Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Med Microbiol Immunol ; 211(2-3): 153-160, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35552511

RESUMEN

Human papillomavirus type 16 (HPV-16) is the predominant genotype worldwide associated with invasive cervical cancer and hence remains as the focus for diagnostic development and vaccine research. L2, the minor capsid protein forms the packaging unit for the HPV genome along with the L1 protein and is primarily associated with transport of genomic DNA to the nucleus. Unlike L1, L2 is known to elicit cross-neutralizing antibodies and thus becomes a suitable candidate for pan-HPV prophylactic vaccine development. In the present study, a total of 148 cervical HPV-16 isolates from Indian women were analyzed by PCR-directed sequencing, phylogenetic analysis and in silico immunoinformatics tools to determine the L2 variations that may impact the immune response and oncogenesis. Ninety-one SNPs translating to 35 non-synonymous amino acid substitutions were observed, of these 16 substitutions are reported in the Indian isolates for the first time. T245A, L266F, S378V and S384A substitutions were significantly associated with high-grade cervical neoplastic status. Multiple substitutions were observed in samples from high-grade cervical neoplastic status as compared to those from normal cervical status (p = 0.027), specifically from the D3 sub-lineage. It was observed that substitution T85A was part of both, B and T cell epitopes recognized by MHC-I molecules; T245A was common to B and T cell epitopes recognized by MHC-II molecules and S122P/A was common to the region recognized by both MHC-I and MHC-II molecules. These findings reporting L2 protein substitutions have implications on cervical oncogenesis and design of next-generation L2-based HPV vaccines.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Anticuerpos Antivirales , Proteínas de la Cápside/genética , Carcinogénesis , Epítopos de Linfocito T , Femenino , Antígenos de Histocompatibilidad Clase II/genética , Papillomavirus Humano 16/genética , Humanos , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Vacunas contra Papillomavirus/genética , Filogenia
16.
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
17.
Virus Res ; 314: 198756, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35364119

RESUMEN

Papillomaviruses (PV) have a wide distribution of hosts, among which human papillomavirus (HPV) has been recognized as the major cause of cervical cancer. HPV is characterized by its high genetic variability with more than 200 genotypes identified, and numerous variants exist within the same genotype. Though phylogenetic incongruence between early gene and late gene of PVs was observed, the recombination in HPV was not taken seriously until the last two decades. The first report of evidence on HPV recombination was published in 2006, in which only intertypic ancient recombination events were identified. Since then, several publications on recombination in HPV provided evidence for intertypic as well as intratypic recombination. Recombination may create challenges on HPV genotyping and vaccination that could cause a great impact in screening and prevention of cervical cancer. Here, we review the literature on recombination and summarize the reasons underlying the difficulties for detecting recombination in HPV. In addition, we analyze the potential consequences of HPV recombination and make further prospects for clinical practice in the future.


Asunto(s)
Alphapapillomavirus , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Neoplasias del Cuello Uterino , Virus no Clasificados , Femenino , Genotipo , Humanos , Papillomaviridae/genética , Vacunas contra Papillomavirus/genética , Filogenia , Recombinación Genética
18.
Microbiol Spectr ; 10(3): e0077922, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35475682

RESUMEN

Human papillomavirus (HPV) is the causative agent of cervical and other cancers and represents a significant global health burden. HPV vaccines demonstrate excellent efficacy in clinical trials and effectiveness in national immunization programmes against the most prevalent genotype, HPV16. It is unclear whether the greater protection conferred by vaccine-induced antibodies, compared to natural infection antibodies, is due to differences in antibody magnitude and/or specificity. We explore the contribution of the surface-exposed loops of the major capsid protein to antigenic domains recognized by vaccine and natural infection neutralizing antibodies. Chimeric pseudoviruses incorporating individual (BC, DE, EF, FG, HI) or combined (All: BC/DE/EF/FG/HI) loop swaps between the target (HPV16) and control (HPV35) genotypes were generated, purified by ultracentrifugation and characterized by SDS-PAGE and electron microscopy. Neutralizing antibody data were subjected to hierarchical clustering and outcomes modeled on the HPV16 capsomer crystal model. Vaccine antibodies exhibited an FG loop preference followed by the EF and HI loops while natural infection antibodies displayed a more diverse pattern, most frequently against the EF loop followed by BC and FG. Both vaccine and natural infection antibodies demonstrated a clear requirement for multiple loops. Crystal modeling of these neutralizing antibody patterns suggested natural infection antibodies typically target the outer rim of the capsomer while vaccine antibodies target the central ring around the capsomer lumen. Chimeric pseudoviruses are useful tools for probing vaccine and natural infection antibody specificity. These data add to the evidence base for the effectiveness of an important public health intervention. IMPORTANCE The human papillomavirus type 16 (HPV16) major virus coat (capsid) protein is a target for antibodies induced by both natural infection and vaccination. Vaccine-induced immunity is highly protective against HPV16-related infection and disease while natural infection associated immunity significantly less so. For this study, we created chimeric functional pseudoviruses based upon an antigenically distant HPV genotype (HPV35) resistant to HPV16-specific antibodies with inserted capsid surface fragments (external loops) from HPV16. By using these chimeric pseudoviruses in functional neutralization assays we were able to highlight specific and distinct areas on the capsid surface recognized by both natural infection and vaccine induced antibodies. These data improve our understanding of the difference between natural infection and vaccine induced HPV16-specific immunity.


