Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.897
Filter
1.
Hum Vaccin Immunother ; 20(1): 2381922, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39113230

ABSTRACT

A rapid review was conducted to explore the implementation determinants of human papillomavirus (HPV) vaccination in the World Health Organization African Region and describe their dynamic relationship. PubMed and Google Scholar were searched in October 2023 to find relevant literature. A total of 64 published studies that reported factors affecting HPV vaccination were identified. Analysis of identified factors yielded 74 implementation determinants of HPV vaccination across the five domains of the Consolidated Framework for Implementation Research (CFIR): two (2.70%) were in the innovation domain, seven (9.46%) were in the outer setting domain, 14 (18.92%) were in the inner setting domain, 37 (50%) were in the individual domain and 14 (18.92%) were in the implementation process domain. A causal loop diagram of these implementation determinants revealed four balancing and seven reinforcing loops. Applying systems lens promoted a more holistic understanding of the implementation determinants of HPV vaccination, exposing leverage points for interventions.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Vaccination , Female , Humans , Africa , Human Papillomavirus Viruses/immunology , Immunization Programs/organization & administration , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Systems Analysis
2.
Hum Vaccin Immunother ; 20(1): 2372883, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38977424

ABSTRACT

Multiple studies have documented low human papillomavirus (HPV) vaccine uptake among Chinese girls. It remains crucial to determine the parental willingness to pay (WTP) HPV vaccine for girls. We conducted a cross-sectional study recruiting 3904 parents with girls aged 9-14 in Shanghai, China, employing an online questionnaire with a convenience sampling strategy. Parental WTP, both range of payment and estimated point value, were determined for themselves (or wives) and daughters. HPV vaccine uptake was 22.44% in mothers and 3.21% in daughters. Respondents favored WTP ≤ 1000 CNY/138 USD for themselves (or wives), whereas showed increasing WTP along with valency of HPV vaccine for daughters (2-valent: 68.62% ≤1000 CNY/138 USD; 4-valent: 56.27% 1001-2000 CNY/138-277 USD; 9-valent: 65.37% ≥2001 CNY/277 USD). Overall, respondents showed higher WTP for daughters (median 2000 CNY/277 USD; IQR 1000-3600 CNY/138-498 USD) than for themselves (2000 CNY/277 USD; 1000-3500 CNY/138-483 USD) or wives (2000 CNY/277 USD; 800-3000 CNY/110-414 USD) (each p < .05). Furthermore, parental WTP was higher for international vaccine and 9-valent vaccine (each p < 0.05). Between two assumed government subsidy scenarios, parental preference for 9-valent vaccine remained consistently high for daughters (approximately 24% in each scenario), whereas preference for themselves (or wives) was sensitive to payment change between the subsidy scenarios. Using a discrete choice experiment, we found domestic vaccine was commonly preferred; however, certain sociodemographic groups preferred multivalent HPV vaccines. In conclusion, the valency of HPV vaccine may influence parental decision-making for daughters, in addition to vaccine price. Our findings would facilitate tailoring the HPV immunization program in China.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Parents , Humans , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/economics , Papillomavirus Vaccines/immunology , Female , China , Cross-Sectional Studies , Child , Adolescent , Papillomavirus Infections/prevention & control , Adult , Parents/psychology , Surveys and Questionnaires , Patient Acceptance of Health Care/statistics & numerical data , Vaccination/economics , Vaccination/psychology , Vaccination/statistics & numerical data , Male , Middle Aged
3.
Front Immunol ; 15: 1362770, 2024.
Article in English | MEDLINE | ID: mdl-38983849

ABSTRACT

The development of human papillomavirus (HPV) vaccines has made substantive progress, as represented by the approval of five prophylactic vaccines since 2006. Generally, the deployment of prophylactic HPV vaccines is effective in preventing newly acquired infections and incidences of HPV-related malignancies. However, there is still a long way to go regarding the prevention of all HPV infections and the eradication of established HPV infections, as well as the subsequent progression to cancer. Optimizing prophylactic HPV vaccines by incorporating L1 proteins from more HPV subtypes, exploring adjuvants that reinforce cellular immune responses to eradicate HPV-infected cells, and developing therapeutic HPV vaccines used either alone or in combination with other cancer therapeutic modalities might bring about a new era getting closer to the vision to get rid of HPV infection and related diseases. Herein, we summarize strategies for the development of HPV vaccines, both prophylactic and therapeutic, with an emphasis on the selection of antigens and adjuvants, as well as implications for vaccine efficacy based on preclinical studies and clinical trials. Additionally, we outline current cutting-edge insights on formulation strategies, dosing schedules, and age expansion among HPV vaccine recipients, which might play important roles in addressing barriers to vaccine uptake, such as vaccine hesitancy and vaccine availability.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Humans , Papillomavirus Vaccines/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Female , Vaccine Development , Adjuvants, Immunologic , Animals , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Papillomaviridae/immunology , Vaccine Efficacy
4.
Pediatrics ; 154(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38978512

