Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 5074, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36977691

RESUMO

Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.


Assuntos
COVID-19 , Vacinas contra Influenza , Influenza Humana , Animais , Humanos , Macaca mulatta , Virossomos , SARS-CoV-2 , Receptor 7 Toll-Like , COVID-19/prevenção & controle , Adjuvantes Imunológicos , Anticorpos Amplamente Neutralizantes , Vacinas contra COVID-19 , Anticorpos Antivirais , Anticorpos Neutralizantes
2.
Sci Rep ; 12(1): 11583, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35803968

RESUMO

The COVID-19 pandemic has caused considerable interest worldwide in antiviral surfaces, and there has been a dramatic increase in the research and development of innovative material systems to reduce virus transmission in the past few years. The International Organization for Standardization (ISO) norms 18,184 and 21,702 are two standard methods to characterize the antiviral properties of porous and non-porous surfaces. However, during the last years of the pandemic, a need for faster and inexpensive characterization of antiviral material was identified. Therefore, a complementary method based on an Inactivated Virus System (InViS) was developed to facilitate the early-stage development of antiviral technologies and quality surveillance of the production of antiviral materials safely and efficiently. The InViS is loaded with a self-quenched fluorescent dye that produces a measurable increase in fluorescence when the viral envelope disintegrates. In the present work, the sensitivity of InViS to viral disintegration by known antiviral agents is demonstrated and its potential to characterize novel materials and surfaces is explored. Finally, the InViS is used to determine the fate of viral particles within facemasks layers, rendering it an interesting tool to support the development of antiviral surface systems for technical and medical applications.


Assuntos
COVID-19 , Vírus , Antivirais/farmacologia , Humanos , Pandemias
3.
Front Bioeng Biotechnol ; 10: 879078, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35669054

RESUMO

Plasmodium falciparum cysteine-rich protective antigen (PfCyRPA) has been identified as a promising blood-stage candidate antigen to include in a broadly cross-reactive malaria vaccine. In the last couple of decades, substantial effort has been committed to the development of scalable cost-effective, robust, and high-yield PfCyRPA production processes. Despite insect cells being a suitable expression system due to their track record for protein production (including vaccine antigens), these are yet to be explored to produce this antigen. In this study, different insect cell lines, culture conditions (baculovirus infection strategy, supplementation schemes, culture temperature modulation), and purification strategies (affinity tags) were explored aiming to develop a scalable, high-yield, and high-quality PfCyRPA for inclusion in a virosome-based malaria vaccine candidate. Supplements with antioxidants improved PfCyRPA volumetric titers by 50% when added at the time of infection. In addition, from three different affinity tags (6x-His, 4x-His, and C-tag) evaluated, the 4x-His affinity tag was the one leading to the highest PfCyRPA purification recovery yields (61%) and production yield (26 mg/L vs. 21 mg/L and 13 mg/L for 6x-His and C-tag, respectively). Noteworthy, PfCyRPA expressed using High Five cells did not show differences in protein quality or stability when compared to its human HEK293 cell counterpart. When formulated in a lipid-based virosome nanoparticle, immunized rabbits developed functional anti-PfCyRPA antibodies that impeded the multiplication of P. falciparum in vitro. This work demonstrates the potential of using IC-BEVS as a qualified platform to produce functional recombinant PfCyRPA protein with the added benefit of being a non-human expression system with short bioprocessing times and high expression levels.

4.
Pharmaceutics ; 14(4)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35456687

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) homotrimeric spike (S) protein is responsible for mediating host cell entry by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, thus being a key viral antigen to target in a coronavirus disease 19 (COVID-19) vaccine. Despite the availability of COVID-19 vaccines, low vaccine coverage as well as unvaccinated and immune compromised subjects are contributing to the emergence of SARS-CoV-2 variants of concern. Therefore, continued development of novel and/or updated vaccines is essential for protecting against such new variants. In this study, we developed a scalable bioprocess using the insect cells-baculovirus expression vector system (IC-BEVS) to produce high-quality S protein, stabilized in its pre-fusion conformation, for inclusion in a virosome-based COVID-19 vaccine candidate. By exploring different bioprocess engineering strategies (i.e., signal peptides, baculovirus transfer vectors, cell lines, infection strategies and formulation buffers), we were able to obtain ~4 mg/L of purified S protein, which, to the best of our knowledge, is the highest value achieved to date using insect cells. In addition, the insect cell-derived S protein exhibited glycan processing similar to mammalian cells and mid-term stability upon storage (up to 90 days at -80 and 4 °C or after 5 freeze-thaw cycles). Noteworthy, antigenicity of S protein, either as single antigen or displayed on the surface of virosomes, was confirmed by ELISA, with binding of ACE2 receptor, pan-SARS antibody CR3022 and neutralizing antibodies to the various epitope clusters on the S protein. Binding capacity was also maintained on virosomes-S stored at 4 °C for 1 month. This work demonstrates the potential of using IC-BEVS to produce the highly glycosylated and complex S protein, without compromising its integrity and antigenicity, to be included in a virosome-based COVID-19 vaccine candidate.

