Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
1.
BMC Biotechnol ; 24(1): 71, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350162

RESUMEN

BACKGROUND: Human papillomavirus type 16 (HPV-16) infection is strongly associated with considerable parts of cervical, neck, and head cancers. Performed investigations have had moderate clinical success, so research to reach an efficient vaccine has been of great interest. In the present study, the immunization potential of a newly designed HPV-16 construct was evaluated in a mouse model. RESULTS: Initially, a construct containing HPV-16 mutant (m) E6/E7 fusion gene was designed and antigen produced in two platforms (i.e., DNA vaccine and recombinant protein). Subsequently, the immunogenicity of these platforms was investigated in five mice) C57BL/6 (groups based on several administration strategies. Three mice groups were immunized recombinant protein, DNA vaccine, and a combination of them, and two other groups were negative controls. The peripheral blood mononuclear cells (PBMCs) proliferation, Interleukin-5 (IL-5) and interferon-γ (IFN-γ) cytokines, IgG1 and IgG2a antibody levels were measured. After two weeks, TC-1 tumor cells were injected into all mice groups, and subsequently further analysis of tumor growth and metastasis and mice survival were performed according to the schedule. Overall, the results obtained from in vitro immunology and tumor cells challenging assays indicated the potential of the mE6/E7 construct as an HPV16 therapeutic vaccine candidate. The results demonstrated a significant increase in IFN-γ cytokine (P value < 0.05) in the Protein/Protein (D) and DNA/Protein (E) groups. This finding was in agreement with in vivo assays. Control groups show a 10.5-fold increase (P value < 0.001) and (C) DNA/DNA group shows a 2.5-fold increase (P value < 0.01) in tumor growth compared to D and E groups. Also, a significant increase in survival of D and E (P value < 0.001) and C (P value < 0.01) groups were observed. CONCLUSIONS: So, according to the findings, the recombinant protein could induce stronger protection compared to the DNA vaccine form. Protein/Protein and DNA/Protein are promising administration strategies for presenting this construct to develop an HPV-16 therapeutic vaccine candidate.


Asunto(s)
Papillomavirus Humano 16 , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales , Proteínas E7 de Papillomavirus , Vacunas contra Papillomavirus , Proteínas Represoras , Vacunas de ADN , Animales , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Ratones , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Vacunas de ADN/inmunología , Vacunas de ADN/genética , Vacunas de ADN/administración & dosificación , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/administración & dosificación , Femenino , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
2.
Virol J ; 21(1): 213, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39252044

RESUMEN

High-risk human papilloma virus (HR-HPV) persistent infection is closely associated with the development of cervical cancer and squamous intraepithelial lesion (SIL).The α-9 HPVs, which is predominantly composed of HR-HPV types, account for 75% of HR-HPV infection in Sichuan. The oncoproteins E6 and E7 of HPV play a crucial role in tumor initiation and progression. Notably, HPV-35 is the only HR-HPV type within the α-9 genus that is not included in the nine-valent HPV prophylactic vaccine. Cervical cell samples obtained from Sichuan were collected for HPV detection and genotyping. Among the 406 HPV-positive samples, 31 HPV-35 were detected, 24 HPV-35 E6 and 26 E7 were successfully amplified and sequenced, five nucleotide mutations in E6 and three in E7 were detected, T232C, T434G of E6 (W78R, I145R) and C67T, G84T of E7 (H23Y, L28F) were non-synonymy mutation. PAML 4.8 server was used to detect positive selection sites of HPV-35 E6, E7, and E6 is W78R. Phyre2 were used to predict and analyze protein structures, W78R made influences on protein structure. IEDB were used to screen epitopes vaccine target for HPV-35 affection therapy, and 5 HPV-35 E6 and 3 HPV-35 E7 most potential epitopes were obtained, the most potential peptides for therapy vaccine design were 79-91YRYSVYGETLEKQ, 45-60FACYDLCIVREGQPY, 124-135RFHNIGGRWTGR of E6; 3-19GEITTLQDYVLDLEPEA, 38-47TIDGPAGQAK, 70-88VQSTHIDIRKLEDLLMGTF of E7 and W78R mainly decreased the epitopes affinity.Conclusions Amino acid substitution in the positive selection sites of HPV-35 E6 and E7 genes have been found to influence protein structure and to decrease the overall affinity of antigen epitopes. This observation aligns with the evolutionary significance of positive selection site, which may confer advantages to the virus by making infected cells more challenging for the immune system to detect, thereby enhancing HPV's adaptability to the host environment. The polymorphism analysis of HPV-35 E6, E7 contributes to the enrichment of α-9 HPV data in Sichuan China, which is instrumental in improving the effectiveness of clinical detection. Furthermore, these findings provide a relevant theoretical foundation for the prevention and treatment of HPV-related diseases.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Humanos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Femenino , China , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/genética , Polimorfismo Genético , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Genotipo , Adulto , Neoplasias del Cuello Uterino/virología , Neoplasias del Cuello Uterino/prevención & control , Epítopos/inmunología , Epítopos/genética , Alphapapillomavirus/genética , Alphapapillomavirus/inmunología , Alphapapillomavirus/clasificación , Persona de Mediana Edad , Mutación , Virus del Papiloma Humano
3.
Viruses ; 16(9)2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39339857