Asunto(s)
Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Cápside , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Papillomavirus Humano 16/química , Papillomavirus Humano 16/genética , Humanos , Papillomaviridae/genética , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/genética
19.
BMC Cancer ; 22(1): 42, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991494

RESUMEN

BACKGROUND: The aim of the study was to investigate the risk of human papillomavirus (HPV) genotyping particularly vaccine genotypes and multiple infections for cervical precancer and cancer, which might contribute to developing genotype-specific screening strategy and assessing potential effects of HPV vaccine. METHODS: The HPV genotypes were identified using the Seq HPV assay on self-collected samples. Hierarchical ranking of each genotype was performed according to positive predictive value (PPV) for cervical intraepithelial neoplasia 2/3 or worse (CIN2+/CIN3+). Multivariate logistic regression model was used to estimate the odds ratios (ORs) with 95% confidence interval (CI) of CIN2+ according to multiplicity of types and vaccine types. RESULTS: A total of 2811 HPV-positive women were analyzed. The five dominant HPV genotypes in high-grade lesions were 16/58/52/33/18. The overall ranking orders were HPV16/33/35/58/31/68/18/ 56/52/66/51/59/45/39 for CIN2+ and HPV16/33/31/58/45/66/52/18/35/56/51/68/59/39 for CIN3+. The risks of single infection versus co-infections with other types lower in the hierarchy having CIN2+ were not statistically significant for HPV16 (multiple infection vs. single infection: OR = 0.8, 95%CI = 0.6-1.1, P = 0.144) or other genotypes (P > 0.0036) after conservative Bonferroni correction. Whether HPV16 was present or not, the risks of single infection versus multiple infection with any number (2, ≥2, or ≥ 3) of types for CIN2+ were not significantly different. In addition, HPV31/33/45/52/58 covered by nonavalent vaccine added 27.5% of CIN2, 23.0% of CIN3, and 12.5% of cancer to the HPV16/18 genotyping. These genotype-groups were at significantly higher risks than genotypes not covered by nonavalent vaccine. Moreover, genotypes covered by nonavalent vaccine contributed to 85.2% of CIN2 lesions, 97.9% of CIN3 and 93.8% of cancers. CONCLUSIONS: Partial extended genotyping such as HPV33/31/58 but not multiplicity of HPV infections could serve as a promising triage for HPV-positive self-samples. Moreover, incidence rates of cervical cancer and precancer were substantial attributable to HPV genotypes covered by current nonavalent vaccination.


Asunto(s)
Técnicas de Genotipaje , Papillomaviridae/genética , Vacunas contra Papillomavirus/genética , Lesiones Precancerosas/virología , Neoplasias del Cuello Uterino/virología , Adulto , Detección Precoz del Cáncer/métodos , Femenino , Genotipo , Humanos , Incidencia , Modelos Logísticos , Persona de Mediana Edad , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Lesiones Precancerosas/epidemiología , Lesiones Precancerosas/prevención & control , Valor Predictivo de las Pruebas , Neoplasias del Cuello Uterino/epidemiología , Neoplasias del Cuello Uterino/prevención & control , Displasia del Cuello del Útero/epidemiología , Displasia del Cuello del Útero/prevención & control , Displasia del Cuello del Útero/virología
20.
J Virol Methods ; 301: 114403, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34890711

RESUMEN

Human papillomavirus (HPV) major capsid protein L1 virus-like particles (VLPs) produced in baculovirus system are highly immunogenic, but the relatively high production cost limits its application in the development of broad-spectrum vaccines. Here we report a novel method for enhancing VLP production in this system. We incorporated respectively 4, 8 or 13 residues truncation mutations in the N-terminus of L1ΔC, a C-terminal 25-residue-deleted L1 of HPV58, to construct three mutants. After expression in Sf9 cells, L1ΔN4C exhibited 2.3-fold higher protein production, 2.0-fold mRNA expression and lower rate of mRNA decay, compared to L1ΔC. More importantly, L1ΔN4C protein was easily purified by two-step chromatography with a VLP yield of up to 60 mg/L (purity > 99 %), 5-fold that of L1ΔC, whereas L1ΔN8C and L1ΔN13C behaved similarly to L1ΔC either in protein or mRNA expression. Moreover, L1ΔN4C VLPs showed similar binding activities with six HPV58 neutralizing monoclonal antibodies and induced comparable level of neutralizing antibody in mice to that of L1ΔC VLPs. Our results demonstrate that certain N- and C-terminal truncations of HPV58 L1 can enhance VLP yield. This method may be used to reduce production costs of other L1VLPs or chimeric VLPs to developing pan-HPV vaccines using baculovirus system.


Asunto(s)
Alphapapillomavirus , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Alphapapillomavirus/metabolismo , Animales , Anticuerpos Antivirales , Baculoviridae/genética , Proteínas de la Cápside/metabolismo , Humanos , Ratones , Proteínas Oncogénicas Virales/genética , Papillomaviridae , Vacunas contra Papillomavirus/química , Vacunas contra Papillomavirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...