5.
Front Immunol ; 13: 788619, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35273592

RESUMO

A virosomal vaccine inducing systemic/mucosal anti-HIV-1 gp41 IgG/IgA had previously protected Chinese-origin rhesus macaques (RMs) against vaginal SHIVSF162P3 challenges. Here, we assessed its efficacy in Indian-origin RMs by intramuscular priming/intranasal boosting (n=12/group). Group K received virosome-P1-peptide alone (harboring the Membrane Proximal External Region), Group L combined virosome-rgp41 plus virosome-P1, and Group M placebo virosomes. Vaccination induced plasma binding but no neutralizing antibodies. Five weeks after boosting, all RMs were challenged intravaginally with low-dose SHIVSF162P3 until persistent systemic infection developed. After SHIV challenge #7, six controls were persistently infected versus only one Group L animal (vaccine efficacy 87%; P=0.0319); Group K was not protected. After a 50% SHIV dose increase starting with challenge #8, protection in Group L was lost. Plasmas/sera were analyzed for IgG phenotypes and effector functions; the former revealed that protection in Group L was significantly associated with increased binding to FcγR2/3(A/B) across several time-points, as were some IgG measurements. Vaginal washes contained low-level anti-gp41 IgGs and IgAs, representing a 1-to-5-fold excess over the SHIV inoculum's gp41 content, possibly explaining loss of protection after the increase in challenge-virus dose. Virosomal gp41-vaccine efficacy was confirmed during the initial seven SHIV challenges in Indian-origin RMs when the SHIV inoculum had at least 100-fold more HIV RNA than acutely infected men's semen. Vaccine protection by virosome-induced IgG and IgA parallels the cooperation between systemically administered IgG1 and mucosally applied dimeric IgA2 monoclonal antibodies that as single-agents provided no/low protection - but when combined, prevented mucosal SHIV transmission in all passively immunized RMs.


Assuntos
Vacinas contra a AIDS , Soropositividade para HIV , HIV-1 , Vírus da Imunodeficiência Símia , Animais , Feminino , Humanos , Imunoglobulina A , Imunoglobulina G , Macaca mulatta , Virossomos
6.
Clin Exp Allergy ; 51(2): 339-349, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33368719

RESUMO

BACKGROUND: Whereas sublingual allergen immunotherapy (AIT) is routinely performed without any adjuvant or delivery system, there is a strong scientific rationale to better target the allergen(s) to oral dendritic cells known to support regulatory immune responses by using appropriate presentation platforms. OBJECTIVE: To identify a safe presentation platform able to enhance allergen-specific tolerance induction. METHODS: Virosomes with membrane-integrated contiguous overlapping peptides (COPs) of Bet v 1 and TLR4 or TLR2/TLR7 agonists were assessed for induction of Bet v 1-specific IgG1, IgG2a and IgE antibodies, hypersensitivity reactions and body temperature drop following subcutaneous injection in naive CD-1 mice. The most promising candidate, Bet v 1 COPs anchored to virosomes with membrane-incorporated TLR4 agonist (Vir.A-Bet v 1 COPs), was further evaluated by the sublingual route in a therapeutic setting in BALB/c mice with birch pollen-induced allergic asthma. Airway hyperresponsiveness, pro-inflammatory cells in bronchoalveolar lavages and polarization of Th cells in the lungs and spleen were then assessed. RESULTS: Both types of adjuvanted virosomes coupled to Bet v 1 COPs triggered a boosted Th1 immunity. Given a more favourable safety profile, Vir.A-Bet v 1 COPs were further evaluated and shown to able to fully reverse asthma symptoms and lung inflammation in a sublingual therapeutic model of birch pollen allergy. CONCLUSIONS AND CLINICAL RELEVANCE: We report herein for the first time on the capacity of a novel and safe presentation platform, that is virosomes with membrane-integrated TLR4 agonist, to improve dramatically sublingual AIT efficacy in a murine model due to its intrinsic dual properties of targeting and stimulating to further promote anti-allergic immune responses. As such, our study paves the ground for further clinical development of this allergen presentation platform for patients suffering from respiratory allergies.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos de Plantas/farmacologia , Asma/imunologia , Imunoglobulina E/efeitos dos fármacos , Imunoglobulina G/efeitos dos fármacos , Rinite Alérgica Sazonal/imunologia , Imunoterapia Sublingual/métodos , Linfócitos T/efeitos dos fármacos , Animais , Antígenos de Plantas/administração & dosagem , Betula/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Camundongos , Peptídeos/administração & dosagem , Peptídeos/farmacologia , Linfócitos T/imunologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Receptor 2 Toll-Like/agonistas , Receptor 4 Toll-Like/agonistas , Receptor 7 Toll-Like/agonistas , Virossomos
7.
NPJ Vaccines ; 5(1): 41, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32435515