RESUMEN

HPV16 is occasionally detected in vaccinated women who received the bivalent HPV16/18 vaccine, usually at low viral loads. This study explored potential differences in HPV16 variants between vaccinated and unvaccinated women. HPV16-postive viral loads were detected in 1.9% (17/875) and 13% (162/760) of vaccinated and unvaccinated women, respectively, showcasing the vaccine's high efficacy. The L1, E6, and URR regions of HPV16 were sequenced from genital swabs from 16 vaccinated and 25 unvaccinated women in the HAVANA (HPV Among Vaccinated And Non-vaccinated Adolescents) study. The majority of HPV16 variants from vaccinated and unvaccinated women clustered similarly with sub-lineages A1 and A2. Additionally, a separate cluster within lineage A was found, with the variants sharing the L1-located SNP A753G (synonymous) and the URR-located SNP T340C, which did not occur in the other variants. Furthermore, four variants from vaccinated women had relatively long branches, but were not characterized by specific SNPs. The frequency of G712A in the URR was the only SNP observed to be marginally higher among vaccinated women than unvaccinated women. Non-synonymous SNPs T266A in the FG-loop of L1 and L83V in E6 were common among variants from vaccinated and unvaccinated women, but present in similar frequencies. In conclusion, the detection of HPV16 in vaccinated (and unvaccinated) women seemed to be the result of random circulation within this study population.


Asunto(s)
Variación Genética , Papillomavirus Humano 16 , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Proteínas Represoras , Humanos , Femenino , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Adolescente , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Eficacia de las Vacunas , Vacunación , Filogenia , Carga Viral , Polimorfismo de Nucleótido Simple , Secuencias Reguladoras de Ácidos Nucleicos , Adulto Joven
4.
Clin Exp Med ; 24(1): 199, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39196444

RESUMEN

Replication-defective viral vector vaccines have several advantages over conventional subunit vaccines, including potent antibody responses, cellular responses critical for eliminating pathogen-infected cells, and the induction of highly immunogenic and durable immune responses without adjuvants. The Human papillomavirus (HPV), a microorganism with over 200 genotypes, plays a crucial role in inducing human tumors, with the majority of HPV-related malignancies expressing HPV proteins. Tumors associated with HPV infection, most of which result from HPV16 infection, include those affecting the cervix, anus, vagina, penis, vulva, and oropharynx. In recent years, the development of therapeutic HPV vaccines utilizing viral vectors for the treatment of premalignant lesions or tumors caused by HPV infection has experienced rapid growth, with numerous research pipelines currently underway. Simultaneously, screening for optimal antigens requires more basic research and more optimized methods. In terms of preclinical research, we present the various models used to assess vaccine efficacy, highlighting their respective advantages and disadvantages. Further, we present current research status of therapeutic vaccines using HPV viral vectors, especially the indications, initial efficacy, combination drugs, etc. In general, this paper summarizes current viral vector therapeutic HPV vaccines in terms of HPV infection, antigen selection, vectors, efficacy evaluation, and progress in clinical trials.


Asunto(s)
Vectores Genéticos , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/administración & dosificación , Humanos , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/terapia , Animales , Femenino , Ensayos Clínicos como Asunto
5.
Vaccine ; 42(24): 126245, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39216181