ABSTRACT

BACKGROUND: Nine-valent human papillomavirus (9vHPV) vaccines can be administered in 2 doses 6 to 12 months apart in adolescents. The impact of extended dose intervals is unknown. We report immunogenicity and safety data in adolescents of a second 9vHPV vaccine dose administered ≥1 year after the first. METHODS: This open-label safety and immunogenicity study (NCT04708041) assessed extended-interval 2-dose regimens of 9vHPV vaccine among adolescents (10 to 15 years) who received 2 9vHPV vaccine doses: the first ≥1 year before enrollment, and second, at enrollment (day 1). We measured serologic responses to vaccine-targeted human papillomavirus (HPV) types at enrollment day 1 (pre-dose 2) and 1 month post-dose 2 (month 1) using a competitive LuminexV® immunoassay. We estimated effects of dose interval on geometric mean titers (GMTs) using regression modeling. Participants reported adverse events (AEs) through 15 days after vaccination. RESULTS: We enrolled 146 adolescents (mean age 13.3 years) with median 25 months since first 9vHPV vaccine dose (range: 12-53 months). Across vaccine-targeted HPV types, GMTs increased from day 1 to month 1; seropositivity at month 1 was 100%. Anti-HPV GMTs at month 1 were not affected by differences in dose interval of 12 to 53 months, based on regression modeling. The most common AEs were mild-to-moderate injection site reactions; no serious AEs were reported. CONCLUSIONS: Extending the interval between first and second 9vHPV vaccine doses to 12 to 53 months did not affect antibody responses, with favorable safety profile. These results support feasibility of extended interval regimens for 9vHPV vaccine.


Subject(s)
Immunization Schedule , Immunogenicity, Vaccine , Papillomavirus Infections , Papillomavirus Vaccines , Humans , Adolescent , Female , Male , Child , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/adverse effects , Papillomavirus Vaccines/immunology , Papillomavirus Infections/prevention & control , Antibodies, Viral/blood
5.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062859

ABSTRACT

To date, more than 400 types of human papillomavirus (HPV) have been identified. Despite the creation of effective prophylactic vaccines against the most common genital HPVs, the viruses remain among the most prevalent pathogens found in humans. According to WHO data, they are the cause of 5% of all cancers. Even more frequent are persistent and recurrent benign lesions such as genital and common warts. HPVs are resistant to many disinfectants and relatively unsusceptible to external conditions. There is still no drug available to inhibit viral replication, and treatment is based on removing lesions or stimulating the host immune system. This paper presents the systematics of HPV and the differences in HPV structure between different genetic types, lineages, and sublineages, based on the literature and GenBank data. We also present the pathogenesis of diseases caused by HPV, with a special focus on the role played by E6, E7, and other viral proteins in the development of benign and cancerous lesions. We discuss further prospects for the treatment of HPV infections, including, among others, substances that block the entry of HPV into cells, inhibitors of viral early proteins, and some substances of plant origin that inhibit viral replication, as well as new possibilities for therapeutic vaccines.


Subject(s)
Papillomaviridae , Papillomavirus Infections , Humans , Papillomavirus Infections/virology , Papillomavirus Infections/drug therapy , Papillomaviridae/physiology , Papillomaviridae/pathogenicity , Papillomaviridae/classification , Papillomaviridae/genetics , Virus Replication/drug effects , Papillomavirus Vaccines/therapeutic use , Papillomavirus Vaccines/immunology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology
6.
Eur Rev Med Pharmacol Sci ; 28(12): 3871-3879, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38946386