RESUMO

The main objective of the MACIVIVA European consortium was to develop new Good Manufacturing Practice pilot lines for manufacturing thermostable vaccines with stabilized antigens on influenza virosomes as enveloped virus-like particles. The HIV-1 gp41-derived antigens anchored in the virosome membrane, along with the adjuvant 3M-052 (TLR7/8 agonist) on the same particle, served as a candidate vaccine for the proof of concept for establishing manufacturing processes, which can be directly applied or adapted to other virosomal vaccines or lipid-based particles. Heat spray-dried powders suitable for nasal or oral delivery, and freeze-dried sublingual tablets were successfully developed as solid dosage forms for mucosal vaccination. The antigenic properties of vaccinal antigens with key gp41 epitopes were maintained, preserving the original immunogenicity of the starting liquid form, and also when solid forms were exposed to high temperature (40 °C) for up to 3 months, with minimal antigen and adjuvant content variation. Virosomes reconstituted from the powder forms remained as free particles with similar size, virosome uptake by antigen-presenting cells in vitro was comparable to virosomes from the liquid form, and the presence of excipients specific to each solid form did not prevent virosome transport to the draining lymph nodes of immunized mice. Virosome integrity was also preserved during exposure to <-15 °C, mimicking accidental freezing conditions. These "ready to use and all-in-one" thermostable needle-free virosomal HIV-1 mucosal vaccines offer the advantage of simplified logistics with a lower dependence on the cold chain during shipments and distribution.

8.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L813-L830, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32073879

RESUMO

Our understanding of mesenchymal cell subsets and their function in human lung affected by aging and in certain disease settings remains poorly described. We use a combination of flow cytometry, prospective cell-sorting strategies, confocal imaging, and modeling of microvessel formation using advanced microfluidic chip technology to characterize mesenchymal cell subtypes in human postnatal and adult lung. Tissue was obtained from patients undergoing elective surgery for congenital pulmonary airway malformations (CPAM) and other airway abnormalities including chronic obstructive pulmonary disease (COPD). In microscopically normal postnatal human lung, there was a fivefold higher mesenchymal compared with epithelial (EpCAM+) fraction, which diminished with age. The mesenchymal fraction composed of CD90+ and CD90+CD73+ cells was enriched in CXCL12 and platelet-derived growth factor receptor-α (PDGFRα) and located in close proximity to EpCAM+ cells in the alveolar region. Surprisingly, alveolar organoids generated from EpCAM+ cells supported by CD90+ subset were immature and displayed dysplastic features. In congenital lung lesions, cystic air spaces and dysplastic alveolar regions were marked with an underlying thick interstitium composed of CD90+ and CD90+PDGFRα+ cells. In postnatal lung, a subset of CD90+ cells coexpresses the pericyte marker CD146 and supports self-assembly of perfusable microvessels. CD90+CD146+ cells from COPD patients fail to support microvessel formation due to fibrinolysis. Targeting the plasmin-plasminogen system during microvessel self-assembly prevented fibrin gel degradation, but microvessels were narrower and excessive contraction blocked perfusion. These data provide important new information regarding the immunophenotypic identity of key mesenchymal lineages and their change in a diverse setting of congenital lung lesions and COPD.