RESUMEN

Persistent infection with high-risk human papillomavirus (HPV) types can lead to the development of cancer in HPV-infected tissues, including the cervix, oropharynx, anus, penis, vagina, and vulva. While current HPV vaccines cover approximately 90 % of cervical cancers, nearly 10 % of cases associated with HPV types not included in the vaccines remain unaddressed, notably HPV59. This study describes the development of a chimeric virus-like particle (VLP) targeting HPV18/45/59, proposed as a vaccine candidate for high-risk HPV type (HPV59) currently lacking commercial vaccines. Given that the majority of neutralizing antibody epitopes are located on the surface loops, we engineered a strategic swap of these loops between the closely related HPV18 and HPV45. This methodology was then extended to incorporate surface loops of HPV59, resulting in the lead candidate construct of the H18-45BCEF-59HI chimeric VLP with two surface loops swapping from HPV45 to HPV18. Characterization confirmed that H18-45BCEF-59HI closely resembled the wild-type (WT) backbone types in particle size and morphology, as verified by Transmission Electron Microscopy (TEM), High-Performance Size-Exclusion Chromatography (HPSEC), and Analytical Ultracentrifugation (AUC), and demonstrated similar thermal stability as evidenced by Differential Scanning Calorimetry (DSC). Immunization studies in mice with the chimeric VLPs assessed their immunogenicity, revealing that the H18-45EF-59HI chimeric VLP exhibited optimal cross-neutralization. Additionally, when produced in a Good Manufacturing Practice (GMP)-like facility, the H18-45BCEF-59HI VLP was selected as a promising vaccine candidate for the prevention of HPV18/45/59 infection. This study not only offers a potential solution to the current vaccination gap but also provides a foundational approach for the design of vaccines targeting viruses with multiple subtypes or variants.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas de Partículas Similares a Virus , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/inmunología , Femenino , Vacunas de Partículas Similares a Virus/inmunología , Animales , Humanos , Ratones , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Papillomavirus Humano 18/inmunología , Papillomavirus Humano 18/genética , Virus del Papiloma Humano
6.
Life Sci Alliance ; 7(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38514186

RESUMEN

Human papillomavirus (HPV) infections account for several human cancers. There is an urgent need to develop therapeutic vaccines for targeting preexisting high-risk HPV (such as HPV 16 and 18) infections and lesions, which are insensitive to preventative vaccines. In this study, we developed a lipid nanoparticle-formulated mRNA-based HPV therapeutic vaccine (mHTV), mHTV-02, targeting the E6/E7 of HPV16 and HPV-18. mHTV-02 dramatically induced antigen-specific cellular immune response and robust memory T-cell immunity in mice, besides significant CD8+ T-cell infiltration and cytotoxicity in TC-1 tumors expressing HPV E6/E7, resulting in tumor regression and prolonged survival in mice. Moreover, evaluation of routes of administration found that intramuscular or intratumoral injection of mHTV-02 displayed significant therapeutic effects. In contrast, intravenous delivery of the vaccine barely showed any benefit in reducing tumor size or improving animal survival. These data together support mHTV-02 as a candidate therapeutic mRNA vaccine via specific administration routes for treating malignancies caused by HPV16 or HPV18 infections.


Asunto(s)
Neoplasias , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Ratones , Animales , Humanos , Vacunas de ARNm , Infecciones por Papillomavirus/prevención & control , Proteínas E7 de Papillomavirus/genética , Neoplasias/terapia , Vacunas contra Papillomavirus/genética
7.
Mol Cancer ; 23(1): 46, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459592

RESUMEN

Nucleic acid vaccines have shown promising potency and efficacy for cancer treatment with robust and specific T-cell responses. Improving the immunogenicity of delivered antigens helps to extend therapeutic efficacy and reduce dose-dependent toxicity. Here, we systematically evaluated chemokine-fused HPV16 E6/E7 antigen to improve the cellular and humoral immune responses induced by nucleotide vaccines in vivo. We found that fusion with different chemokines shifted the nature of the immune response against the antigens. Although a number of chemokines were able to amplify specific CD8 + T-cell or humoral response alone or simultaneously. CCL11 was identified as the most potent chemokine in improving immunogenicity, promoting specific CD8 + T-cell stemness and generating tumor rejection. Fusing CCL11 with E6/E7 antigen as a therapeutic DNA vaccine significantly improved treatment effectiveness and caused eradication of established large tumors in 92% tumor-bearing mice (n = 25). Fusion antigens with CCL11 expanded the TCR diversity of specific T cells and induced the infiltration of activated specific T cells, neutrophils, macrophages and dendritic cells (DCs) into the tumor, which created a comprehensive immune microenvironment lethal to tumor. Combination of the DNA vaccine with anti-CTLA4 treatment further enhanced the therapeutic effect. In addition, CCL11 could also be used for mRNA vaccine design. To summarize, CCL11 might be a potent T cell enhancer against cancer.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Proteínas Oncogénicas Virales , Vacunas contra Papillomavirus , Vacunas de ADN , Animales , Ratones , Vacunación Basada en Ácidos Nucleicos , Vacunas de ADN/genética , Vacunas contra Papillomavirus/genética , Neoplasias/genética , Neoplasias/terapia , Linfocitos T CD8-positivos , Proteínas E7 de Papillomavirus/genética , Proteínas Oncogénicas Virales/genética , Ratones Endogámicos C57BL , Microambiente Tumoral
8.
Life Sci ; 340: 122456, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38266814

