RA, Parry NR, Gough GW, et al. Regression of canine oral papillomas is associated with infiltration of CD4+ and CD8+ lymphocytes. Virology. 2001; 283: 31–39.
[37]
Iwatsuki K, Tagami H, Takigawa M, Yamada M. Plane warts under spontaneous regression. Immunopathologic study on cellular constituents leading to the inflammatory reaction. Archives of Dermatology. 1986; 122: 655–659.
[38]
Coleman N, Birley HD, Renton AM, Hanna NF, Ryait BK, Byrne M, et al. Immunological events in regressing genital warts. American Journal of Clinical Pathology. 1994; 102: 768–774.
[39]
Adler DH, Wallace M, Bennie T, Abar B, Meiring TL, Williamson AL, et al. Cumulative Impact of HIV and Multiple Concurrent Human Papillomavirus Infections on the Risk of Cervical Dysplasia. Advances in Virology. 2016; 2016: 7310894.
[40]
Taku O, Businge CB, Mdaka ML, Phohlo K, Basera W, Garcia-Jardon M, et al. Human papillomavirus prevalence and risk factors among HIV-negative and HIV-positive women residing in rural Eastern Cape, South Africa. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases. 2020; 95: 176–182.
[41]
Bushara O, Krogh K, Weinberg SE, Finkelman BS, Sun L, Liao J, et al. Human Immunodeficiency Virus Infection Promotes Human Papillomavirus-Mediated Anal Squamous Carcinogenesis: An Immunologic and Pathobiologic Review. Pathobiology: Journal of Immunopathology, Molecular and Cellular Biology. 2022; 89: 1–12.
[42]
Bayurova E, Jansons J, Skrastina D, Smirnova O, Mezale D, Kostyusheva A, et al. HIV-1 Reverse Transcriptase Promotes Tumor Growth and Metastasis Formation via ROS-Dependent Upregulation of Twist. Oxidative Medicine and Cellular Longevity. 2019; 2019: 6016278.
[43]
Bogale AL, Belay NB, Medhin G, Ali JH. Molecular epidemiology of human papillomavirus among HIV infected women in developing countries: systematic review and meta-analysis. Virology Journal. 2020; 17: 179.
[44]
Chambuso RS, Gray CM, Kaambo E, Rebello G, Ramesar R. Impact of Host Molecular Genetic Variations and HIV/HPV Co-infection on Cervical Cancer Progression: A Systematic review. Oncomedicine. 2018; 3: 82–93.
[45]
Chambuso RS. Human Immunodeficiency Virus/Human Papillomavirus co-infection and host molecular genetics of cervical carcinoma. 2019.
[46]
Nunnari G, Smith JA, Daniel R. HIV-1 Tat and AIDS-associated cancer: targeting the cellular anti-cancer barrier? Journal of Experimental & Clinical Cancer Research: CR. 2008; 27: 3.
[47]
Barillari G, Palladino C, Bacigalupo I, Leone P, Falchi M, Ensoli B. Entrance of the Tat protein of HIV-1 into human uterine cervical carcinoma cells causes upregulation of HPV-E6 expression and a decrease in p53 protein levels. Oncology Letters. 2016; 12: 2389–2394.
[48]
Lien K, Mayer W, Herrera R, Rosbe K, Tugizov SM. HIV-1 proteins gp120 and tat induce the epithelial-mesenchymal transition in oral and genital mucosal epithelial cells. PLoS ONE. 2019; 14: e0226343.
[49]
Ensoli B, Moretti S, Borsetti A, Maggiorella MT, Buttò S, Picconi O, et al. New insights into pathogenesis point to HIV-1 Tat as a key vaccine target. Archives of Virology. 2021; 166: 2955–2974.
[50]
Pérez-González A, Cachay E, Ocampo A, Poveda E. Update on the Epidemiological Features and Clinical Implications of Human Papillomavirus Infection (HPV) and Human Immunodeficiency Virus (HIV) Coinfection. Microorganisms. 2022; 10: 1047.
[51]
Nyagol J, Leucci E, Onnis A, De Falco G, Tigli C, Sanseverino F, et al. The effects of HIV-1 Tat protein on cell cycle during cervical carcinogenesis. Cancer Biology & Therapy. 2006; 5: 684–690.