Assuntos
Imunomodulação/imunologia , Células-Tronco Mesenquimais/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo , Adolescente , Biomarcadores/metabolismo , Antígeno CD146/imunologia , Antígeno CD146/metabolismo , Separação Celular/métodos , Criança , Pré-Escolar , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Feminino , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Lactente , Recém-Nascido , Masculino , Células-Tronco Mesenquimais/imunologia , Microvasos/imunologia , Microvasos/metabolismo , Pericitos/imunologia , Pericitos/metabolismo , Estudos Prospectivos
9.
NPJ Vaccines ; 5: 9, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32025340

RESUMO

The Plasmodium falciparum (Pf) cysteine-rich protective antigen (PfCyRPA) has emerged as a promising blood-stage candidate antigen for inclusion into a broadly cross-reactive malaria vaccine. This highly conserved protein among various geographical strains plays a key role in the red blood cell invasion process by P. falciparum merozoites, and antibodies against PfCyRPA can efficiently prevent the entry of the malaria parasites into red blood cells. The aim of the present study was to develop a human-compatible formulation of the PfCyRPA vaccine candidate and confirming its activity in preclinical studies. Recombinant PfCyRPA expressed in HEK 293 cells was chemically coupled to phosphoethanolamine and then incorporated into the membrane of unadjuvanted influenza virosomes approved as antigen delivery system for humans. Laboratory animals were immunised with the virosome-based PfCyRPA vaccine to determine its immunogenic properties and in particular, its capacity to elicit parasite binding and growth-inhibitory antibodies. The vaccine elicited in mice and rabbits high titers of PfCyRPA-specific antibodies that bound to the blood-stage parasites. At a concentration of 10 mg/mL, purified total serum IgG from immunised rabbits inhibited parasite growth in vitro by about 80%. Furthermore, in a P. falciparum infection mouse model, passive transfer of 10 mg of purified total IgG from PfCyRPA vaccinated rabbits reduced the in vivo parasite load by 77%. Influenza virosomes thus represent a suitable antigen delivery system for the induction of protective antibodies against the recombinant PfCyRPA, designating it as a highly suitable component for inclusion into a multivalent and multi-stage virosomal malaria vaccine.

10.
Front Immunol ; 8: 359, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28439267

RESUMO

Pulmonary administration of biomimetic nanoparticles loaded with antigen may represent an effective strategy to directly modulate adaptive immune responses in the respiratory tract. Depending on the design, virosomes may not only serve as biomimetic antigen carriers but are also endowed with intrinsic immune-stimulatory properties. We designed fluorescently labeled influenza-derived virosomes and liposome controls coupled to the model antigen ovalbumin to investigate uptake, phenotype changes, and antigen processing by antigen-presenting cells exposed to such particles in different respiratory tract compartments. Both virosomes and liposomes were captured by pulmonary macrophages and dendritic cells alike and induced activation in particle-bearing cells by upregulation of costimulatory markers such as CD40, CD80, CD86, PD-L1, PD-L2, and ICOS-L. Though antigen processing and accumulation of both coupled and soluble antigen was similar between virosomes and liposomes, only ovalbumin-coupled virosomes generated a strong antigen-specific CD4+ T cell proliferation. Pulmonary administrated antigen-coupled virosomes therefore effectively induced adaptive immune responses and may be utilized in novel preventive or therapeutic approaches in the respiratory tract.

11.
Nanomedicine ; 13(5): 1725-1737, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28214610

RESUMO

There is considerable interest to develop antigen-carriers for immune-modulatory clinical applications, but insufficient information is available on their effects on antigen-presenting cells. We employed virosomes coupled to ovalbumin (OVA) to study their interaction with murine bone marrow-derived dendritic cells (BMDCs) and modulation of downstream T cell responses. BMDCs were treated in vitro with virosomes or liposomes prior to determining BMDC phenotype, viability, and intracellular trafficking. Antigen-specific CD4+ T cell activation was measured by co-culture of BMDCs with DO11.10 CD4+ T cells. Compared to liposomes, virosomes were rapidly taken up. Neither nanocarrier type affected BMDC viability, nor did a moderate degree of activation differ for markers such as CD40, CD80, CD86. Virosome uptake occurred via clathrin-mediated endocytosis and phagocytosis, with co-localization in late endosomes. Only BMDCs treated with OVA-coupled virosomes induced enhanced OVA-specific CD4+ T cell proliferation. Antigen-coupled virosomes are endowed with an intrinsic ability to modulate DC-dependent adaptive immune responses.