RESUMEN

AIMS: Human papillomavirus (HPV) infections are highly prevalent globally. While preventive HPV vaccines exist, therapeutic vaccines are needed to treat existing HPV lesions and malignancies. This study evaluated the immunostimulatory and anti-tumor effects of three therapeutic vaccine candidates based on the recombinant protein, tumor cell lysate (TCL), and engineered exosome (Exo) harboring the heat shock protein 27 (Hsp27)-E7 fusion construct in mouse model. MAIN METHODS: At first, the recombinant Hsp27-E7 protein was generated in E. coli expression system. Then, tumor cell lysates-based and engineered exosomes-based vaccine constructs harboring green fluorescent protein (GFP) and Hsp27-E7 were produced using lentiviral system. Finally, their immunological and antitumor effects were investigated in both prophylactic and therapeutic experiments. KEY FINDINGS: Our data showed that the recombinant Hsp27-E7 protein, TCL-Hsp27-E7 and Exo-Hsp27-E7 regimens can induce the highest level of IFN-γ, TNF-α and Granzyme B, respectively. The percentage of tumor-free mice was identical for three vaccine strategies (survival rate: 75 %) in both prophylactic and therapeutic experiments. Generally, the TCL-Hsp27-E7, Exo-Hsp27-E7 and recombinant Hsp27-E7 protein regimens induced effective immune responses toward Th1 and CTL activity, and subsequently antitumor effects in mouse model. SIGNIFICANCE: Regarding to higher Granzyme B secretion, lower tumor growth and more safety, the Exo-Hsp27-E7 regimen can be considered as the most promising HPV vaccination strategy.


Asunto(s)
Exosomas , Neoplasias , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Humanos , Animales , Ratones , Vacunas contra Papillomavirus/genética , Granzimas/metabolismo , Proteínas de Choque Térmico HSP27 , Exosomas/metabolismo , Infecciones por Papillomavirus/prevención & control , Escherichia coli/metabolismo , Proteínas E7 de Papillomavirus/genética , Ratones Endogámicos C57BL
9.
Infect Dis (Lond) ; 56(1): 66-72, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37994805

RESUMEN

BACKGROUND: Human papillomavirus (HPV) infection contributes to approximately 5% of the worldwide cancer burden. The three-dose HPV vaccine has demonstrated immunogenicity and efficacy. Humoral responses may be critical for preventing, controlling, and/or eliminating HPV infection. Using data from the HITCH cohort, we analysed humoral immune response to HPV vaccination among women in relation to the phylogenetic relatedness of HPV genotypes. METHODS: We included 96 women aged 18-24 years attending college or university in Montreal, Canada. Participants provided blood samples at enrolment and five follow-up visits. Antibody response to bacterially expressed L1 and E6 glutathione S-transferase fusion proteins of multiple Alphapapillomavirus types, and to virus-like particles (VLP-L1) of HPV16 and HPV18 were measured using multiplex serology. We assessed correlations between antibody seroreactivities using Pearson correlations (r). RESULTS: At enrolment, 87.7% of participants were unvaccinated, 2.4% had received one, 3.2% two, and 6.7% three doses of HPV vaccine. The corresponding L1 seropositivity to any HPV was 41.2%, 83.3%, 100%, and 97.0%. Between-type correlations for L1 seroreactivities increased with the number of vaccine doses, from one to three. Among the latter, the strongest correlations were observed for HPV58-HPV33 (Pearson correlation [r] = 0.96; α9-species); HPV11-HPV6 (r = 0.96; α10-species); HPV45-HPV18 (r = 0.95; α7-species), and HPV68-HPV59 (r = 0.95; α7-species). CONCLUSIONS: Correlations between HPV-specific antibody seroreactivities are affected by phylogenetic relatedness, with anti-L1 correlations becoming stronger with the number of vaccine doses received.