ABSTRACT

OBJECTIVE: The World Health Organization (WHO) declared that the human papillomavirus (HPV) is the most widespread infection that affects women's reproductive system. HPV is a serious concern to women's health, as it has a negative impact on women's quality of life. Approximately 70% of all occurrences of cervical cancer globally are caused by HPV strains 16 and 18. A few studies have found that HPV vaccinations play a significant role in protecting women against HPV infections. This study aims to identify the effectiveness of the HPV vaccine and to examine the influence of this vaccine on women's health. MATERIALS AND METHODS: EBSCO, PubMed, Cochrane, Google Scholar, Science Direct, and ProQuest were selected as electronic databases for systematic research. The inclusion criteria encompassed studies published in English from January 2019 to August 2023, evaluating the effectiveness of the HPV vaccine in women aged 18-76 years globally. This review included different types of studies, including cross-sectional, retrospective cohort, original randomized controlled trials, and prospective studies. Moreover, the included studies were evaluated using the Jonna Briggs Institute (JBI) methodological quality checklist tool. Two reviewers assessed the methodological quality of all studies using JBI guidelines. RESULTS: The search identified 11,095 articles, 19 of which were included in this review. Significant findings were found regarding the relationship between HPV vaccines and women's health. CONCLUSIONS: This review highlights the importance of the HPV vaccine and its administration among women to promote their health and prevent future infections.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Women's Health , Humans , Female , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Papillomavirus Infections/prevention & control , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Adolescent , Adult , Middle Aged , Quality of Life , Young Adult , Aged
7.
Sex Transm Infect ; 100(5): 271-280, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38914474

ABSTRACT

OBJECTIVES: Populations who seek HIV pre-exposure prophylaxis (PrEP) are disproportionately affected by hepatitis A virus (HAV), hepatitis B virus (HBV) and human papillomavirus (HPV). We examined immunity/vaccination against these infections among participants in the Ontario PrEP cohort study (ON-PrEP). METHODS: ON-PrEP is a prospective cohort of HIV-negative PrEP users from 10 Ontario clinics. We descriptively analysed baseline immunity/vaccination against HAV (IgG reactive), HBV (hepatitis B surface antibody >10) and HPV (self-reported three-dose vaccination). We further performed multivariable logistic regression to identify characteristics associated with baseline immunity/vaccination. We used cumulative incidence functions to describe vaccine uptake among participants non-immune at baseline. RESULTS: Of 633 eligible participants, 59.1% were white, 85.8% were male and 79.6% were gay. We found baseline evidence of immunity/vaccination against HAV, HBV and HPV in 69.2%, 81.2% and 16.8% of PrEP-experienced participants and 58.9%, 70.3% and 10.4% of PrEP-naïve participants, respectively. Characteristics associated with baseline HAV immunity were greater PrEP duration (adjusted OR (aOR) 1.41/year, 95% CI 1.09 to 1.84), frequent sexually transmitted and bloodborne infection (STBBI) testing (aOR 2.38, 95% CI 1.15 to 4.92) and HBV immunity (aOR 3.53, 95% CI 2.09 to 5.98). Characteristics associated with baseline HBV immunity were living in Toronto (aOR 3.54, 95% CI 1.87 to 6.70) or Ottawa (aOR 2.76, 95% CI 1.41 to 5.40), self-identifying as racialised (aOR 2.23, 95% CI 1.19 to 4.18), greater PrEP duration (aOR 1.39/year, 95% CI 1.02 to 1.90) and HAV immunity (aOR 3.75, 95% CI 2.19 to 6.41). Characteristics associated with baseline HPV vaccination were being aged ≤26 years (aOR 9.28, 95% CI 2.11 to 40.77), annual income between CAD$60 000 and CAD$119 000 (aOR 3.42, 95% CI 1.40 to 8.34), frequent STBBI testing (aOR 7.00, 95% CI 1.38 to 35.46) and HAV immunity (aOR 6.96, 95% CI 2.00 to 24.25). Among those non-immune at baseline, overall cumulative probability of immunity/vaccination was 0.70, 0.60 and 0.53 among PrEP-experienced participants and 0.93, 0.80 and 0.70 among PrEP-naïve participants for HAV, HBV and HPV, respectively. CONCLUSIONS: Baseline immunity to HAV/HBV was common, and a sizeable proportion of non-immune participants were vaccinated during follow-up. However, HPV vaccination was uncommon. Continued efforts should be made to remove barriers to HPV vaccination such as cost, inclusion in clinical guidelines and provider recommendation.