Assuntos
Linfócitos T CD4-Positivos , Células Dendríticas , Virossomos , Imunidade Adaptativa , Animais , Antígenos , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina , Linfócitos T Reguladores , Células Th1
12.
PLoS One ; 11(9): e0163539, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27685460

RESUMO

The respiratory tract with its ease of access, vast surface area and dense network of antigen-presenting cells (APCs) represents an ideal target for immune-modulation. Bio-mimetic nanocarriers such as virosomes may provide immunomodulatory properties to treat diseases such as allergic asthma. In our study we employed a triple co-culture model of epithelial cells, macrophages and dendritic cells to simulate the human airway barrier. The epithelial cell line 16HBE was grown on inserts and supplemented with human blood monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) for exposure to influenza virosomes and liposomes. Additionally, primary human nasal epithelial cells (PHNEC) and EpCAM+ epithelial progenitor cell mono-cultures were utilized to simulate epithelium from large and smaller airways, respectively. To assess particle uptake and phenotype change, cell cultures were analyzed by flow cytometry and pro-inflammatory cytokine concentrations were measured by ELISA. All cell types internalized virosomes more efficiently than liposomes in both mono- and co-cultures. APCs like MDMs and MDDCs showed the highest uptake capacity. Virosome and liposome treatment caused a moderate degree of activation in MDDCs from mono-cultures and induced an increased cytokine production in co-cultures. In epithelial cells, virosome uptake was increased compared to liposomes in both mono- and co-cultures with EpCAM+ epithelial progenitor cells showing highest uptake capacity. In conclusion, all cell types successfully internalized both nanocarriers with virosomes being taken up by a higher proportion of cells and at a higher rate inducing limited activation of MDDCs. Thus virosomes may represent ideal carrier antigen systems to modulate mucosal immune responses in the respiratory tract without causing excessive inflammatory changes.

13.
mBio ; 6(3): e00724, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26037125

RESUMO

UNLABELLED: Vaginal inflammation (vaginitis) is the most common disease caused by the human-pathogenic fungus Candida albicans. Secretory aspartyl proteinases (Sap) are major virulence traits of C. albicans that have been suggested to play a role in vaginitis. To dissect the mechanisms by which Sap play this role, Sap2, a dominantly expressed member of the Sap family and a putative constituent of an anti-Candida vaccine, was used. Injection of full-length Sap2 into the mouse vagina caused local neutrophil influx and accumulation of the inflammasome-dependent interleukin-1ß (IL-1ß) but not of inflammasome-independent tumor necrosis factor alpha. Sap2 could be replaced by other Sap, while no inflammation was induced by the vaccine antigen, the N-terminal-truncated, enzymatically inactive tSap2. Anti-Sap2 antibodies, in particular Fab from a human combinatorial antibody library, inhibited or abolished the inflammatory response, provided the antibodies were able, like the Sap inhibitor Pepstatin A, to inhibit Sap enzyme activity. The same antibodies and Pepstatin A also inhibited neutrophil influx and cytokine production stimulated by C. albicans intravaginal injection, and a mutant strain lacking SAP1, SAP2, and SAP3 was unable to cause vaginal inflammation. Sap2 induced expression of activated caspase-1 in murine and human vaginal epithelial cells. Caspase-1 inhibition downregulated IL-1ß and IL-18 production by vaginal epithelial cells, and blockade of the IL-1ß receptor strongly reduced neutrophil influx. Overall, the data suggest that some Sap, particularly Sap2, are proinflammatory proteins in vivo and can mediate the inflammasome-dependent, acute inflammatory response of vaginal epithelial cells to C. albicans. These findings support the notion that vaccine-induced or passively administered anti-Sap antibodies could contribute to control vaginitis. IMPORTANCE: Candidal vaginitis is an acute inflammatory disease that affects many women of fertile age, with no definitive cure and, in its recurrent forms, causing true devastation of quality of life. Unraveling the fungal factors causing inflammation is important to be able to devise novel tools to fight the disease. In an experimental murine model, we have discovered that aspartyl proteinases, particularly Sap2, may cause the same inflammatory signs of vaginitis caused by the fungus and that anti-Sap antibodies and the protease inhibitor Pepstatin A almost equally inhibit Sap- and C. albicans-induced inflammation. Sap-induced vaginitis is an early event during vaginal infection, is uncoupled from fungal growth, and requires Sap and caspase-1 enzymatic activities to occur, suggesting that Sap or products of Sap activity activate an inflammasome sensor of epithelial cells. Our data support the notion that anti-Sap antibodies could help control the essence of candidal vaginitis, i.e., the inflammatory response.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Candida albicans/enzimologia , Candidíase Vulvovaginal/patologia , Proteínas Fúngicas/metabolismo , Fatores de Virulência/metabolismo , Animais , Candida albicans/crescimento & desenvolvimento , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Inflamassomos/metabolismo , Camundongos , Neutrófilos/imunologia , Vagina/patologia
14.
Immunology ; 145(1): 71-81, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25471322