Asunto(s)
Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Femenino , Humanos , Virus del Papiloma Humano , Infecciones por Papillomavirus/prevención & control , Estudios de Cohortes , Filogenia , Anticuerpos Antivirales , Papillomavirus Humano 18 , Vacunación , Papillomaviridae/genética , Vacunas contra Papillomavirus/genética , Genotipo
10.
Microbiol Immunol ; 68(2): 65-74, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38105559

RESUMEN

Cervical cancer is caused mostly by human papillomavirus (HPV), and several HPV vaccines have been developed to prevent its onset. Vaccines include antigens as well as adjuvants, with adjuvants playing an important role in activating the innate immune responses necessary for inducing adaptive immunological responses. Recent research has shown the presence of trained immunity inside the innate immune system. However, trained immunity conferred by HPV vaccinations is not well understood. In this work, we explored the innate immune responses and trained immunity caused by two HPV vaccines, Cervarix and Gardasil. Cervarix includes monophosphoryl lipid A and an aluminum adjuvant, and it significantly increased the expression of IL-6 and IFN-ß mRNAs in RAW264.7 cells. On the contrary, Gardasil, which only includes an aluminum adjuvant, exhibited little cytokine expression but increased the expression of TLRs. Furthermore, Cervarix significantly increased IL-1ß secretion from mouse macrophages, while Gardasil only mildly induced IL-1ß secretion. Interestingly, initial stimulation with Gardasil enhanced the expression of IL-6 and TNF-α mRNAs upon secondary stimulation with TLR ligands, indicating that Gardasil induced trained immunity in macrophages. Moreover, Gardasil injection into mice resulted in enhanced TNF-α production in sera following secondary TLR stimulation. Our findings suggest that HPV vaccinations have the ability to induce trained immunity that modulate TLR ligand responses.


Asunto(s)
Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Humanos , Animales , Ratones , Citocinas , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18 , Factor de Necrosis Tumoral alfa , Interleucina-6/genética , Inmunidad Entrenada , Infecciones por Papillomavirus/prevención & control , Aluminio , Vacunas contra Papillomavirus/genética , Adyuvantes Inmunológicos , Receptores Toll-Like
11.
BMC Res Notes ; 16(1): 164, 2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37550734

RESUMEN

INTRODUCTION: In humans, approximately 5% of all cancers are attributable to HPV infection. Prophylactic vaccines can inhibit viral migration and persistence. However, further studies are still required to develop such treatments. To achieve this goal, we designed a therapeutic HPV DNA vaccine encoding a construct of E6/E7/L1 and used NSP4 antigen as an adjuvant to assess the efficiency of this construct in generating antigen-specific antitumor immune responses. MATERIALS AND METHODS: Sixty female C57BL/6 mice (6-8 weeks old) were purchased from the Institute Pasteur of Iran. Through a subcutaneous (s.c) injection of a suspension of 100 µl PBS containing 106 TC-1 cells/mouse in the back side, 30 of them became cancerous, while 30 of them were healthy control mice. To amplify E6/E7/L1-pcDNA3 and NSP4-pcDNA3, the competent cells of DH5α and to generate a tumor, TC-1 cell line was used. Mice were then immunized with the HPV DNA vaccine. Cell proliferation was assessed by MTT assay. Finally, cytokine responses (IL-4, IL-12, IFN- γ) were measured in the supernatant of mice spleen cells. RESULT: Mice receiving the NSP4/E6-E7-L1 vaccine had the highest stimulatory index compared to other groups, although it was not statistically significant. Interleukin 4/12 and IFN-γ production were significantly higher in E6-E7-L1 / NSP4 group and E6-E7-L1 group compared to other groups (P < 0.05). Among different groups, E6/E7/L1 + NSP4 group was able to slow down the tumor growth process, but it was not significant (p > 0.05). Among the aforementioned cytokines, IFN-γ and IL-12 are among the cytokines that stimulate the Th1 pathway and IL-4 cytokine stimulates the Th2 pathway and B lymphocytes. CONCLUSION: Our data revealed that the present vaccine can reduce tumor size, and cytokine measurement showed that it stimulates innate and acquired immune responses, thus it can be a therapeutic vaccine in the tumor-bearing mice model.


Asunto(s)
Neoplasias , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas de ADN , Humanos , Femenino , Animales , Ratones , Vacunas de ADN/genética , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Linfocitos T Citotóxicos , Interleucina-4 , Infecciones por Papillomavirus/prevención & control , Ratones Endogámicos C57BL , Vacunas contra Papillomavirus/genética , Adyuvantes Inmunológicos , ADN , Citocinas , Interleucina-12
12.
J Mol Biol ; 435(13): 168096, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37086948