Subject(s)
HIV Infections , Hepatitis A , Hepatitis B , Papillomavirus Infections , Papillomavirus Vaccines , Pre-Exposure Prophylaxis , Adult , Female , Humans , Male , Middle Aged , Young Adult , Hepatitis A/prevention & control , Hepatitis A/immunology , Hepatitis A Vaccines/administration & dosage , Hepatitis A Vaccines/immunology , Hepatitis B/prevention & control , Hepatitis B/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/immunology , HIV Infections/prevention & control , Ontario , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Pre-Exposure Prophylaxis/statistics & numerical data , Prospective Studies , Vaccination/statistics & numerical data
8.
J Med Virol ; 96(6): e29732, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874202

ABSTRACT

Neutralizing antibodies (NAbs) are considered the primary mechanism of vaccine-mediated protection against human papillomaviruses (HPV), the causative agent of cervical cancer. However, the minimum level of NAb needed for protection is currently unknown. The HPV pseudovirion-based neutralization assay (PBNA) is the gold standard method for assessing HPV antibody responses but is time-consuming and labor-intensive. With the development of higher valency HPV vaccines, alternative serological assays with the capacity for multiplexing would improve efficiency and output. Here we describe a multiplex bead-based immunoassay to characterize the antibody responses to the seven oncogenic HPV types (HPV16/18/31/33/45/52/58) contained in the current licensed nonavalent HPV vaccine. This assay can measure antibody isotypes and subclasses (total IgG, IgM, IgA1-2, IgG1-4), and can be adapted to measure other antibody features (e.g., Fc receptors) that contribute to vaccine immunity. When tested with serum samples from unvaccinated and vaccinated individuals, we found high concordance between HPV-specific IgG using this multiplex assay and NAbs measured with PBNA. Overall, this assay is high-throughput, sample-sparing, and time-saving, providing an alternative to existing assays for the measurement and characterization of HPV antibody responses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Immunoglobulin G , Papillomavirus Infections , Papillomavirus Vaccines , Humans , Antibodies, Viral/blood , Immunoassay/methods , Female , Papillomavirus Infections/diagnosis , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Immunoglobulin G/blood , Papillomaviridae/immunology , Human Papillomavirus Viruses
9.
Viral Immunol ; 37(5): 221-239, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38841885

ABSTRACT

Human papillomavirus (HPV) is a circular, double-stranded DNA virus and recognized as the most prevalent sexually transmitted infectious agent worldwide. The HPV life cycle encompasses three primary stages. First, the virus infiltrates the basal cells of the stratified epidermis. Second, there is a low-level expression of viral genes and preservation of the viral genome in the basal layer. Lastly, productive replication of HPV occurs in differentiated cells. An effective immune response, involving various immune cells, including innate immunity, keratinocytes, dendritic cells, and natural killer T cells, is instrumental in clearing HPV infection and thwarting the development of HPV-associated tumors. Vaccines have demonstrated their efficacy in preventing genital warts, high-grade precancerous lesions, and cancers in females. In males, the vaccines can also aid in preventing genital warts, anal precancerous lesions, and cancer. This comprehensive review aims to provide a thorough and detailed exploration of HPV infections, delving into its genetic characteristics, life cycle, pathogenesis, and the role of high-risk and low-risk HPV strains. In addition, this review seeks to elucidate the intricate immune interactions that govern HPV infections, spanning from innate immunity to adaptive immune responses, as well as examining the evasion mechanisms used by the virus. Furthermore, the article discusses the current landscape of HPV vaccines and common treatments, contributing to a holistic understanding of HPV and its associated diseases.


Subject(s)
Papillomaviridae , Papillomavirus Infections , Papillomavirus Vaccines , Humans , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Female , Papillomaviridae/immunology , Papillomaviridae/genetics , Vaccination Coverage , Neoplasms/immunology , Neoplasms/therapy , Male , Immunity, Innate , Adaptive Immunity
10.
Mol Ther ; 32(7): 2340-2356, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38715363

ABSTRACT

Human papillomavirus (HPV) 16 and 18 infections are related to many human cancers. Despite several preventive vaccines for high-risk (hr) HPVs, there is still an urgent need to develop therapeutic HPV vaccines for targeting pre-existing hrHPV infections and lesions. In this study, we developed a lipid nanoparticle (LNP)-formulated mRNA-based HPV therapeutic vaccine (mHTV)-03E2, simultaneously targeting the E2/E6/E7 of both HPV16 and HPV18. mHTV-03E2 dramatically induced antigen-specific cellular immune responses, leading to significant CD8+ T cell infiltration and cytotoxicity in TC-1 tumors derived from primary lung epithelial cells of C57BL/6 mice expressing HPV E6/E7 antigens, mediated significant tumor regression, and prolonged animal survival, in a dose-dependent manner. We further demonstrated significant T cell immunity against HPV16/18 E6/E7 antigens for up to 4 months post-vaccination in immunological and distant tumor rechallenging experiments, suggesting robust memory T cell immunity against relapse. Finally, mHTV-03E2 synergized with immune checkpoint blockade to inhibit tumor growth and extend animal survival, indicating the potential in combination therapy. We conclude that mHTV-03E2 is an excellent candidate therapeutic mRNA vaccine for treating malignancies caused by HPV16 or HPV18 infections.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , RNA, Messenger , Animals , Mice , Papillomavirus Vaccines/immunology , Humans , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Infections/therapy , Papillomavirus Infections/prevention & control , Female , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Nanoparticles/chemistry , Human papillomavirus 16/immunology , Human papillomavirus 16/genetics , Mice, Inbred C57BL , Human papillomavirus 18/immunology , Human papillomavirus 18/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/genetics , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , CD8-Positive T-Lymphocytes/immunology , Repressor Proteins/immunology , Repressor Proteins/genetics , DNA-Binding Proteins , Liposomes
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 378-382, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710522

ABSTRACT

One of the most prevalent malignancies in women is cervical cancer. Cervical cancer is mostly brought on by chronic high-risk human papillomavirus 16 (HPV16) and HPV18 infection. Currently, the widely used HPV vaccines are the bivalent Cervarix, the tetravalent Gardasil, and the 9-valent Gardasil-9.There are differences in T cell effector molecule changes, B cell antibody level, duration, age and the injection after vaccination of the three vaccines.


Subject(s)
B-Lymphocytes , Papillomavirus Vaccines , T-Lymphocytes , Humans , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Female , T-Lymphocytes/immunology , B-Lymphocytes/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Vaccination , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage , Human Papillomavirus Viruses
12.
PLoS One ; 19(5): e0304080, 2024.
Article in English | MEDLINE | ID: mdl-38768231

ABSTRACT

Human Papillomavirus (HPV) prophylactic vaccination has proven effective in preventing new infections, but it does not treat existing HPV infections or associated diseases. Hence, there is still an important reservoir of HPV in adults, as vaccination programs are mainly focused on young women. The primary objective of this non-randomized, open-label trial is to evaluate if a 3-dose regimen of Gardasil-9 in HPV16/18-positive women could reduce the infective capacity of their body fluids. We aim to assess if vaccine-induced antibodies could neutralize virions present in the mucosa, thus preventing the release of infective particles and HPV transmission to sexual partners. As our main endpoint, the E1^E4-HaCaT model will be used to assess the infectivity rate of cervical, anal and oral samples, obtained from women before and after vaccination. HPV DNA positivity, virion production, seroconversion, and the presence of antibodies in the exudates, will be evaluated to attribute infectivity reduction to vaccination. Our study will recruit two different cohorts (RIFT-HPV1 and RIFT-HPV2) of non-vaccinated adult women. RIFT-HPV1 will include subjects with an HPV16/18 positive cervical test and no apparent cervical lesions or cervical lesions eligible for conservative treatment. RIFT-HPV2 will include subjects with an HPV16/18 positive anal test and no apparent anal lesions or anal lesions eligible for conservative treatment, as well as women with an HPV16/18 positive cervical test and HPV-associated vulvar lesions. Subjects complying with inclusion criteria for both cohorts will be recruited to the main cohort, RIFT-HPV1. Three doses of Gardasil-9 will be administered intramuscularly at visit 1 (0 months), visit 2 (2 months) and visit 3 (6 months). Even though prophylactic HPV vaccines would not eliminate a pre-existing infection, our results will determine if HPV vaccination could be considered as a new complementary strategy to prevent HPV-associated diseases by reducing viral spread. Trial registration: https://clinicaltrials.gov/ct2/show/NCT05334706.