RESUMO

This study examined specific antibody and T-cell responses associated with experimental malaria infection or malaria vaccination, in malaria-naive human volunteers within phase I/IIa vaccine trials, with a view to investigating inter-relationships between these types of response. Malaria infection was via five bites of Plasmodium falciparum-infected mosquitoes, with individuals reaching patent infection by 11-12 days, having harboured four or five blood-stage cycles before drug clearance. Infection elicited a robust antibody response against merozoite surface protein-119 , correlating with parasite load. Classical class switching was seen from an early IgM to an IgG1-dominant response of increasing affinity. Malaria-specific T-cell responses were detected in the form of interferon-γ and interleukin-4 (IL-4) ELIspot, but their magnitude did not correlate with the magnitude of antibody or its avidity, or with parasite load. Different individuals who were immunized with a virosome vaccine comprising influenza antigens combined with P. falciparum antigens, demonstrated pre-existing interferon-γ, IL-2 and IL-5 ELIspot responses against the influenza antigens, and showed boosting of anti-influenza T-cell responses only for IL-5. The large IgG1-dominated anti-parasite responses showed limited correlation with T-cell responses for magnitude or avidity, both parameters being only negatively correlated for IL-5 secretion versus anti-apical membrane antigen-1 antibody titres. Overall, these findings suggest that cognate T-cell responses across a range of magnitudes contribute towards driving potentially effective antibody responses in infection-induced and vaccine-induced immunity against malaria, and their existence during immunization is beneficial, but magnitudes are mostly not inter-related.


Assuntos
Anticorpos Antiprotozoários/imunologia , Formação de Anticorpos/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Citocinas/imunologia , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/patologia , Malária Falciparum/prevenção & controle , Masculino , Proteína 1 de Superfície de Merozoito/administração & dosagem , Proteína 1 de Superfície de Merozoito/imunologia , Pessoa de Meia-Idade
15.
Expert Rev Vaccines ; 12(7): 779-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23885823

RESUMO

The basic concept of virosomes is the controlled in vitro assembly of virus-like particles from purified components. The first generation of influenza virosomes developed two decades ago is successfully applied in licensed vaccines, providing a solid clinical safety and efficacy track record for the technology. In the meantime, a second generation of influenza virosomes has evolved as a carrier and adjuvant system, which is currently applied in preclinical and clinical stage vaccine candidates targeting various prophylactic and therapeutic indications. The inclusion of additional components to optimize particle assembly, to stabilize the formulations, or to enhance the immunostimulatory properties have further improved and broadened the applicability of the platform.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Orthomyxoviridae/genética , Vacinas/administração & dosagem , Vacinas/imunologia , Virossomos/administração & dosagem , Humanos , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/imunologia , Virossomos/genética
16.
PLoS One ; 8(2): e55438, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437055

RESUMO

UNLABELLED: Mucosal antibodies harboring various antiviral activities may best protect mucosal surfaces against early HIV-1 entry at mucosal sites and they should be ideally induced by prophylactic HIV-1 vaccines for optimal prevention of sexually transmitted HIV-1. A phase I, double-blind, randomized, placebo-controlled trial was conducted in twenty-four healthy HIV-uninfected young women. The study objectives were to assess the safety, tolerability and immunogenicity of virosomes harboring surface HIV-1 gp41-derived P1 lipidated peptides (MYM-V101). Participants received placebo or MYM-V101 vaccine at 10 µg/dose or 50 µg/dose intramuscularly at week 0 and 8, and intranasally at week 16 and 24. MYM-V101 was safe and well-tolerated at both doses administered by the intramuscular and intranasal routes, with the majority of subjects remaining free of local and general symptoms. P1-specific serum IgGs and IgAs were induced in all high dose recipients after the first injection. After the last vaccination, vaginal and rectal P1-specific IgGs could be detected in all high dose recipients. Approximately 63% and 43% of the low and high dose recipients were respectively tested positive for vaginal P1-IgAs, while 29% of the subjects from the high dose group tested positive for rectal IgAs. Serum samples had total specific IgG and IgA antibody concentrations ≥ 0.4 µg/mL, while mucosal samples were usually below 0.01 µg/mL. Vaginal secretions from MYM-V101 vaccinated subjects were inhibiting HIV-1 transcytosis but had no detectable neutralizing activity. P1-specific Th1 responses could not be detected on PBMC. This study demonstrates the excellent safety and tolerability of MYM-V101, eliciting systemic and mucosal antibodies in the majority of subjects. Vaccine-induced mucosal anti-gp41 antibodies toward conserved gp41 motifs were harboring HIV-1 transcytosis inhibition activity and may contribute to reduce sexually-transmitted HIV-1. TRIAL REGISTRATION: ClinicalTrials.gov NCT01084343.


Assuntos
Vacinas contra a AIDS/efeitos adversos , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/imunologia , Mucosa/imunologia , Fragmentos de Peptídeos/imunologia , Vacinação , Vacinas de Subunidades Antigênicas/efeitos adversos , Virossomos/imunologia , Vacinas contra a AIDS/imunologia , Adulto , Sequência de Aminoácidos , Fármacos Anti-HIV/imunologia , Especificidade de Anticorpos/imunologia , Feminino , Anticorpos Anti-HIV/sangue , Proteína gp41 do Envelope de HIV/química , Infecções por HIV/sangue , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , Saúde , Humanos , Imunidade Celular/imunologia , Injeções Intramusculares , Dados de Sequência Molecular , Mucosa/virologia , Fragmentos de Peptídeos/química , Transcitose/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Adulto Jovem
17.
Vaccine ; 30(30): 4490-8, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22561143

RESUMO

A novel vaccine (PEV7) consisting of a truncated, recombinant aspartyl proteinase-2 of Candida albicans incorporated into influenza virosomes was studied. This vaccine candidate generated a potent serum antibody response in mouse and rat following intramuscular immunization. Anti-Sap2 IgG and IgA were also detected in the vaginal fluid of rats following intravaginal or intramuscular plus intravaginal administration. In a rat model of candidal vaginitis, PEV7 induced significant, long-lasting, likely antibody-mediated, protection following intravaginal route of immunization. PEV7 was also found to be safe in a repeated-dose toxicological study in rats. Overall, these data provide a sound basis to envisage the clinical development of this new candidate vaccine against candidal vaginitis.


Assuntos
Ácido Aspártico Endopeptidases/imunologia , Candidíase Vulvovaginal/prevenção & controle , Proteínas Fúngicas/imunologia , Vacinas Fúngicas/administração & dosagem , Virossomos/administração & dosagem , Administração Intravaginal , Animais , Anticorpos Antifúngicos/sangue , Candida albicans/imunologia , Candidíase Vulvovaginal/imunologia , Reações Cruzadas , Feminino , Vacinas Fúngicas/imunologia , Masculino , Camundongos , Orthomyxoviridae/imunologia , Ratos , Ratos Wistar , Testes de Toxicidade , Virossomos/imunologia
18.
Malar J ; 10: 359, 2011 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-22166048

RESUMO

BACKGROUND: In clinical trials, immunopotentiating reconstituted influenza virosomes (IRIVs) have shown great potential as a versatile antigen delivery platform for synthetic peptides derived from Plasmodium falciparum antigens. This study describes the immunogenicity of a virosomally-formulated recombinant fusion protein comprising domains of the two malaria vaccine candidate antigens MSP3 and GLURP. METHODS: The highly purified recombinant protein GMZ2 was coupled to phosphatidylethanolamine and the conjugates incorporated into the membrane of IRIVs. The immunogenicity of this adjuvant-free virosomal formulation was compared to GMZ2 formulated with the adjuvants Montanide ISA 720 and Alum in three mouse strains with different genetic backgrounds. RESULTS: Intramuscular injections of all three candidate vaccine formulations induced GMZ2-specific antibody responses in all mice tested. In general, the humoral immune response in outbred NMRI mice was stronger than that in inbred BALB/c and C57BL/6 mice. ELISA with the recombinant antigens demonstrated immunodominance of the GLURP component over the MSP3 component. However, compared to the Al(OH)(3)-adjuvanted formulation the two other formulations elicited in NMRI mice a larger proportion of anti-MSP3 antibodies. Analyses of the induced GMZ2-specific IgG subclass profiles showed for all three formulations a predominance of the IgG1 isotype. Immune sera against all three formulations exhibited cross-reactivity with in vitro cultivated blood-stage parasites. Immunofluorescence and immunoblot competition experiments showed that both components of the hybrid protein induced IgG cross-reactive with the corresponding native proteins. CONCLUSION: A virosomal formulation of the chimeric protein GMZ2 induced P. falciparum blood stage parasite cross-reactive IgG responses specific for both MSP3 and GLURP. GMZ2 thus represents a candidate component suitable for inclusion into a multi-valent virosomal malaria vaccine and influenza virosomes represent a versatile antigen delivery system suitable for adjuvant-free immunization with recombinant proteins.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/genética , Reações Cruzadas , Imunoglobulina G/sangue , Injeções Intramusculares , Vacinas Antimaláricas/administração & dosagem , Manitol/administração & dosagem , Manitol/análogos & derivados , Camundongos , Ácidos Oleicos/administração & dosagem , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Virossomais/administração & dosagem , Vacinas Virossomais/imunologia
19.
Expert Rev Vaccines ; 10(4): 437-46, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21506642

RESUMO

Influenza virosomes have been used for more than 10 years in commercial vaccines. The technology has been further developed as a carrier and adjuvant system for subunit vaccines, in particular for synthetic peptides. The extensive amount of preclinical and clinical data supports the notion that influenza virosomes represent a platform technology that ensures robust and long-lasting immune responses against subunit antigens with an excellent safety profile. Structurally and functionally, virosomes are enveloped virus-like particles, although they are assembled in vitro. This unique feature ensures a tight control of their composition and at the same time provides the flexibility to adapt the particle to various types of antigens. The mode of action of virosomes is complex and includes carrier as well as immune-stimulatory functions.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Vacinas contra Influenza/imunologia , Virossomos/administração & dosagem , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Virossomais/administração & dosagem , Vacinas Virossomais/imunologia
20.
Immunity ; 34(2): 269-80, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21315623

RESUMO

Human immunodeficiency virus (HIV)-1 is mainly transmitted mucosally during sexual intercourse. We therefore evaluated the protective efficacy of a vaccine active at mucosal sites. Macaca mulatta monkeys were immunized via both the intramuscular and intranasal routes with an HIV-1 vaccine made of gp41-subunit antigens grafted on virosomes, a safe delivery carrier approved in humans with self-adjuvant properties. Six months after 13 vaginal challenges with simian-HIV (SHIV)-SF162P3, four out of five vaccinated animals remained virus-negative, and the fifth was only transiently infected. None of the five animals seroconverted to p27gag-SIV. In contrast, all 6 placebo-vaccinated animals became infected and seroconverted. All protected animals showed gp41-specific vaginal IgAs with HIV-1 transcytosis-blocking properties and vaginal IgGs with neutralizing and/or antibody-dependent cellular-cytotoxicity activities. In contrast, plasma IgGs totally lacked virus-neutralizing activity. The protection observed challenges the paradigm whereby circulating antiviral antibodies are required for protection against HIV-1 infection and may serve in designing a human vaccine against HIV-1-AIDS.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Anticorpos Anti-HIV/biossíntese , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/imunologia , Imunização , Macaca mulatta/imunologia , Fragmentos de Peptídeos/imunologia , Vagina/imunologia , Virossomos/imunologia , Vacinas contra a AIDS/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos , Sítios de Ligação , Feminino , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/administração & dosagem , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , Soropositividade para HIV , Dados de Sequência Molecular , Fragmentos de Peptídeos/administração & dosagem , Transcitose , Viremia/imunologia , Viremia/prevenção & controle , Viremia/transmissão , Produtos do Gene gag do Vírus da Imunodeficiência Humana/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...