RESUMEN

Human papilloma virus (HPV) infections are associated with almost all cervical cancers and to a lower extend also with anogenital or oropharyngeal cancers. HPV proteins expressed in HPV-associated tumors are attractive antigens for cancer vaccination strategies as self-tolerance, which is associated with most endogenous tumor-associated antigens, does not need to be overcome. In this study, we generated a live attenuated cancer vaccine based on the chimeric vesicular stomatitis virus VSV-GP, which has previously proven to be a potent vaccine vector and oncolytic virus. Genes at an earlier position in the genome more to the 3' end are expressed stronger compared to genes located further downstream. By inserting an HPV16-derived antigen cassette consisting of E2, E6 and E7 into VSV-GP either at first (HPVp1) or fifth (HPVp5) position in VSV-GP's genome we aimed to analyze the effect of vaccine antigen position and consequently expression level on viral fitness, immunogenicity, and anti-tumoral efficacy in a syngeneic mouse tumor model. HPVp1 expressed higher amounts of HPV antigens compared to HPVp5 in vitro but had a slightly delayed replication kinetic which overall translated into increased HPV-specific T cell responses upon vaccination of mice. Immunization with both vectors protected mice in prophylactic and in therapeutic TC-1 tumor models with HPVp1 being more effective in the prophylactic setting. Taken together, VSV-GP is a promising candidate as therapeutic HPV vaccine and first position of the vaccine antigen in a VSV-derived vector seems to be superior to fifth position.


Asunto(s)
Neoplasias , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vesiculovirus , Animales , Humanos , Ratones , Virus del Papiloma Humano , Ratones Endogámicos C57BL , Neoplasias/terapia , Neoplasias/virología , Infecciones por Papillomavirus/terapia , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/uso terapéutico , Vacunas Atenuadas , Neoplasias Experimentales
13.
Sci Transl Med ; 15(686): eabn3464, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36867683

RESUMEN

As mRNA vaccines have proved to be very successful in battling the coronavirus disease 2019 (COVID-19) pandemic, this new modality has attracted widespread interest for the development of potent vaccines against other infectious diseases and cancer. Cervical cancer caused by persistent human papillomavirus (HPV) infection is a major cause of cancer-related deaths in women, and the development of safe and effective therapeutic strategies is urgently needed. In the present study, we compared the performance of three different mRNA vaccine modalities to target tumors associated with HPV-16 infection in mice. We generated lipid nanoparticle (LNP)-encapsulated self-amplifying mRNA as well as unmodified and nucleoside-modified non-replicating mRNA vaccines encoding a chimeric protein derived from the fusion of the HPV-16 E7 oncoprotein and the herpes simplex virus type 1 glycoprotein D (gDE7). We demonstrated that single low-dose immunizations with any of the three gDE7 mRNA vaccines induced activation of E7-specific CD8+ T cells, generated memory T cell responses capable of preventing tumor relapses, and eradicated subcutaneous tumors at different growth stages. In addition, the gDE7 mRNA-LNP vaccines induced potent tumor protection in two different orthotopic mouse tumor models after administration of a single vaccine dose. Last, comparative studies demonstrated that all three gDE7 mRNA-LNP vaccines proved to be superior to gDE7 DNA and gDE7 recombinant protein vaccines. Collectively, we demonstrated the immunogenicity and therapeutic efficacy of three different mRNA vaccines in extensive comparative experiments. Our data support further evaluation of these mRNA vaccines in clinical trials.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas de ADN , Animales , Femenino , Ratones , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Inmunización , Ratones Endogámicos C57BL , Neoplasias/terapia , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/genética , Proteínas Recombinantes , ARN Mensajero/genética
14.
Adv Exp Med Biol ; 1407: 85-103, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36920693

RESUMEN

Papillomavirus is difficult to culture in vitro, which limits its related research. The development of pseudotyped virus technology provides a valuable research tool for virus infectivity research, vaccine evaluation, infection inhibitor evaluation, and so on. Depending on the application fields, different measures have been developed to generate various kinds of pseudotyped papillomavirus. L1-based and L2-based HPV vaccines should be evaluated using different pseudotyped virus system. Pseudotyped papillomavirus animal models need high-titer pseudotyped virus and unique handling procedure to generate robust results. This paper reviewed the development, optimization, standardization, and application of various pseudotyped papillomavirus methods.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Animales , Proteínas de la Cápside/genética , Proteínas Oncogénicas Virales/genética , Pseudotipado Viral , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/uso terapéutico , Infecciones por Papillomavirus/prevención & control , Anticuerpos Antivirales , Papillomaviridae/genética
15.
Proc Natl Acad Sci U S A ; 120(5): e2215091120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36696444