Subject(s)
Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18 , Human papillomavirus 16 , Human papillomavirus 18 , Papillomavirus Infections , Adolescent , Adult , Female , Humans , Young Adult , Antibodies, Viral/immunology , Cervix Uteri/virology , DNA, Viral , Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Vaccination/methods , Clinical Trials as Topic , Evaluation Studies as Topic
13.
Virol J ; 21(1): 124, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822328

ABSTRACT

Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , mRNA Vaccines , Humans , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/therapy , Female , Papillomaviridae/immunology , Papillomaviridae/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Human Papillomavirus Viruses
14.
Hum Vaccin Immunother ; 20(1): 2352908, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38780076

ABSTRACT

Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.


Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.


Subject(s)
Epitopes, T-Lymphocyte , Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Vaccines, DNA , Female , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Human papillomavirus 16/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Epitopes, T-Lymphocyte/immunology , Animals , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Papillomavirus E7 Proteins/immunology , Mice , Humans , T-Lymphocytes, Cytotoxic/immunology , Repressor Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Mice, Inbred C57BL , Interferon-gamma/metabolism , Interferon-gamma/immunology
16.
Hum Vaccin Immunother ; 20(1): 2343552, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38723789

ABSTRACT

The main aim of our study was to investigate the specific contribution of a 9-valent human papillomavirus vaccine (9vHPV) to the recurrence risk of cervical intraepithelial neoplasia grade 2 or worse (CIN2+) in women vaccinated post-excision. Therefore, we conducted a retrospective monocentric cohort study in women aged 22-49 years undergoing conization between 2014 and 2023. The 9vHPV-vaccinated women were matched to unvaccinated women for age and follow-up duration in a 1:2 ratio to eliminate allocation bias. The risk of CIN2+ recurrence was estimated by the incidence rate ratio using Poisson regression with adjustment for comorbidities, smoking status, nulliparity, CIN grade, positive cone margin, and HPV genotypes. The CIN2+ recurrence rates in 147 women enrolled in the analysis were 18 and 2 cases per 100,000 person-days for unvaccinated and vaccinated women, respectively, during a mean follow-up period of 30 months (±22 months). A reduction in CIN2+ recurrences by 90% (95% confidence interval: 12-99%) was documented in 9vHPV-vaccinated participants compared to women undergoing only surgical excision. Moreover, vaccinated women with a positive cone margin showed a 42% (though non-significant) reduction in relapse (p = .661). Full post-conization vaccination with the 9vHPV contributed to an additional reduction in the risk of CIN2+ recurrence. This finding is consistent with current knowledge and suggests a high adjuvant effect of the 9vHPV vaccine.


Subject(s)
Neoplasm Recurrence, Local , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Humans , Female , Retrospective Studies , Adult , Middle Aged , Uterine Cervical Dysplasia/prevention & control , Uterine Cervical Dysplasia/virology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Young Adult , Neoplasm Recurrence, Local/prevention & control , Conization/methods , Vaccination
17.
Nat Commun ; 15(1): 3679, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693149

ABSTRACT

HPV vaccination with concomitant HPV-based screening of young women has been proposed for faster cervical cancer elimination. We describe the baseline results of a population-based trial of this strategy to reduce the incidence of HPV. All 89,547 women born 1994-1999 and resident in the capital region of Sweden were personally invited to concomitant HPV vaccination and HPV screening with 26,125 women (29.2%) enrolled between 2021-05-03 and 2022-12-31. Baseline HPV genotyping of cervical samples from the study participants finds, compared to pre-vaccination prevalences, a strong decline of HPV16 and 18 in birth cohorts previously offered vaccination, some decline for cross-protected HPV types but no decline for HPV types not targeted by vaccines. Our dynamic transmission modelling predicts that the trial could reduce the incidence of high-risk HPV infections among the 1994-1998 cohorts by 62-64% in 3 years. Baseline results are prevalences of HPV infection, validated transmission model projections, and power estimates for evaluating HPV incidence reductions at follow-up (+/-0.1% with 99.9% confidence). In conclusion, concomitant HPV vaccination and HPV screening appears to be a realistic option for faster cervical cancer elimination. Clinicaltrials.gov identifier: NCT04910802; EudraCT number: 2020-001169-34.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/epidemiology , Papillomavirus Infections/epidemiology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/therapeutic use , Adult , Sweden/epidemiology , Young Adult , Vaccination , Adolescent , Incidence , Mass Screening , Prevalence , Middle Aged , Early Detection of Cancer , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Human papillomavirus 18/genetics , Human papillomavirus 18/immunology , Human Papillomavirus Viruses
18.
Vaccine ; 42(14): 3277-3281, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38627144