[52]
Dehlendorff C, Baandrup L, Kjaer SK. Real-World Effectiveness of Human Papillomavirus Vaccination Against Vulvovaginal High-Grade Precancerous Lesions and Cancers. Journal of the National Cancer Institute. 2021; 113: 869–874.
[53]
International Agency for Research on Cancer W. GLOBOCAN 2012: estimated cancer incidence, mortality and prevalence worldwide in 2012. 2012.
[54]
WHO. WHO guideline for screening and treatment of cervical pre-cancer lesions for cervical cancer prevention. 2nd edn. World Health Organization: Geneva. 2021.
[55]
Deschuyteneer M, Elouahabi A, Plainchamp D, Plisnier M, Soete D, Corazza Y, et al. Molecular and structural characterization of the L1 virus-like particles that are used as vaccine antigens in Cervarix™, the AS04-adjuvanted HPV-16 and -18 cervical cancer vaccine. Human Vaccines. 2010; 6: 407–419.
[56]
Zhao Q, Modis Y, High K, Towne V, Meng Y, Wang Y, et al. Disassembly and reassembly of human papillomavirus virus-like particles produces more virion-like antibody reactivity. Virology Journal. 2012; 9: 52.
[57]
Mikamo H, Yamagishi Y, Murata S, Yokokawa R, Han SR, Wakana A, et al. Efficacy, safety, and immunogenicity of a quadrivalent HPV vaccine in Japanese men: A randomized, Phase 3, placebo-controlled study. Vaccine. 2019; 37: 1651–1658.
[58]
Kjaer SK, Nygård M, Dillner J, Brooke Marshall J, Radley D, Li M, et al. A 12-Year Follow-up on the Long-Term Effectiveness of the Quadrivalent Human Papillomavirus Vaccine in 4 Nordic Countries. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America. 2018; 66: 339–345.
[59]
Tsang SH, Sampson JN, Schussler J, Porras C, Wagner S, Boland J, et al. Durability of Cross-Protection by Different Schedules of the Bivalent HPV Vaccine: The CVT Trial. Journal of the National Cancer Institute. 2020; 112: 1030–1037.
[60]
Huh WK, Joura EA, Giuliano AR, Iversen OE, de Andrade RP, Ault KA, et al. 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 (London, England). 2017; 390: 2143–2159.
[61]
Carse S, Bergant M, Schäfer G. Advances in Targeting HPV Infection as Potential Alternative Prophylactic Means. International Journal of Molecular Sciences. 2021; 22: 2201.
[62]
Zhang Q, Chen CZ, Swaroop M, Xu M, Wang L, Lee J, et al. Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. Cell Discovery. 2020; 6: 80.
[63]
Ling J, Li J, Khan A, Lundkvist Å, Li JP. Is heparan sulfate a target for inhibition of RNA virus infection? American Journal of Physiology. Cell Physiology. 2022; 322: C605–C613.
[64]
Cagno V, Tseligka ED, Jones ST, Tapparel C. Heparan Sulfate Proteoglycans and Viral Attachment: True Receptors or Adaptation Bias? Viruses. 2019; 11: 596.
[65]
Kalia M, Chandra V, Rahman SA, Sehgal D, Jameel S. Heparan sulfate proteoglycans are required for cellular binding of the hepatitis E virus ORF2 capsid protein and for viral infection. Journal of Virology. 2009; 83: 12714–12724.
[66]
Zhang P, Moreno R, Lambert PF, DiMaio D. Cell-penetrating peptide inhibits retromer-mediated human papillomavirus trafficking during virus entry. Proceedings of the National Academy of Sciences of the United States of America. 2020; 117: 6121–6128.
[67]
Zhang P, Monteiro da Silva G, Deatherage C, Burd C, DiMaio D. Cell-Penetrating Peptide Mediates Intracellular Membrane Passage of Human Papillomavirus L2 Protein to Trigger Retrograde Trafficking. Cell. 2018; 174: 1465–1476.e13.
[68]
Kohli E, Causse S, Baverel V, Dubrez L, Borges-Bonan N, Demidov O, et al. Endoplasmic Reticulum Chaperones in Viral Infection: Therapeutic Perspectives. Microbiology and Molecular Biology Reviews: MMBR. 2021; 85: e0003521.
[69]
Coluccio LM. Myosins, A Superfamily of Molecular Motors. Series. Advances in Experimental Medicine and Biology. 2020.
[70]
Sankovski E, Abroi A, Ustav M, Jr, Ustav M. Nuclear myosin 1 associates with papillomavirus E2 regulatory protein and influences viral replication. Virology. 2018; 514: 142–155.
[71]
Oswald E, Reinz E, Voit R, Aubin F, Alonso A, Auvinen E. Human papillomavirus type 8 E7 protein binds nuclear myosin 1c and downregulates the expression of pre-rRNA. Virus Genes. 2017; 53: 807–813.
[72]
Pankaj S, Nazneen S, Kumari S, Kumari A, Kumari A, Kumari J, et al. Comparison of conventional Pap smear and liquid-based cytology: A study of cervical cancer screening at a tertiary care center in Bihar. Indian Journal of Cancer. 2018; 55: 80–83.
[73]
Nkwabong E, Laure Bessi Badjan I, Sando Z. Pap smear accuracy for the diagnosis of cervical precancerous lesions. Tropical Doctor. 2019; 49: 34–39.
[74]
Kitchen FL, Cox CM. Papanicolaou Smear. StatPearls Publishing: Treasure Island (FL). 2022.
[75]
Walker HK, Hall WD, Hurst JW. Clinical Methods: The History, Physical, and Laboratory Examinations. 3rd edn. Butterworths: Boston. 1990.
[76]
Nanda K, McCrory DC, Myers ER, Bastian LA, Hasselblad V, Hickey JD, et al. Accuracy of the Papanicolaou test in screening for and follow-up of cervical cytologic abnormalities: a systematic review. Annals of Internal Medicine. 2000; 132: 810–819.
[77]
Koliopoulos G, Nyaga VN, Santesso N, Bryant A, Martin-Hirsch PP, Mustafa RA, et al. Cytology versus HPV testing for cervical cancer screening in the general population. The Cochrane Database of Systematic Reviews. 2017; 8: CD008587.
[78]
Specificity, sensitivity and cost. Nature Reviews Cancer. 2007; 7: 893.
[79]
Barut MU, Kale A, Kuyumcuoğlu U, Bozkurt M, Ağaçayak E, Özekinci S, et al. Analysis of Sensitivity, Specificity, and Positive and Negative Predictive Values of Smear and Colposcopy in Diagnosis of Premalignant and Malignant Cervical Lesions. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 2015; 21: 3860–3867.
[80]
Habbema D, De Kok IMCM, Brown ML. Cervical cancer screening in the United States and the Netherlands: a tale of two countries. The Milbank Quarterly. 2012; 90: 5–37.
[81]
Denny L, Kuhn L, Hu CC, Tsai WY, Wright TC, Jr. Human papillomavirus-based cervical cancer prevention: long-term results of a randomized screening trial. Journal of the National Cancer Institute. 2010; 102: 1557–1567.
[82]
Clarke MA, Gradissimo A, Schiffman M, Lam J, Sollecito CC, Fetterman B, et al. Human Papillomavirus DNA Methylation as a Biomarker for Cervical Precancer: Consistency across 12 Genotypes and Potential Impact on Management of HPV-Positive Women. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2018; 24: 2194–2202.
[83]
Hu L, Bell D, Antani S, Xue Z, Yu K, Horning MP, et al. An Observational Study of Deep Learning and Automated Evaluation of Cervical Images for Cancer Screening. Journal of the National Cancer Institute. 2019; 111: 923–932.
[84]
Sørbye SW, Fismen S, Gutteberg TJ, Mortensen ES, Skjeldestad FE. Primary cervical cancer screening with an HPV mRNA test: a prospective cohort study. BMJ Open. 2016; 6: e011981.
[85]
Wentzensen N, Fetterman B, Castle PE, Schiffman M, Wood SN, Stiemerling E, et al. p16/Ki-67 Dual Stain Cytology for Detection of Cervical Precancer in HPV-Positive Women. Journal of the National Cancer Institute. 2015; 107: djv257.
[86]
Lie AK, Kristensen G. Human papillomavirus E6/E7 mRNA testing as a predictive marker for cervical carcinoma. Expert Review of Molecular Diagnostics. 2008; 8: 405–415.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top