RESUMEN

A foundational principle of rational vaccinology is that vaccine structure plays a critical role in determining therapeutic efficacy, but in order to establish fundamental, effective, and translatable vaccine design parameters, a highly modular and well-defined platform is required. Herein, we report a DNA dendron vaccine, a molecular nanostructure that consists of an adjuvant DNA strand that splits into multiple DNA branches with a varied number of conjugated peptide antigens that is capable of dendritic cell uptake, immune activation, and potent cancer killing. We leveraged the well-defined architecture and chemical modularity of the DNA dendron to study structure-function relationships that dictate molecular vaccine efficacy, particularly regarding the delivery of immune-activating DNA sequences and antigenic peptides on a single chemical construct. We investigated how adjuvant and antigen placement and number impact dendron cellular uptake and immune activation, in vitro. These parameters also played a significant role in raising a potent and specific immune response against target cancer cells. By gaining this structural understanding of molecular vaccines, DNA dendrons successfully treated a mouse cervical human papillomavirus TC-1 cancer model, in vivo, where the vaccine structure defined its efficacy; the top-performing design effectively reduced tumor burden (<150 mm3 through day 30) and maintained 100% survival through 44 d after tumor inoculation.


Asunto(s)
Vacunas contra el Cáncer , Dendrímeros , Vacunas contra Papillomavirus , Neoplasias del Cuello Uterino , Vacunas de ADN , Animales , Femenino , Ratones , Humanos , Dendrímeros/farmacología , Neoplasias del Cuello Uterino/prevención & control , ADN , Péptidos , Vacunas contra Papillomavirus/genética
16.
Prep Biochem Biotechnol ; 53(2): 148-156, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35302435

RESUMEN

Cervical cancer caused by Human papillomavirus (HPV) is one of the most common causes of cancer death in women worldwide. Even though the disease can be avoided by immunization, the expensive price of HPV vaccines makes it hard to be accessed by women in middle-low-income countries. Thus, the development of generic HPV vaccines is needed to address inequalities in life-saving access. This study aimed to develop the HPV52 L1 VLP-based recombinant vaccine using Pichia pastoris expression system. The l1 gene was codon-optimized based on P. pastoris codon usage resulting CAI value of 0.804. The gene was inserted into the pD902 plasmid under the regulation of the AOX1 promoter. The linear plasmid was transformed into P. pastoris BG10 genome and screened in YPD medium containing zeocin antibiotic. Colony of transformant that grown on highest zeocin concentration was characterized by genomic PCR and sequencing. The positive clone was selected and expressed using BMGY/BMMY medium induced with various methanol concentrations. The SDS-PAGE and Western blot analyses showed that 55 kDa L1 protein was successfully expressed using an optimum concentration of 1% methanol. The self-assembly of HPV52 L1 protein was also proven using TEM analysis. Moreover, we also analyzed the B-cell epitope of HPV52 L1 protein based on several criteria, including antigenicity, surface accessibility, flexibility, and hydrophilicity. We assumed that epitope 476GLQARPKLKRPASSAPRTSTKKKKV500 could be developed as an epitope-based vaccine with a neutralizing antibody response toward HPV52 infection. Finally, our study provided the alternative for developing low-cost HPV vaccines, either VLP or epitope-based.


Asunto(s)
Virus del Papiloma Humano , Vacunas contra Papillomavirus , Femenino , Humanos , Metanol/metabolismo , Proteínas de la Cápside/genética , Pichia/genética , Pichia/metabolismo , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/metabolismo , Epítopos/metabolismo , Codón/metabolismo
17.
Virology ; 575: 63-73, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36070626

RESUMEN

Human alphapapillomaviruses (αHPV) infect genital mucosa, and a high-risk subset is a necessary cause of cervical cancer. Licensed L1 virus-like particle (VLP) vaccines offer immunity against the nine most common αHPV associated with cervical cancer and genital warts. However, vaccination with an αHPV L2-based multimer vaccine, α11-88x5, protected mice and rabbits from vaginal and skin challenge with diverse αHPV types. While generally clinically inapparent, human betapapillomaviruses (ßHPV) are possibly associated with cutaneous squamous cell carcinoma (CSCC) in epidermodysplasia verruciformis (EV) and immunocompromised patients. Here we show that α11-88x5 vaccination protected wild type and EV model mice against HPV5 challenge. Passive transfer of antiserum conferred protection independently of Fc receptors (FcR) or Gr-1+ phagocytes. Antisera demonstrated robust antibody titers against ten ßHPV by L1/L2 VLP ELISA and neutralized and protected against challenge by 3 additional ßHPV (HPV49/76/96). Thus, unlike the licensed vaccines, α11-88x5 vaccination elicits broad immunity against αHPV and ßHPV.


