Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 406
Filter
1.
J Virol ; 96(13): e0056622, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35703545

ABSTRACT

The family of human papillomaviruses (HPV) includes over 400 genotypes. Genus α genotypes generally infect the anogenital mucosa, and a subset of these HPV are a necessary, but not sufficient, cause of cervical cancer. Of the 13 high-risk (HR) and 11 intermediate-risk (IR) HPV associated with cervical cancer, genotypes 16 and 18 cause 50% and 20% of cases, respectively, whereas HPV16 dominates in other anogenital and oropharyngeal cancers. A plethora of ßHPVs are associated with cutaneous squamous cell carcinoma (CSCC), especially in sun-exposed skin sites of epidermodysplasia verruciformis (EV), AIDS, and immunosuppressed patients. Licensed L1 virus-like particle (VLP) vaccines, such as Gardasil 9, target a subset of αHPV but no ßHPV. To comprehensively target both α- and ßHPVs, we developed a two-component VLP vaccine, RG2-VLP, in which L2 protective epitopes derived from a conserved αHPV epitope (amino acids 17 to 36 of HPV16 L2) and a consensus ßHPV sequence in the same region are displayed within the DE loop of HPV16 and HPV18 L1 VLP, respectively. Unlike vaccination with Gardasil 9, vaccination of wild-type and EV model mice (Tmc6Δ/Δ or Tmc8Δ/Δ) with RG2-VLP induced robust L2-specific antibody titers and protected against ß-type HPV5. RG2-VLP protected rabbits against 17 αHPV, including those not covered by Gardasil 9. HPV16- and HPV18-specific neutralizing antibody responses were similar between RG2-VLP- and Gardasil 9-vaccinated animals. However, only transfer of RG2-VLP antiserum effectively protected naive mice from challenge with all ßHPVs tested. Taken together, these observations suggest RG2-VLP's potential as a broad-spectrum vaccine to prevent αHPV-driven anogenital, oropharyngeal, and ßHPV-associated cutaneous cancers. IMPORTANCE Licensed preventive HPV vaccines are composed of VLPs derived by expression of major capsid protein L1. They confer protection generally restricted to infection by the αHPVs targeted by the up-to-9-valent vaccine, and their associated anogenital cancers and genital warts, but do not target ßHPV that are associated with CSCC in EV and immunocompromised patients. We describe the development of a two-antigen vaccine protective in animal models against known oncogenic αHPVs as well as diverse ßHPVs by incorporation into HPV16 and HPV18 L1 VLP of 20-amino-acid conserved protective epitopes derived from minor capsid protein L2.


Subject(s)
Alphapapillomavirus , Carcinoma, Squamous Cell , Papillomaviridae , Papillomavirus Infections , Papillomavirus Vaccines , Vaccines, Virus-Like Particle , Alphapapillomavirus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Capsid Proteins/immunology , Carcinoma, Squamous Cell/prevention & control , Epitopes/immunology , Female , Human papillomavirus 16/immunology , Humans , Mice , Mice, Inbred BALB C , Papillomaviridae/immunology , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Rabbits , Vaccines, Virus-Like Particle/immunology
2.
Front Immunol ; 13: 782198, 2022.
Article in English | MEDLINE | ID: mdl-35185881

ABSTRACT

Misunderstanding temporal coincidence of adverse events during mass vaccination and invalid assessment of possible safety concerns have negative effects on immunization programs, leading to low immunization coverage. We conducted this systematic review and meta-analysis to identify the incidence rates of GBS that are temporally associated with viral vaccine administration but might not be attributable to the vaccines. By literature search in Embase and PubMed, we included 48 publications and 2,110,441,600 participants. The pooled incidence rate of GBS was 3.09 per million persons (95% confidence interval [CI]: 2.67 to 3.51) within six weeks of vaccination, equally 2.47 per 100,000 person-year (95%CI: 2.14 to 2.81). Subgroup analyses illustrated that the pooled rates were 2.77 per million persons (95%CI: 2.47 to 3.07) for individuals who received the influenza vaccine and 2.44 per million persons (95%CI: 0.97 to 3.91) for human papillomavirus (HPV) vaccines, respectively. Our findings evidence the GBS-associated safety of virus vaccines. We present a reference for the evaluation of post-vaccination GBS rates in mass immunization campaigns, including the SARS-CoV-2 vaccine.


Subject(s)
COVID-19 Vaccines/adverse effects , Guillain-Barre Syndrome/epidemiology , Influenza Vaccines/adverse effects , Mass Vaccination/adverse effects , Papillomavirus Vaccines/adverse effects , Alphapapillomavirus/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Population Surveillance , SARS-CoV-2/immunology
3.
Sci Rep ; 12(1): 2227, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140326

ABSTRACT

To assess the dynamics of human papillomavirus (HPV) serology, we analyzed HPV6-,11-,16-,18-, and 45 antibodies in infants during the first 36 months of their life. Serial serum samples of 276/327 mother-child pairs were collected at baseline (mothers) and at months 1, 2, 6, 12, 24 and 36 (offspring), and tested for HPVL1-antibodies using the GST-L1 assay. Concordance between maternal and infant HPV-antibody levels remained high until month-6 (p < = 0.001), indicating maternal antibody transfer. At 1 month, 40-62% of the infants tested seropositive to any of the 5 HPV-types. Between 1-3 years of age, 53% (58/109) of the children born to HPV-seronegative mothers tested HPV-seropositive. Times to positive seroconversion varied between13.4 and 18.7 months, and times to negative seroconversion (decay) between 8.5 and 9.9 months. Significant independent predictors of infants' seroconversion to LR-HPV were hand warts and mother's history of oral warts and seroconversion to LR-HPV. No predictors of seroconversion to HR-HPV were identified. Maternal HPV-IgG-antibodies are transferred to her offspring and remain detectable for 6 months, corroborating the IgG molecule's half-life. Seroconversion to HPV-genotypes 6, 11, 16 and 18 was confirmed among children born to HPV-seronegative mothers, implicating an immune response to these HPV-genotypes during early infancy.


Subject(s)
Alphapapillomavirus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Papillomavirus Infections/blood , Papillomavirus Infections/immunology , Seroconversion , Child, Preschool , Correlation of Data , Female , Finland/epidemiology , Humans , Infant , Infectious Disease Transmission, Vertical/statistics & numerical data , Kaplan-Meier Estimate , Longitudinal Studies , Male , Models, Statistical , Mothers , Papillomavirus Infections/epidemiology , Papillomavirus Infections/transmission , Pregnancy , Pregnancy Trimester, Third/blood , Pregnancy Trimester, Third/immunology , Prevalence , Prospective Studies , Seroepidemiologic Studies , Warts
4.
Virology ; 567: 15-25, 2022 02.
Article in English | MEDLINE | ID: mdl-34942562

ABSTRACT

HPV68 is a common HR-HPV, its persistent infection is closely related with the occurrence of cervical cancer. In this study, 2939 (27.60%, 2939/10650) positive samples were detected, and 174 (5.92%, 174/2939) were HPV68. 150 HPV68 E6-E7 were successful sequenced, 4 non-synonymous mutations were detected in E6, and E7 were 12. N133S non-synonymous mutations of HPV 68 E6 and C67G, T68 A/M of HPV68 E7 are E6, E7 positive selection sites, they all located in the key domains and major motifs of E6/E7 protein, the above amino-acid substitutions changed the protein structure, disturbed the interaction with other protein or cellular factors and make a difference in epitopes affinity, may affect the pathogenicity and adaptability of HPV68 to the environment. The enrichment of HPV68 data is of great significance for understanding the inherent geographical and biological differences of HPV68 in China.


Subject(s)
Alphapapillomavirus/genetics , Mutation , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/epidemiology , Alphapapillomavirus/chemistry , Alphapapillomavirus/classification , Alphapapillomavirus/immunology , Amino Acid Sequence , Amino Acid Substitution , B-Lymphocytes/immunology , B-Lymphocytes/virology , Binding Sites , Cervix Uteri/immunology , Cervix Uteri/virology , China/epidemiology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Female , Genotype , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Models, Molecular , Molecular Typing , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/chemistry , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Phylogeny , Prevalence , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , T-Lymphocytes/immunology , T-Lymphocytes/virology
5.
J Cancer Res Ther ; 17(6): 1468-1472, 2021.
Article in English | MEDLINE | ID: mdl-34916379

ABSTRACT

BACKGROUND: Human papilloma virus (HPV) has been widely implicated in cervical carcinogenesis and 90% of carcinoma cervix cases are due to high-risk HPV infection. This study was done to find the high-risk HPV genotypes in the rural women of Lucknow, North India. MATERIALS AND METHODS: HPV-DNA testing has been carried out in 130 cases of squamous intraepithelial lesions (SILs) of the cervix to find HPV status and type of high-risk HPV genotype infecting the rural women. These cases were collected from the rural cervical cancer screening program carried out in the villages of West Lucknow, North India. RESULTS: HPV status in 130 SIL cases revealed HPV positivity in only 17 cases (13.1%), whereas the remaining 113 cases were HPV negative (86.9%). HPV genotypes detected in the study were HPV-18, HPV-31, HPV-33, and HPV-35. HPV positivity was found highly associated with the young and sexually active group of women complaining of vaginal discharge. High HPV infection rate was also seen with multiparity and illiteracy as majority of women attending the camps were multiparous and illiterate. CONCLUSIONS: The present study revealed highly oncogenic HPV-18 alone or in combination with multiple infections of high-risk genotypes - 31, 33, and 35 - in the rural women of Lucknow, North India. Since HPV vaccine currently available in India is for HPV-16 and HPV-18 combined, efforts should be made to make region-specific vaccine according to their prevalence in that particular state of the country to provide effective HPV vaccination.


Subject(s)
Alphapapillomavirus/genetics , Papillomavirus Infections/epidemiology , Rural Population/statistics & numerical data , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/virology , Adult , Age Factors , Alphapapillomavirus/immunology , Alphapapillomavirus/isolation & purification , Carcinogenesis/pathology , Cross-Sectional Studies , DNA, Viral/genetics , DNA, Viral/isolation & purification , Female , Genotyping Techniques , Humans , India/epidemiology , Papillomavirus Infections/diagnosis , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Risk Factors , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/prevention & control , Young Adult , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/prevention & control
6.
Lima; Instituto Nacional de Salud; dic. 2021.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1354025

ABSTRACT

ANTECEDENTES: Las personas infectadas con VIH tienen mayor prevalencia y persistencia de infección por VPH, lo cual produce mayor riesgo de desarrollar enfermedades relacionadas con el VPH, incluyendo cáncer, y adquirir una enfermedad de progresión más rápida. Debido a ello, los programas de inmunización contra VPH son una prioridad de salud pública y una estrategia particularmente importante en la población infantil infectada o expuesta al VIH. OBJETIVO: Describir la evidencia sobre la eficacia, seguridad y recomendaciones de uso de vacunas contra el virus del papiloma humano (VPH) en niños expuestos e infectados con VIH. MÉTODO: Búsqueda electrónica de estudios publicados en español o inglés en PubMed, Cochrane Library, Web of Science y LILACS hasta el 11 de diciembre de 2021. Adicionalmente, se realizó una búsqueda en PubMed y repositorios de organismos elaboradores de Guías de Práctica Clínica. La selección de estudios fue desarrollada por un solo revisor. RESULTADOS: Inmunogenicidad contra VPH-6 El porcentaje de infectados y expuestos pero no infectados con VIH que alcanzó seropositividad fue 84.4% y 92.3% tras una dosis y 82.2% y 100% tras la tercera dosis. En infectados con VIH, la seropositividad después de completar tres dosis varió entre 96.6-100% al primer mes, y 88.5% a los 18 meses. Una cuarta dosis dos años después produjo seropositividad en el 97% a los 2 años, 99% a los 3.5 años y 95% a los 5 años. Inmunogenicidad contra VPH-11: El porcentaje de infectados y expuestos pero no infectados con VIH que alcanzó seropositividad fue 83.1% y 94.5% tras una dosis, y 84.4% y 100% tras la tercera dosis. En infectados con VIH, la seropositividad después de tres dosis varió entre 97.2-100% al primer mes y 84.6% a los 18 meses. Una cuarta dosis produjo seropositividad de 97%, 99% y 98% después de 2, 3.5 y 4-5 años. Inmunogenicidad contra VPH-16: El porcentaje de infectados y expuestos pero no infectados con VIH que alcanzó seropositividad fue 87.7% y 98.9% tras una dosis y 92.2% y 100% tras la tercera dosis. En infectados con VIH, la seropositividad luego de tres dosis varió entre 98.3-100% al primer mes y 100% a los 18 meses. Una cuarta dosis produjo seropositividad de 99% a los 2 a 3.5 años, y de 98% entre los 4 a 5 años. Inmunogenicidad contra VPH-18: La seropositividad en infectados y expuestos pero no infectados con VIH fue 62.3% y 86.8% tras una dosis, y 61.1% y 81.8% tras la tercera dosis. En infectados con VIH, la seropositividad después de tres dosis fue 72.5% a los 6 meses y 72% a los 18 meses. Una cuarta dosis produjo seropositividad en 81% de infectados a los 2 años, 77% a los 3.5 años, y 74% entre los 4 a 5 años. Eventos adversos asociados a la vacunación: Los eventos adversos (EA) fueron reportados en dos estudios y alcanzaron a 49-74.4% de infectados con VIH. Entre un 45.7-64% de los EA correspondieron a molestias en el sitio de inyección. Los EA sistémicos fueron principalmente fatiga y dolor de cabeza (11.4%). En un estudio se reportaron dos EA graves relacionados con la vacunación: nefritis y elevación de alanina aminostransferasa, resueltos sin necesidad de interrumpir el esquema de vacunación. Recomendaciones sobre la vacunación contra VHA en niños con VIH: Todas las GPC recomiendan inmunizar contra VPH. Las GPC de Ecuador, AEPCC, CDC y Colombia recomiendan inmunizar infectados con VIH independientemente de su sexo, mientras que OMS recomienda inmunizar solo a niñas, aunque su recomendación no es explícita para población con VIH. Ecuador y CDC recomiendan aplicar tres dosis y OMS recomienda aplicar dos dosis. En todas las GPC, la edad de inicio recomendada es a los 9 años, aunque Colombia hace la distinción de iniciar a los 11 años en varones. Ecuador, AEPCC y CDC recomiendan inmunizar contra VPH hasta los 26 años. CONCLUSIONES: En niños y adolescentes infectados con VIH, una serie de tres dosis de vacuna contra VPH produjo seropositividad superior a 80% para anticuerpos contra VPH-6, 11 y 16 en todos los periodos de tiempo evaluados, que abarcan un seguimiento de hasta 18 meses posteriores a finalizar la serie. Una dosis de refuerzo a los dos años logró seropositividad cercana al 100% que se mantuvo estable, incluso en periodos de seguimiento de hasta 4-5 años. En relación al genotipo 18 del VPH, la inmunización con tres dosis de vacuna tetravalente contra VPH logró baja seropositividad, entre 62-72%. Una dosis de refuerzo aumentó ligeramente la seropositividad hasta un 74% a los 4-5 años. A pesar de la seropositividad superior al 80% para los genotipos 6, 11 y 16 del VPH, fue consistente observar concentraciones más bajas de anticuerpos en infectados con VIH, en comparación con poblaciones no infectadas. La probabilidad de alcanzar seropositividad o concentraciones más altas de anticuerpos para los diferentes genotipos evaluados se incrementó con una menor edad de inicio de la inmunización, supresión o carga viral de VIH más baja, y niveles más altos de CD4 y CD8. La inmunización contra VPH produjo eventos adversos en 49-74% de participantes, siendo más de la mitad de ellos relacionados con dolor en la zona de punción. Los principales eventos adversos sistémicos fueron dolor de cabeza y fatiga transitorios. Eventos adversos graves asociados a la inmunización fueron muy poco frecuentes y resueltos sin necesidad de interrumpir el esquema de vacunación. Las GPC incluidas recomiendan vacunar contra VPH a la población infantil infectada con VIH independiente del sexo, excepto por OMS que recomienda inmunizar solo a niñas, aunque su recomendación no es explícita para población con VIH. La edad de inicio mayormente suele ser a los 9 años con un esquema de tres dosis.


Subject(s)
Humans , Child , Acquired Immunodeficiency Syndrome/physiopathology , Papillomavirus Infections/prevention & control , Alphapapillomavirus/immunology , Papillomavirus Vaccines/administration & dosage , Efficacy , Cost-Benefit Analysis
7.
Front Immunol ; 12: 742861, 2021.
Article in English | MEDLINE | ID: mdl-34759925

ABSTRACT

Background: Cervical cancer - caused by persistent High Risk Human Papilloma Virus (HR HPV) infections - is the second most common cancer affecting women globally. HIV infection increases the risk for HPV persistence, associated disease progression and malignant cell transformation. We therefore hypothesized that this risk increase is directly linked to HIV infection associated dysfunction or depletion of HPV-oncoprotein-specific T-cell responses. Methods: The 2H study specifically included HIV+ and HIV- women with and without cervical lesions and cancer to analyze HPV oncogene-specific T cell responses in relation to HPV infection, cervical lesion status and HIV status. Oncoprotein E6 and E7 specific T-cell responses were quantified for the most relevant types HPV16, 18 and 45 and control antigens (CMV-pp65) and M.tb-PPD in 373 women, using fresh peripheral blood mononuclear cells in an IFN-γ release ELISpot assay. Results: Overall, systemic E6- and E7-oncoprotein-specific T-cell responses were infrequent and of low magnitude, when compared to CMV-pp65 and M.tb-PPD (p < 0.001 for all HR HPV types). Within HIV negative women infected with either HPV16, 18 or 45, HPV16 infected women had lowest frequency of autologous-type-E6/E7-specific T-cell responses (33%, 16/49), as compared to HPV18 (46% (6/13), p = 0.516) and HPV45 (69% (9/13), p = 0.026) infected women. Prevalent HPV18 and 45, but not HPV16 infections were linked to detectable oncoprotein-specific T-cell responses, and for these infections, HIV infection significantly diminished T-cell responses targeting the autologous infecting genotype. Within women living with HIV, low CD4 T-cell counts, detectable HIV viremia as well as cancerous and precancerous lesions were significantly associated with depletion of HPV oncoprotein-specific T-cell responses. Discussion: Depletion of HPV-oncoprotein-specific T-cell responses likely contributes to the increased risk for HR HPV persistence and associated cancerogenesis in women living with HIV. The low inherent immunogenicity of HPV16 oncoproteins may contribute to the exceptional potential for cancerogenesis associated with HPV16 infections.


Subject(s)
Alphapapillomavirus/immunology , Coinfection/immunology , HIV Infections/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/immunology , T-Lymphocytes/immunology , Adult , Female , Humans , Middle Aged , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology
8.
Infect Genet Evol ; 95: 105084, 2021 11.
Article in English | MEDLINE | ID: mdl-34547435

ABSTRACT

OBJECTIVE: Human papillomavirus (HPV) is the main cause of cervical cancer, the 4th prominent cause of death in women globally. Previous vaccine development projects have led to several approved prophylactic vaccines available commercially, all of which are made using major capsid-based (L1). Administration of minor capsid protein (L2) gave rise to the second generation investigational prophylactic HPV vaccines, none of which are approved yet due to low immunogenicity provided by the L2 capsid protein. On the other hand, post-translation proteins, E6 and E7, have been utilized to develop experimental therapeutic vaccines. Here, in silico designing of a therapeutic and prophylactic vaccine against HPV16 is performed. METHODS: In this study, several immunoinformatic and computational tools were administered to identify and design a vaccine construct with dual prophylactic and therapeutic applications consisting of several epitope regions on L2, E6, and E7 proteins of HPV16. RESULTS: Immunodominant epitope regions (aa 12-23 and 78-78 of L2 protein, aa 11-27 of E6 protein, and aa 70-89 of E7 protein) were employed, which offered adequate immunogenicity to induce immune responses. Resuscitation-promoting factors (RpfB and RpfE) of Mycobacterium tuberculosis were integrated in two separate constructs as TLR4 agonists to act as vaccine adjuvants. Following physiochemical and structural evaluations carried out by various bioinformatics tools, the designed constructs were modeled and validated, resulting in two 3D structures. Molecular docking and molecular dynamic simulations suggested stable ligand-receptor interactions between the designed construct and TLR4. CONCLUSION: Ultimately, this study led to suggest the designed construct as a potential vaccine candidate with both prophylactic and therapeutic applications against HPV by promoting Th1, Th2, CTL, and B cell immune responses, which should be further confirmed in experimental studies.


Subject(s)
Alphapapillomavirus/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/chemistry , Vaccine Development , Molecular Docking Simulation , Molecular Dynamics Simulation , Papillomavirus Vaccines/pharmacology
9.
Nature ; 597(7875): 279-284, 2021 09.
Article in English | MEDLINE | ID: mdl-34471285

ABSTRACT

T cells are important in tumour immunity but a better understanding is needed of the differentiation of antigen-specific T cells in human cancer1,2. Here we studied CD8 T cells in patients with human papillomavirus (HPV)-positive head and neck cancer and identified several epitopes derived from HPV E2, E5 and E6 proteins that allowed us to analyse virus-specific CD8 T cells using major histocompatibility complex (MHC) class I tetramers. HPV-specific CD8 T cells expressed PD-1 and were detectable in the tumour at levels that ranged from 0.1% to 10% of tumour-infiltrating CD8 T lymphocytes (TILs) for a given epitope. Single-cell RNA-sequencing analyses of tetramer-sorted HPV-specific PD-1+ CD8 TILs revealed three transcriptionally distinct subsets. One subset expressed TCF7 and other genes associated with PD-1+ stem-like CD8 T cells that are critical for maintaining T cell responses in conditions of antigen persistence. The second subset expressed more effector molecules, representing a transitory cell population, and the third subset was characterized by a terminally differentiated gene signature. T cell receptor clonotypes were shared between the three subsets and pseudotime analysis suggested a hypothetical differentiation trajectory from stem-like to transitory to terminally differentiated cells. More notably, HPV-specific PD-1+TCF-1+ stem-like TILs proliferated and differentiated into more effector-like cells after in vitro stimulation with the cognate HPV peptide, whereas the more terminally differentiated cells did not proliferate. The presence of functional HPV-specific PD-1+TCF-1+CD45RO+ stem-like CD8 T cells with proliferative capacity shows that the cellular machinery to respond to PD-1 blockade exists in HPV-positive head and neck cancer, supporting the further investigation of PD-1 targeted therapies in this malignancy. Furthermore, HPV therapeutic vaccination efforts have focused on E6 and E7 proteins; our results suggest that E2 and E5 should also be considered for inclusion as vaccine antigens to elicit tumour-reactive CD8 T cell responses of maximal breadth.


Subject(s)
Alphapapillomavirus/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , Stem Cells/cytology , Alphapapillomavirus/isolation & purification , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/immunology , Humans , Lymphocytes, Tumor-Infiltrating/classification , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , RNA-Seq , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , Stem Cells/immunology , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , Transcription, Genetic
10.
Viruses ; 13(7)2021 07 17.
Article in English | MEDLINE | ID: mdl-34372596

ABSTRACT

Persistent infection with high-risk human papillomaviruses (HPVs) is the major risk factor associated with development of anogenital and oropharyngeal cancers. Initial infection by HPVs occurs into basal epithelial cells where viral genomes are established as nuclear episomes and persist until cleared by the immune response. Productive replication or amplification occurs upon differentiation and is dependent upon activation of the ataxia-telangiectasia mutated (ATM), ataxia telangiectasia and RAD3-related (ATR) DNA damage repair (DDR) pathways. In addition to activating DDR pathways, HPVs must escape innate immune surveillance mechanisms by antagonizing sensors, adaptors, interferons and antiviral gene expression. Both DDR and innate immune pathways are key host mechanisms that crosstalk with each other to maintain homeostasis of cells persistently infected with HPVs. Interestingly, it is still not fully understood why some HPV infections get cleared while others do not. Targeting of these two processes with antiviral therapies may provide opportunities for treatment of cancers caused by high-risk HPVs.


Subject(s)
Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , DNA Damage , DNA Repair , Host-Pathogen Interactions/immunology , Immunity, Innate , Cell Differentiation , Genome, Viral , Humans , Virus Replication
11.
Eur J Pharm Biopharm ; 168: 131-138, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34438020

ABSTRACT

Currently licensed vaccines require a cold-chain to maintain efficacy. This cold-chain requirement reduces the availability of vaccines in resource-poor areas of the world. Commercially available human papillomavirus (HPV) vaccines protect against the most common HPV types related to cervical cancer; however, their impact is limited in many regions due to cold-chain requirements. The goal of this study was to test the thermostability of an adjuvanted, trivalent HPV L1 capsomere-based vaccine (containing HPV types 16, 18, and 31) that was formulated by using lyophilization to embed the antigens within a solid, glassy matrix. Thermal stabilities were determined by storing the vaccine formulations for 3 months at 50 °C, followed by immunization of BALB/c mice and measurement of antibody responses. Antibody responses to capsomere vaccines formulated with alum were unchanged after storage for 3 months at 50 °C. Neutralizing responses to these vaccines were unchanged by high-temperature storage, and were equivalent to those generated after administration of the commercially available liquid HPV vaccine Gardasil®9.


Subject(s)
Alphapapillomavirus/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/chemistry , Animals , Capsid Proteins/immunology , Drug Stability , Drug Storage , Female , Humans , Mice , Mice, Inbred BALB C , Papillomavirus Infections/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Temperature , Time Factors
12.
Viruses ; 13(6)2021 06 08.
Article in English | MEDLINE | ID: mdl-34201028

ABSTRACT

Human papilloma virus (HPV) is the most common sexually transmitted infection worldwide causing a variety of benign and malignant conditions. A significant portion of the global population is infected with HPV, with the virus attributed to causing up to 5% of cancers worldwide. Bivalent, quadrivalent, and nine-valent vaccinations exist to aid in the prevention of these diseases and have been proven to be effective at preventing both benign and malignant disease. While vaccination is readily accessible in more developed countries, barriers exist to worldwide distribution and acceptance of vaccination. Vaccination and screening of HPV infection when used in combination are proven and predicted to decrease HPV related pathology. Improvements in vaccination formulations, for treatment as well as prevention, are actively being sought from a variety of mechanisms. Despite these advancements, and the data supporting their efficacy, there has been substantial delay in obtaining adequate vaccination coverage. In reviewing these challenges and looking forward to new vaccine development-especially within the current pandemic-it is clear from the challenges of HPV we require methods to more effectively encourage vaccination, ways to dispel vaccination myths as they occur, and implement better processes for vaccine distribution globally.


Subject(s)
Alphapapillomavirus/immunology , Papillomaviridae/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Vaccination , Female , Humans , Mass Screening , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Vaccine Development
13.
Front Immunol ; 12: 652054, 2021.
Article in English | MEDLINE | ID: mdl-34305889

ABSTRACT

Head and neck cancer (HNC) is the sixth most common malignancy worldwide; head and neck squamous cell carcinoma (HNSCC) account for the most cases of HNC. Past smoking and alcohol consumption are common risk factors of HNSCC; however, an increasing number of cases associated with human papillomavirus (HPV) infection have been reported in recent years. The treatment of HNSCC is integrated and multimodal including traditional surgery, radiotherapy, chemotherapy, and targeted therapy. Since pembrolizumab was approved in 2016, an increasing number of studies have focused on immunotherapy. However, not all of HNSCC patients have a better outcome on immunotherapy. Immunotherapy has been reported to be more effective in HPV-positive patients, but its molecular mechanism is still unclear. Some researchers have proposed that the high proportion of infiltrating immune cells in HPV-positive tumors and the difference in immune checkpoint expression level may be the reasons for their better response. As a result, a series of individualized immunotherapy trials have also been conducted in HPV-positive patients. This paper summarizes the current status of HNSCC immunotherapy, individualized immunotherapy in HPV-positive patients, and immune differences in HPV-positive tumors to provide new insights into HNSCC immunotherapy and try to identify patients who may benefit from immunotherapy.


Subject(s)
Head and Neck Neoplasms/therapy , Immunotherapy/methods , Neoplasm Recurrence, Local/therapy , Papillomavirus Infections/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Alphapapillomavirus/immunology , Antineoplastic Agents, Immunological/therapeutic use , Chemoradiotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/methods , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/virology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/virology , Neoplasm Staging , Papillomavirus Infections/immunology , Papillomavirus Infections/mortality , Papillomavirus Infections/virology , Progression-Free Survival , Randomized Controlled Trials as Topic , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/virology
14.
Biotechnol Lett ; 43(9): 1933-1944, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34313864

ABSTRACT

OBJECTIVES: Human papillomavirus infection (HPV) is the most common viral infection which is causes of cervical, penal, vulvar, anal and, oropharyngeal cancer. E7 protein of HPV is a suitable target for induction of T cell responses and controlling HPV-related cancer. The aim of the current study was to designed and evaluated a novel fusion protein containing the different E7 proteins of the HPV 16, 18, 6 and 11, linked to the cell-penetrating peptide HIV-1 Tat 49-57, in order to improve cytotoxic immune responses in in-vitro and in-vivo. RESULTS: In this study whole sequence of HPV16,18,6,11 E7-Tat (47-57) and HPV16,18,6,11 E7 cloned into the vector and expressed in E. coli (BL21). The purified protein was confirmed by SDS page and western blotting and then injected into the C57BL/6 mice. The efficiency of the fusion protein vaccine was assessed by antibody response assay, cytokine assay (IL-4 and IFN-γ), CD + 8 cytotoxicity assay and tumor challenge experiment. Result showed that fusion proteins containing Adjuvant (IFA,CFA) could express higher titer of antibody. Also, we showed that vaccination with E7-Tat and, E7-Tat-ADJ induced high frequencies of E7-specific CD8 + T cells and CD107a expression as well as IFN-γ level and enhanced long-term survival in the therapeutic animal models. CONCLUSION: Our finding suggested that this novel fusion protein vaccine was able to induce therapeutic efficacy and immunogenicity by improving CD8 + T cell in TC-1 tumor bearing mice; so this vaccine may be appreciated for research against HPV and tumor immunotherapies.


Subject(s)
Alphapapillomavirus/metabolism , HIV-1/genetics , Lung Neoplasms/virology , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Peptide Fragments/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/growth & development , Female , HIV-1/metabolism , Human papillomavirus 11/genetics , Human papillomavirus 11/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Human papillomavirus 18/genetics , Human papillomavirus 18/metabolism , Human papillomavirus 6/genetics , Human papillomavirus 6/metabolism , Humans , Lung Neoplasms/prevention & control , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism
15.
PLoS Med ; 18(6): e1003588, 2021 06.
Article in English | MEDLINE | ID: mdl-34097688

ABSTRACT

BACKGROUND: Cervical cancer elimination through human papillomavirus (HPV) vaccination programs requires the attainment of herd effect. Due to its uniquely high basic reproduction number, the vaccination coverage required to achieve herd effect against HPV type 16 exceeds what is attainable in most populations. We have compared how gender-neutral and girls-only vaccination strategies create herd effect against HPV16 under moderate vaccination coverage achieved in a population-based, community-randomized trial. METHODS AND FINDINGS: In 2007-2010, the 1992-1995 birth cohorts of 33 Finnish communities were randomized to receive gender-neutral HPV vaccination (Arm A), girls-only HPV vaccination (Arm B), or no HPV vaccination (Arm C) (11 communities per trial arm). HPV16/18/31/33/35/45 seroprevalence differences between the pre-vaccination era (2005-2010) and post-vaccination era (2011-2016) were compared between all 8,022 unvaccinated women <23 years old and resident in the 33 communities during 2005-2016 (2,657, 2,691, and 2,674 in Arms A, B, and C, respectively). Post- versus pre-vaccination-era HPV seroprevalence ratios (PRs) were compared by arm. Possible outcome misclassification was quantified via probabilistic bias analysis. An HPV16 and HPV18 seroprevalence reduction was observed post-vaccination in the gender-neutral vaccination arm in the entire study population (PR16 = 0.64, 95% CI 0.10-0.85; PR18 = 0.72, 95% CI 0.22-0.96) and for HPV16 also in the herpes simplex virus type 2 seropositive core group (PR16 = 0.64, 95% CI 0.50-0.81). Observed reductions in HPV31/33/35/45 seroprevalence (PR31/33/35/45 = 0.88, 95% CI 0.81-0.97) were replicated in Arm C (PR31/33/35/45 = 0.79, 95% CI 0.69-0.90). CONCLUSIONS: In this study we only observed herd effect against HPV16/18 after gender-neutral vaccination with moderate vaccination coverage. With only moderate vaccination coverage, a gender-neutral vaccination strategy can facilitate the control of even HPV16. Our findings may have limited transportability to other vaccination coverage levels. TRIAL REGISTRATION: ClinicalTrials.gov number NCT00534638, https://clinicaltrials.gov/ct2/show/NCT00534638.


Subject(s)
Alphapapillomavirus/immunology , Antibodies, Viral/blood , Immunity, Herd , Immunogenicity, Vaccine , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/prevention & control , Vaccination , Adolescent , Child , Cross-Sectional Studies , Female , Finland/epidemiology , Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Humans , Male , Papillomavirus Infections/epidemiology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Pregnancy , Randomized Controlled Trials as Topic , Seroepidemiologic Studies , Serologic Tests , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Young Adult
17.
Lancet Infect Dis ; 21(10): 1458-1468, 2021 10.
Article in English | MEDLINE | ID: mdl-34081923

ABSTRACT

BACKGROUND: Quadrivalent and bivalent vaccines against oncogenic human papillomavirus (HPV) are used worldwide with different reported overall efficacies against HPV infections. Although protective concentrations of vaccine-induced antibodies are still not formally defined, we evaluated the sustainability of neutralising antibodies in vaccine trial participants 2-12 years after vaccination and the correlation with reported vaccine efficacy. METHODS: We did a follow-up analysis of data from the Finnish cohorts of two international, randomised, double-blind, phase 3 trials of HPV vaccines, PATRICIA (bivalent, HPV16 and 18) and FUTURE II (quadrivalent, HPV6, 11, 16, and 18). In 2002 and 2004-05, respectively, Finnish girls aged 16-17 years participated in one of these two trials and consented to health registry follow-up with the Finnish Cancer Registry. The cohorts were also linked with the Finnish Maternity Cohort (FMC) that collects first-trimester serum samples from nearly all pregnant Finnish women, resulting in 2046 post-vaccination serum samples obtained during up to 12 years of follow-up. We obtained serum samples from the FMC-based follow-up of the FUTURE II trial (from the quadrivalent vaccine recipients) and the PATRICIA trial (from corresponding bivalent vaccine recipients who were aligned by follow-up time, and matched by the number of pregnancies). We assessed neutralising antibody concentrations (type-specific seroprevalence) to HPV6, 16, and 18, and cross-neutralising antibody responses to non-vaccine HPV types 31, 33, 45, 52, and 58 from 2 to 12 years after vaccination. FINDINGS: Up to Dec 31, 2016, we obtained and analysed 577 serum samples from the quadrivalent vaccine recipients and 568 from the bivalent vaccine recipients. In 681 first-pregnancy serum samples, neutralising antibodies to HPV6, 16, and 18 were generally found up to 12 years after vaccination. However, 51 (15%) of 339 quadrivalent vaccine recipients had no detectable HPV18 neutralising antibodies 2-12 years after vaccination, whereas all 342 corresponding bivalent vaccine recipients had HPV18 neutralising antibodies.. In seropositive quadrivalent vaccine recipients, HPV16 geometric mean titres (GMT) halved by years 5-7 (GMT 3679, 95% CI 2377 to 4708) compared with years 2-4 (6642, 2371 to 13 717). Between 5 and 12 years after vaccination, GMT of neutralising antibodies to HPV16 and 18 were 5·7 times and 12·4 times higher, respectively, in seropositive bivalent vaccine recipients than in the quadrivalent vaccine recipients. Cross-neutralising antibodies to HPV31, 33, 45, 52, and 58 were more prevalent in the bivalent vaccine recipients but, when measurable, sustainable up to 12 years after vaccination with similar GMTs in both vaccine cohorts. Seroprevalence for HPV16, 31, 33, 52, and 58 significantly correlated with vaccine efficacy against persistent HPV infections in the bivalent vaccine recipients only (rs=0·90, 95% CI 0·09 to 0·99, p=0·037, compared with rs=0·62, 95% CI -0·58 to 0·97, p=0·27 for the quadrivalent vaccine recipients). Correlation of protection with prevalence of neutralising or cross-neutralising HPV antibodies was not significant in the quadrivalent vaccine recipients. INTERPRETATION: The observed significant differences in the immunogenicity of the two vaccines are in line with the differences in their cross-protective efficacy. Protective HPV vaccine-induced antibody titres can be detected up to 12 years after vaccination. FUNDING: Academy of Finland and Finnish Cancer Foundation.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Papillomavirus Infections/blood , Papillomavirus Vaccines/immunology , Adolescent , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cohort Studies , Cross Reactions , Double-Blind Method , Female , Finland , Follow-Up Studies , Humans , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology
18.
Front Immunol ; 12: 626582, 2021.
Article in English | MEDLINE | ID: mdl-34054800

ABSTRACT

There is a need to increase the vaccine completion rates in women who have already received human papillomavirus (HPV) vaccines. With vaccines requiring multiple doses, designing a vaccination control program and increasing the proportion of women who complete vaccination are critical and remain as huge challenges. Currently, there are no published reports on the differences in the background characteristics between postpartum women who are vaccinated or unvaccinated against HPV. This study aimed to determine the vaccination rates of the second and third doses of HPV vaccination utilizing an achievable HPV vaccination program in postpartum women. In this retrospective study, 243 postpartum women attending Chiayi Chang Gung Memorial Hospital between March and September 2014 were enrolled. These women were classified into two groups: one group received the HPV vaccine under a practical, controlled postpartum HPV vaccination program, and the other group did not. The rates for the second and third rounds of HPV vaccination in postpartum women were calculated. The differences in the background characteristics between the two groups were determined using the Student's t test, chi-square test or Fisher's exact test, and the multiple logistic models, as appropriate. Under the controlled postpartum HPV vaccination program, the completion rate for the three doses of postpartum HPV vaccination was 97.2%. Significant differences were observed according to maternal age, gender of the newborn, and postpartum Pap smear results between the two groups in our study. In conclusion, the controlled postpartum HPV vaccination program is a reasonable method for achieving an excellent completion rate for the three doses of postpartum HPV vaccination and may be a good model for any multiple-dose vaccination protocol.


Subject(s)
Alphapapillomavirus/immunology , Immunization Programs , Immunization Schedule , Medication Adherence , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Postpartum Period , Uterine Cervical Neoplasms/prevention & control , Vaccination Refusal , Adolescent , Adult , Alphapapillomavirus/pathogenicity , Female , Humans , Middle Aged , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/adverse effects , Pregnancy , Retrospective Studies , Risk Factors , Time Factors , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Young Adult
19.
J Gen Virol ; 102(5)2021 05.
Article in English | MEDLINE | ID: mdl-34043499

ABSTRACT

Serum antibody levels can be used to measure the humoral immune response against human papillomaviruses (HPV). We developed and validated a rapid, technically simple and relatively inexpensive multiplex non-competitive Luminex-based immunoassay (ncLIA) to measure total IgG antibody levels against four HPV types. For the assay's solid phase, virus-like particles (VLPs) of HPV6, 11, 16 and 18 were bound to heparin-coated beads. HPV serum antibody levels binding to the VLPs were quantified using a phycoerithrin-conjugated secondary polyclonal donkey anti-human IgG antibody. Standardization and validation of the ncLIA were performed using 96 paired serum and genital samples from participants in the HITCH cohort study, including young women (aged 18-24 years) and their male sexual partners (aged 18+) in Montreal, Canada. Results from the ncLIA were compared to a validated Luminex immunoassay from PPD laboratories using Pearson's correlation coefficients, receiver operating characteristic curves and logistic regression. Our assay had good inter- and intra-assay variability. The correlation of serum antibody levels between the ncLIA and validation assay was highest for HPV16 and HPV11 (r=0.90), followed by HPV6 (r=0.86) and HPV18 (r=0.67). The ncLIA was better able to predict HPV DNA positivity in genital samples than the validation assay for HPV16 [area under the curve (AUC) 0.65 versus 0.52, P=0.001] and HPV18 [AUC 0.71 versus 0.57, P=0.024]. AUCs for HPV6 and HPV11 were similar between the two assays (0.70 versus 0.71, P=0.59, and 0.88 versus 0.96, P=0.08, respectively). The developed ncLIA is useful for measuring total IgG antibody response following natural infection or vaccination against four HPV VLPs included in the quadrivalent vaccine.


Subject(s)
Alphapapillomavirus/classification , Alphapapillomavirus/isolation & purification , Antibodies, Viral/blood , Papillomavirus Infections/diagnosis , Adolescent , Alphapapillomavirus/immunology , Canada , Cohort Studies , Female , Humans , Immunoassay , Immunoglobulin G/blood , Male , Papillomavirus Infections/blood , Papillomavirus Infections/virology , Reproducibility of Results , Sensitivity and Specificity , Serologic Tests , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...