ABSTRACT

BACKGROUND: Persistent human papillomavirus (HPV) infection can cause anogenital and oropharyngeal cancers. Many HPV infections and HPV-associated cancers are vaccine-preventable. Studies suggest long-term persistence of vaccine-induced antibodies. However, data are limited among Alaska Native people. METHODS: During 2011-2014, we enrolled Alaska Native children aged 9-14 years who received a 3-dose series of quadrivalent HPV vaccine (4vHPV). We collected sera at 1 month and 1, 2, 3, and 5 years post-vaccination to evaluate trends in type-specific immunoglobulin G antibody concentrations for the 4vHPV types (HPV 6/11/16/18). RESULTS: All participants (N = 469) had detectable antibodies against all 4vHPV types at all timepoints post-vaccination. For all 4vHPV types, antibody levels peaked by 1 month post-vaccination and gradually declined in subsequent years. At 5 years post-vaccination, antibody levels were higher among children who received 4vHPV at a younger age. CONCLUSIONS: Alaska Native children maintained antibodies against all 4vHPV types at 5 years post-vaccination.


Subject(s)
Alaska Natives , Antibodies, Viral , Immunogenicity, Vaccine , Papillomavirus Infections , Humans , Child , Adolescent , Female , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Antibodies, Viral/blood , Male , Alaska Natives/statistics & numerical data , Alaska , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/immunology , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18/administration & dosage , Vaccination , Immunoglobulin G/blood , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage
19.
Viruses ; 16(4)2024 04 20.
Article in English | MEDLINE | ID: mdl-38675981

ABSTRACT

The incidence and mortality rate of cervical cancer in Croatia remains a health challenge despite screening efforts. Besides the persistent infection with HPV, the development of cancer is also associated with some cofactors. The goal of this study was to assess circulating HPV genotypes and risk factors for the development of cervical precancer after almost 16 years from the onset of HPV vaccination in Croatia. In this study, a total of 321 women attending gynecological care were evaluated. Relevant medical and demographic information, including cytology, were collected. HPV genotyping was performed by PCR. Comparing the HPV types found in circulation in the pre-vaccination (1999-2015) and post-vaccination periods (2020-2023), a statistically significant reduction in HPV 31 was noted, while the overall prevalence increased in the post-vaccination period. Besides the expected HPV positivity as a risk factor, the history of smoking was associated with LSIL or worse cytology at enrollment. For the first time, this population study revealed a statistically significant shift in the HPV genotype in the post-vaccination period, as well as the confirmation of risk factors for the development of abnormal cytology among Croatian women.


Subject(s)
Genotype , Papillomaviridae , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Adult , Female , Humans , Middle Aged , Young Adult , Croatia/epidemiology , Papillomaviridae/genetics , Papillomaviridae/classification , Papillomavirus Infections/epidemiology , Papillomavirus Infections/virology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Prevalence , Risk Factors , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/prevention & control , Vaccination
20.
Expert Rev Vaccines ; 23(1): 523-534, 2024.
Article in English | MEDLINE | ID: mdl-38682812

ABSTRACT

BACKGROUND: Traditional vaccine development, often a lengthy and costly process of three separated phases. However, the swift development of COVID-19 vaccines highlighted the critical importance of accelerating the approval of vaccines. This article showcases a seamless phase 2/3 trial design to expedite the development process, particularly for multi-valent vaccines. RESEARCH DESIGN AND METHODS: This study utilizes simulation to compare the performance of seamless phase 2/3 design with that of conventional trial design, specifically by re-envisioning a 9-valent HPV vaccine trial. Across three cases, several key performance metrics are evaluated: overall power, type I error rate, average sample size, trial duration, the percentage of early stop, and the accuracy of dose selection. RESULTS: On average, when the experimental vaccine was assumed to be effective, the seamless design that performed interim analyses based solely on efficacy saved 555.73 subjects, shortened trials by 10.29 months, and increased power by 3.70%. When the experimental vaccine was less effective than control, it saved an average of 887.73 subjects while maintaining the type I error rate below 0.025. CONCLUSION: The seamless design proves to be a compelling strategy for vaccine development, given its versatility in early stopping, re-estimating sample sizes, and shortening trial durations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Research Design , Vaccine Development , Humans , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Vaccine Development/methods , Sample Size , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Computer Simulation
SELECTION OF CITATIONS
SEARCH DETAIL