Asunto(s)
Alphapapillomavirus , Betapapillomavirus , Carcinoma de Células Escamosas , Epidermodisplasia Verruciforme , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Neoplasias Cutáneas , Neoplasias del Cuello Uterino , Vacunas de Partículas Similares a Virus , Animales , Betapapillomavirus/genética , Proteínas de la Cápside , Epidermodisplasia Verruciforme/prevención & control , Femenino , Humanos , Sueros Inmunes , Ratones , Vacunas contra Papillomavirus/genética , Conejos , Receptores Fc , Vacunación
18.
Vaccine ; 40(42): 6141-6152, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36117002

RESUMEN

Persistent infection of high-risk human papillomavirus (HPV) is a leading cause of some cancers, including cervical cancer. However, with over 20 carcinogenic HPV types, it is difficult to design a multivalent vaccine that can offer complete protection. Here, we describe the design and optimization of a HPV51/69/26 triple-type chimeric virus-like particle (VLP) for vaccine development. Using E. coli and a serial N-terminal truncation strategy, we created double- and triple-type chimeric VLPs through loop-swapping at equivalent surface loops. The lead candidate, H69-51BC-26FG, conferred similar particulate properties as that of its parental VLPs and comparable immunogenicity against HPV51, -69 and -26. When produced in a GMP-like facility, these H69-51BC-26FG VLPs were verified to have excellent qualities for the development of a multivalent HPV vaccine. This study showcases an amenable way to create a single VLP using type-specific epitope clustering for the design of a triple-type vaccine.


Asunto(s)
Alphapapillomavirus , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas de Partículas Similares a Virus , Animales , Anticuerpos Antivirales , Proteínas de la Cápside , Epítopos , Escherichia coli/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Papillomaviridae/genética , Vacunas contra Papillomavirus/genética , Vacunas Combinadas , Vacunas de Partículas Similares a Virus/genética
19.
Sci Rep ; 12(1): 9521, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35681036

RESUMEN

Cervical cancer is the most common gynaecological cancer and reaches an alarming stage. HPVs are considered the main causative agents for cervical cancer and other sexually transmitted infections across the globe. Currently, three prophylactic vaccines are available against HPV infections with no therapeutic values. Due to a lack of effective therapeutic and prophylactic measures, the HPV infection is spreading in an uncontrolled manner. Next-generation of vaccine is needed to have both prophylactic and therapeutic values against HPV. Here first time we have designed a multi-epitope chimeric vaccine using the most oncogenic strain HPV 16 and HPV 18 through an immunoinformatic approach. In this study, we have used the L1, E5, E6 and E7 oncoproteins from both HPV 16 and HPV 18 strains for epitope prediction. Our recombinant chimeric vaccine construct consists, selected helper and cytotoxic T cell epitopes. Our computational analysis suggests that this chimeric construct is highly stable, non-toxic and also capable of inducing both cell-mediated and humoral immune responses. Furthermore, in silico cloning of the multi-epitope chimeric vaccine construct was done and the stabilization of the vaccine construct is validated with molecular dynamics simulation studies. Finally, our results indicated that our construct could be used for an effective prophylactic and therapeutic vaccine against HPV.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Neoplasias del Cuello Uterino , Epítopos de Linfocito T/genética , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 18 , Humanos , Papillomaviridae , Vacunas contra Papillomavirus/genética , Vacunas Sintéticas
20.
Epidemics ; 39: 100584, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35636314

RESUMEN

Human papillomaviruses are common sexually transmitted infections, caused by a large diversity of genotypes. In the context of vaccination against a subgroup of genotypes, better understanding the role of genotype interactions and human sexual behavior on genotype dynamics is essential. Herein, we present an individual-based model that integrates realistic heterosexual partnership behaviors and simulates interactions between vaccine and non-vaccine genotypes. Genotype interactions were considered, assuming a previous vaccine-genotype infection shortened (competition) or extended (synergy) the duration of a secondary non-vaccine-genotype infection. Sexual behavior determined papillomavirus acquisition and transmission: only 19.5% of active individuals at most 1 partner r during the year, but > 80% of those with ≥ 2 partners, were infected before vaccine introduction. The pre-vaccination situation was consistent with all genotype interaction scenarios. These genotype interactions, despite being undetectable during the pre-vaccination era, markedly impacted genotype prevalence after vaccination started, with a significant increase/decrease of non-vaccine genotypes prevalence for respectively competitive/synergistic interactions. These prevalence changes were more pronounced in individuals with ≤ 3 partners per year (up to 30% of prevalence modification assuming 65% vaccine coverage) but barely visible for individuals with > 3 partners per year (at most 0.30%). Results suggest the presence of genotype interaction, which is consistent with the pre-vaccine situation, may impact the dynamics of non-vaccine genotypes, particularly in less active individuals.


Asunto(s)
Coinfección , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Genotipo , Humanos , Papillomaviridae/genética , Infecciones por Papillomavirus/epidemiología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/genética , Prevalencia , Conducta Sexual , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA