Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vaccines (Basel) ; 12(4)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38675801

RESUMEN

To protect older adults against influenza A virus (IAV) infection, innovative strategies are imperative to overcome the decrease in protective immune response with age. One approach involves the boosting of CD8+ T cells at middle age that were previously induced by natural infection. At this stage, the immune system is still fit. Given the high conservation of T-cell epitopes within internal viral proteins, such a response may confer lasting protection against evolving influenza strains at older age, also reducing the high number of influenza immunizations currently required. However, at the time of vaccination, some individuals may have been more recently exposed to IAV than others, which could affect the T-cell response. We therefore investigated the fundamental principle of how the interval between the last infection and booster immunization during middle age influences the CD8+ T-cell response. To model this, female mice were infected at either 6 or 9 months of age and subsequently received a heterosubtypic infection booster at middle age (12 months). Before the booster infection, 6-month-primed mice displayed lower IAV-specific CD8+ T-cell responses in the spleen and lung than 9-month-primed mice. Both groups were better protected against the subsequent heterosubtypic booster infection compared to naïve mice. Notably, despite the different CD8+ T-cell levels between the 6-month- and 9-month-primed mice, we observed comparable responses after booster infection, based on IFNγ responses, and IAV-specific T-cell frequencies and repertoire diversity. Lung-derived CD8+ T cells of 6- and 9-month-primed mice expressed similar levels of tissue-resident memory-T-cell markers 30 days post booster infection. These data suggest that the IAV-specific CD8+ T-cell response after boosting is not influenced by the time post priming.

2.
NPJ Vaccines ; 8(1): 116, 2023 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-37573454

RESUMEN

CD8 + T cells are promising targets for vaccination against influenza A virus (IAV) infection. Their induction via peptide vaccination is not trivial, because peptides are weakly immunogenic. One strategy to overcome this is by vaccination with chemically enhanced altered peptide ligands (CPLs), which have improved MHC-binding and immunogenicity. It remains unknown how peptide-modification affects the resulting immune response. We studied the effect of CPLs derived from the influenza M158-66 epitope (GILGFVFTL) on the T-cell response. In HLA-A2*0201 transgenic mice, CPL-vaccination led to higher T-cell frequencies, but only a small percentage of the induced T cells recognized the GILG-wildtype (WT) peptide. CPL-vaccination resulted in a lower richness of the GILG-WT-specific T-cell repertoire and no improved protection against IAV-infection compared to GILG-WT peptide-vaccination. One CPL even appeared to enhance pathology after IAV-challenge. CPL-vaccination thus induces T cells not targeting the original peptide, which may lead to potential unwanted side effects.

3.
Rev Med Virol ; 33(3): e2330, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35119149

RESUMEN

This review sought to assess the efficacy, effectiveness and safety of high-dose inactivated influenza vaccines (HD-IIV) for the prevention of laboratory-confirmed influenza in individuals aged 18 years or older. A systematic literature search was conducted in electronic databases and grey literature sources up to 7 February 2020. Randomised controlled trials (RCTs) and non-randomised studies of interventions (NRSIs) were included. The search returned 28,846 records, of which 36 studies were included. HD-IIV was shown to have higher relative vaccine efficacy in preventing influenza compared with standard-dose influenza vaccines (SD-IIV3) in older adults (Vaccine effectiveness (VE) = 24%, 95% CI 10-37, one RCT). One NRSI demonstrated significant effect for HD-IIV3 against influenza B (VE = 89%, 95% CI 47-100), but not for influenza A(H3N2) (VE = 22%, 95% CI -82 to 66) when compared with no vaccination in older adults. HD-IIV3 showed significant relative effect compared with SD-IIV3 for influenza-related hospitalisation (VE = 11.8%, 95% CI 6.4-17.0, two NRSIs), influenza- or pneumonia-related hospitalisation (VE = 13.7%, 95% CI 9.5-17.7, three NRSIs), influenza-related hospital encounters (VE = 13.1%, 95% CI 8.4-17.7, five NRSIs), and influenza-related office visits (VE = 3.5%, 95% CI 1.5-5.5, two NRSIs). For safety, HD-IIV were associated with significantly higher rates of local and systemic adverse events compared with SD-IIV (combined local reactions, pain at injection site, swelling, induration, headache, chills and malaise). From limited data, compared with SD-IIV, HD-IIV were found to be more effective in the prevention of laboratory-confirmed influenza, for a range of proxy outcome measures, and associated with more adverse events.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Adolescente , Anciano , Humanos , Gripe Humana/prevención & control , Estaciones del Año , Vacunación/efectos adversos , Vacunas de Productos Inactivados/efectos adversos
4.
Sci Adv ; 8(50): eadc9937, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36516261

RESUMEN

Universal influenza vaccines should protect against continuously evolving and newly emerging influenza viruses. T cells may be an essential target of such vaccines, as they can clear infected cells through recognition of conserved influenza virus epitopes. We evaluated a novel T cell-inducing nucleoside-modified messenger RNA (mRNA) vaccine that encodes the conserved nucleoprotein, matrix protein 1, and polymerase basic protein 1 of an H1N1 influenza virus. To mimic the human situation, we applied the mRNA vaccine as a prime-boost regimen in naïve ferrets (mimicking young children) and as a booster in influenza-experienced ferrets (mimicking adults). The vaccine induced and boosted broadly reactive T cells in the circulation, bone marrow, and respiratory tract. Booster vaccination enhanced protection against heterosubtypic infection with a potential pandemic H7N9 influenza virus in influenza-experienced ferrets. Our findings show that mRNA vaccines encoding internal influenza virus proteins represent a promising strategy to induce broadly protective T cell immunity against influenza viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Niño , Animales , Humanos , Preescolar , Hurones/genética , Gripe Humana/prevención & control , ARN Mensajero/genética , Subtipo H7N9 del Virus de la Influenza A/genética , Linfocitos T
5.
Microbiol Spectr ; 10(6): e0138622, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36301107

RESUMEN

Nonpharmaceutical interventions (NPIs) to contain the SARS-CoV-2 pandemic drastically reduced human-to-human interactions, decreasing the circulation of other respiratory viruses, as well. Consequently, influenza virus circulation, which is normally responsible for 3 to 5 million hospitalizations per year globally, was significantly reduced. With the downscaling of the NPI countermeasures, there is a concern for increased influenza disease, particularly in individuals suffering from postacute effects of SARS-CoV-2 infection. To investigate this, we performed a sequential influenza H1N1 infection 4 weeks after an initial SARS-CoV-2 infection in ferrets. Upon H1N1 infection, ferrets that were previously infected with SARS-CoV-2 showed an increased tendency to develop clinical signs, compared to the control H1N1-infected animals. A histopathological analysis indicated only a slight increase for type II pneumocyte hyperplasia and bronchitis. Thus, the effects of the sequential infection appeared minor. However, ferrets were infected with B.1.351-SARS-CoV-2, the beta variant of concern, which replicated poorly in our model. The histopathology of the respiratory organs was mostly resolved 4 weeks after the SARS-CoV-2 infection, with only reminiscent histopathological features in the upper respiratory tract. Nevertheless, SARS-CoV-2 specific cellular and humoral responses were observed, confirming an established infection. On account of a modest trend toward the enhancement of the influenza disease, even upon a mild SARS-CoV-2 infection, our findings suggest that a stronger SARS-CoV-2 infection and its consequent, long-term effects could have a greater impact on the outcome of disease after a sequential influenza infection. Hence, the influenza vaccination of individuals suffering from postacute SARS-CoV-2 infection effects may be considered an avertible measure for such a scenario. IMPORTANCE During the COVID-19 pandemic, the use of face masks, social distancing, and isolation were effective not only in decreasing the circulation of SARS-CoV-2 but also in reducing other respiratory viruses, such as influenza. With fewer restrictions currently in place, influenza is slowly returning. In the meantime, people who are still suffering from long-COVID could be more vulnerable to an influenza virus infection and could develop a more severe influenza disease. This study provides directions to the effect of a previous SARS-CoV-2 exposure on influenza disease severity in a ferret model. This model is highly valuable to test sequential infections under controlled settings for translation to humans. We could not induce clear long-term COVID-19 effects, as the SARS-CoV-2 infections in the ferrets were mild. However, we still observed a slight increase in influenza disease severity compared to ferrets that had not encountered SARS-CoV-2 before. Therefore, it may be advisable to include long-COVID patients as a risk group for influenza vaccination.


Asunto(s)
COVID-19 , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Animales , Humanos , SARS-CoV-2 , Hurones , Síndrome Post Agudo de COVID-19 , Pandemias
6.
J Virol ; 96(14): e0073222, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35862678

RESUMEN

H2N2 influenza virus, the causative agent of the 1957 "Asian flu" pandemic, has disappeared from circulation. However, H2-influenza viruses are still circulating in avian reservoirs. Combined with the waning of H2N2-specific immunity in the human population, there is a risk of reintroduction of H2N2 influenza virus. Vaccines could help in preventing a future pandemic, but to assess their efficacy animal models are required. We therefore set out to expand the ferret model for H2N2 influenza disease by infecting ferrets intranasally or intratracheally with four different H2N2 viruses to investigate their influence on the severity of disease. The H2N2 viruses were collected either during the pandemic or near the end of H2N2 circulation and covered both clade I and clade II viruses. Infection of ferrets with the different viruses showed that viral replication, disease, and pathology differed markedly between virus isolates and infection routes. Intranasal inoculation induced a severe to mild rhinitis, depending on the virus isolate, and did not lead to lung infection or pathology. When administered intratracheally, isolates that successfully replicated in the lower respiratory tract (LRT) induced a nonlethal disease that resembles that of a moderate pneumonia in humans. Differences in viral replication and disease between viruses could be associated with their binding preference for α2,3- and α2,6-sialic acid. The model presented here could facilitate the development of a new generation of H2N2 influenza vaccines. IMPORTANCE In 1957 the world was subjected to a pandemic caused by an influenza A virus of the subtype H2N2. Although the virus disappeared in 1968, H2 viruses continue to circulate in avian reservoirs. It is therefore possible that the H2N2 influenza virus will be reintroduced into the human population, which can lead to another pandemic. The impact of a new H2N2 influenza pandemic can be mitigated by vaccination. However, these vaccines first need to be developed and tested in animal models. In preparation for this, we expanded the ferret model to mimic the different facets of human H2N2 influenza infection and disease. This model can be used for the development and evaluation of new H2N2 influenza vaccines.


Asunto(s)
Subtipo H2N2 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Replicación Viral , Animales , Aves , Modelos Animales de Enfermedad , Hurones/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza , Humanos , Subtipo H2N2 del Virus de la Influenza A/fisiología , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae/patología , Vacunación
7.
J Med Chem ; 65(4): 2836-2847, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34328726

RESUMEN

The SARS-CoV-2 viral spike protein S receptor-binding domain (S-RBD) binds ACE2 on host cells to initiate molecular events, resulting in intracellular release of the viral genome. Therefore, antagonists of this interaction could allow a modality for therapeutic intervention. Peptides can inhibit the S-RBD:ACE2 interaction by interacting with the protein-protein interface. In this study, protein contact atlas data and molecular dynamics simulations were used to locate interaction hotspots on the secondary structure elements α1, α2, α3, ß3, and ß4 of ACE2. We designed a library of discontinuous peptides based upon a combination of the hotspot interactions, which were synthesized and screened in a bioluminescence-based assay. The peptides demonstrated high efficacy in antagonizing the SARS-CoV-2 S-RBD:ACE2 interaction and were validated by microscale thermophoresis which demonstrated strong binding affinity (∼10 nM) of these peptides to S-RBD. We anticipate that such discontinuous peptides may hold the potential for an efficient therapeutic treatment for COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Péptidos/farmacología , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Sitios de Unión/efectos de los fármacos , Células Cultivadas , Células HEK293 , Humanos , Modelos Moleculares , Péptidos/síntesis química , Péptidos/química , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo
8.
Front Immunol ; 12: 750229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745122

RESUMEN

Improving COVID-19 intervention strategies partly relies on animal models to study SARS-CoV-2 disease and immunity. In our pursuit to establish a model for severe COVID-19, we inoculated young and adult male ferrets intranasally or intratracheally with SARS-CoV-2. Intranasal inoculation established an infection in all ferrets, with viral dissemination into the brain and gut. Upon intratracheal inoculation only adult ferrets became infected. However, neither inoculation route induced observable COVID-19 symptoms. Despite this, a persistent inflammation in the nasal turbinates was prominent in especially young ferrets and follicular hyperplasia in the bronchi developed 21 days post infection. These effects -if sustained- might resemble long-COVID. Respiratory and systemic cellular responses and antibody responses were induced only in animals with an established infection. We conclude that intranasally-infected ferrets resemble asymptomatic COVID-19 and possibly aspects of long-COVID. Combined with the increasing portfolio to measure adaptive immunity, ferrets are a relevant model for SARS-CoV-2 vaccine research.


Asunto(s)
Bronquios/patología , COVID-19/complicaciones , COVID-19/inmunología , Hurones/inmunología , SARS-CoV-2/fisiología , Administración Intranasal , Factores de Edad , Animales , Enfermedades Asintomáticas , Modelos Animales de Enfermedad , Hurones/virología , Humanos , Hiperplasia , Inmunidad Celular , Inmunidad Humoral , Inyección Intratimpánica , Masculino , Internalización del Virus , Síndrome Post Agudo de COVID-19
9.
Commun Biol ; 3(1): 564, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037319

RESUMEN

Traditional influenza vaccines primarily induce a narrow antibody response that offers no protection against heterosubtypic infections. Murine studies have shown that T cells can protect against a broad range of influenza strains. However, ferrets are a more potent model for studying immune correlates of protection in influenza infection. We therefore set out to investigate the role of systemic and respiratory T cells in the protection against heterosubtypic influenza A infections in ferrets. H1N1-priming induced systemic and respiratory T cells that responded against pandemic H2N2 and correlated with reduced viral replication and disease. CD8-positive T cell responses in the upper and lower respiratory tract were exceptionally high. We additionally confirmed that H2N2-responsive T cells are present in healthy human blood donors. These findings underline the importance of the T cell response in influenza immunity and show that T cells are a potent target for future universal influenza vaccines.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H2N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Reacciones Cruzadas/inmunología , Femenino , Hurones , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H2N2 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Pulmón/citología , Pulmón/inmunología , Masculino , Infecciones por Orthomyxoviridae/prevención & control , Sistema Respiratorio/citología , Sistema Respiratorio/inmunología , Estaciones del Año , Replicación Viral/inmunología
10.
NPJ Vaccines ; 5(1): 38, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32411401

RESUMEN

Until universal influenza vaccines become available, pandemic preparedness should include developing classical vaccines against potential pandemic influenza subtypes. We here show that addition of SWE adjuvant, a squalene-in-water emulsion, to H7N9 split influenza vaccine clearly enhanced functional antibody responses in ferrets. These were cross-reactive against H7N9 strains from different lineages and newly emerged H7N9 variants. Both vaccine formulations protected in almost all cases against severe pneumonia induced by intratracheal infection of ferrets with H7N9 influenza; however, the SWE adjuvant enhanced protection against virus replication and disease. Correlation analysis and curve fitting showed that both VN- and NI-titers were better predictors for protection than HI-titers. Moreover, we show that novel algorithms can assist in better interpretation of large data sets generated in preclinical studies. Cluster analysis showed that the adjuvanted vaccine results in robust immunity and protection, whereas the response to the non-adjuvanted vaccine is heterogeneous, such that the protection balance may be more easily tipped toward severe disease. Finally, cluster analysis indicated that the dose-sparing capacity of the adjuvant is at least a factor six, which greatly increases vaccine availability in a pandemic situation.

11.
J Clin Virol ; 128: 104412, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32416600

RESUMEN

The final months of 2019 witnessed the emergence of a novel coronavirus in the human population. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has since spread across the globe and is posing a major burden on society. Measures taken to reduce its spread critically depend on timely and accurate identification of virus-infected individuals by the most sensitive and specific method available, i.e. real-time reverse transcriptase PCR (RT-PCR). Many commercial kits have recently become available, but their performance has not yet been independently assessed. The aim of this study was to compare basic analytical and clinical performance of selected RT-PCR kits from seven different manufacturers (Altona Diagnostics, BGI, CerTest Biotec, KH Medical, PrimerDesign, R-Biopharm AG, and Seegene). We used serial dilutions of viral RNA to establish PCR efficiency and estimate the 95 % limit of detection (LOD95). Furthermore, we ran a panel of SARS-CoV-2-positive clinical samples (n = 13) for a preliminary evaluation of clinical sensitivity. Finally, we used clinical samples positive for non-coronavirus respiratory viral infections (n = 6) and a panel of RNA from related human coronaviruses to evaluate assay specificity. PCR efficiency was ≥96 % for all assays and the estimated LOD95 varied within a 6-fold range. Using clinical samples, we observed some variations in detection rate between kits. Importantly, none of the assays showed cross-reactivity with other respiratory (corona)viruses, except as expected for the SARS-CoV-1 E-gene. We conclude that all RT-PCR kits assessed in this study may be used for routine diagnostics of COVID-19 in patients by experienced molecular diagnostic laboratories.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Betacoronavirus , COVID-19 , Humanos , Pandemias , Neumonía Viral , ADN Polimerasa Dirigida por ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2
12.
Front Immunol ; 9: 525, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593747

RESUMEN

Influenza peptide antigens coding for conserved T cell epitopes have the capacity to induce cross-protective influenza-specific immunity. Short peptide antigens used as a vaccine, however, often show poor immunogenicity. In this study, we demonstrate that whole-inactivated influenza virus (WIV) acts as an adjuvant for influenza peptide antigens, as shown by the induction of peptide-specific CD8+ T cells in HLA-A2.1 transgenic mice upon vaccination with the influenza-M1-derived GILGFVFTL peptide (GIL), formulated with WIV. By screening various concentrations of GIL and WIV, we found that both components contributed to the GIL-specific T cell response. Whereas co-localization of the peptide antigen and WIV adjuvant was found to be important, neither physical association between peptide and WIV nor fusogenic activity of WIV were relevant for the adjuvant effect of WIV. We furthermore show that WIV may adjuvate T cell responses to a variety of peptides, using pools of either conserved wild-type influenza peptides or chemically altered peptide ligands. This study shows the potential of WIV as an adjuvant for influenza peptides. The simple formulation process and the solid safety record of WIV make this an attractive adjuvant for T cell peptides, and may also be used for non-influenza antigens.


Asunto(s)
Adyuvantes Inmunológicos , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Subtipo H5N1 del Virus de la Influenza A , Péptidos/inmunología , Inactivación de Virus , Animales , Femenino , Ratones Transgénicos
13.
PLoS One ; 11(6): e0156462, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27333291

RESUMEN

T cells are essential players in the defense against infection. By targeting the MHC class I antigen-presenting pathway with peptide-based vaccines, antigen-specific T cells can be induced. However, low immunogenicity of peptides poses a challenge. Here, we set out to increase immunogenicity of influenza-specific CD8+ T cell epitopes. By substituting amino acids in wild type sequences with non-proteogenic amino acids, affinity for MHC can be increased, which may ultimately enhance cytotoxic CD8+ T cell responses. Since preventive vaccines against viruses should induce a broad immune response, we used this method to optimize influenza-specific epitopes of varying dominance. For this purpose, HLA-A*0201 epitopes GILGFVFTL, FMYSDFHFI and NMLSTVLGV were selected in order of decreasing MHC-affinity and dominance. For all epitopes, we designed chemically enhanced altered peptide ligands (CPLs) that exhibited greater binding affinity than their WT counterparts; even binding scores of the high affinity GILGFVFTL epitope could be improved. When HLA-A*0201 transgenic mice were vaccinated with selected CPLs, at least 2 out of 4 CPLs of each epitope showed an increase in IFN-γ responses of splenocytes. Moreover, modification of the low affinity epitope NMLSTVLGV led to an increase in the number of mice that responded. By optimizing three additional influenza epitopes specific for HLA-A*0301, we show that this strategy can be extended to other alleles. Thus, enhancing binding affinity of peptides provides a valuable tool to improve the immunogenicity and range of preventive T cell-targeted peptide vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Epítopos Inmunodominantes/inmunología , Gripe Humana/inmunología , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Epítopos de Linfocito T/química , Citometría de Flujo , Polarización de Fluorescencia , Antígeno HLA-A2/inmunología , Antígeno HLA-A3/inmunología , Humanos , Epítopos Inmunodominantes/química , Activación de Linfocitos/inmunología , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Péptidos/química , Unión Proteica , Vacunación
14.
Mol Ther ; 24(5): 991-1002, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26796670

RESUMEN

Avian influenza viruses continue to cross the species barrier, and if such viruses become transmissible among humans, it would pose a great threat to public health. Since its emergence in China in 2013, H7N9 has caused considerable morbidity and mortality. In the absence of a universal influenza vaccine, preparedness includes development of subtype-specific vaccines. In this study, we developed and evaluated in ferrets an intranasal live attenuated influenza vaccine (LAIV) against H7N9 based on the A/Leningrad/134/17/57 (H2N2) cold-adapted master donor virus. We demonstrate that the LAIV is attenuated and safe in ferrets and induces high hemagglutination- and neuraminidase-inhibiting and virus-neutralizing titers. The antibodies against hemagglutinin were also cross-reactive with divergent H7 strains. To assess efficacy, we used an intratracheal challenge ferret model in which an acute severe viral pneumonia is induced that closely resembles viral pneumonia observed in severe human cases. A single- and two-dose strategy provided complete protection against severe pneumonia and prevented virus replication. The protective effect of the two-dose strategy appeared better than the single dose only on the microscopic level in the lungs. We observed, however, an increased lymphocytic infiltration after challenge in single-vaccinated animals and hypothesize that this a side effect of the model.


Asunto(s)
Bronconeumonía/prevención & control , Subtipo H7N9 del Virus de la Influenza A/fisiología , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Vacunas Atenuadas/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/inmunología , Bronconeumonía/inmunología , Modelos Animales de Enfermedad , Femenino , Hurones , Humanos , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Vacunas Atenuadas/inmunología , Replicación Viral/efectos de los fármacos
15.
Hum Vaccin Immunother ; 10(7): 1935-48, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25424803

RESUMEN

New and reemerging infectious diseases call for innovative and efficient control strategies of which fast vaccine design and development represent an important element. In emergency situations, when time is limited, identification and use of correlates of protection (COPs) may play a key role as a strategic tool for accelerated vaccine design, testing, and licensure. We propose that general rules for COP-based vaccine design can be extracted from the existing knowledge of protective immune responses against a large spectrum of relevant viral and bacterial pathogens. Herein, we focus on the applicability of this approach by reviewing the established and up-coming COPs for influenza in the context of traditional and a wide array of new vaccine concepts. The lessons learnt from this field may be applied more generally to COP-based accelerated vaccine design for emerging infections.


Asunto(s)
Vacunas Bacterianas/inmunología , Vacunas Bacterianas/aislamiento & purificación , Biomarcadores/análisis , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Vacunas Virales/inmunología , Vacunas Virales/aislamiento & purificación , Humanos , Factores de Tiempo
16.
PLoS One ; 9(7): e102339, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25058039

RESUMEN

H2N2 Influenza A caused the Asian flu pandemic in 1957, circulated for more than 10 years and disappeared from the human population after 1968. Given that people born after 1968 are naïve to H2N2, that the virus still circulates in wild birds and that this influenza subtype has a proven pandemic track record, H2N2 is regarded as a potential pandemic threat. To prepare for an H2N2 pandemic, here we developed and tested in mice and ferrets two live attenuated influenza vaccines based on the haemagglutinins of the two different H2N2 lineages that circulated at the end of the cycle, using the well characterized A/Leningrad/134/17/57 (H2N2) master donor virus as the backbone. The vaccine strains containing the HA and NA of A/California/1/66 (clade 1) or A/Tokyo/3/67 (clade 2) showed a temperature sensitive and cold adapted phenotype and a reduced reproduction that was limited to the respiratory tract of mice, suggesting that the vaccines may be safe for use in humans. Both vaccine strains induced haemagglutination inhibition titers in mice. Vaccination abolished virus replication in the nose and lung and protected mice from weight loss after homologous and heterologous challenge with the respective donor wild type strains. In ferrets, the live attenuated vaccines induced high virus neutralizing, haemagglutination and neuraminidase inhibition titers, however; the vaccine based on the A/California/1/66 wt virus induced higher homologous and better cross-reactive antibody responses than the A/Tokyo/3/67 based vaccine. In line with this observation, was the higher virus reduction observed in the throat and nose of ferrets vaccinated with this vaccine after challenge with either of the wild type donor viruses. Moreover, both vaccines clearly reduced the infection-induced rhinitis observed in placebo-vaccinated ferrets. The results favor the vaccine based on the A/California/1/66 isolate, which will be evaluated in a clinical study.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Subtipo H2N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Pandemias/prevención & control , Virus Reordenados/inmunología , Animales , Evaluación Preclínica de Medicamentos , Femenino , Hurones , Expresión Génica , Hemaglutininas Virales/genética , Hemaglutininas Virales/inmunología , Humanos , Inmunización , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos CBA , Neuraminidasa/genética , Neuraminidasa/inmunología , Nariz/efectos de los fármacos , Nariz/inmunología , Nariz/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/genética , Vacunas Atenuadas , Replicación Viral
17.
Front Immunol ; 5: 171, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24795718

RESUMEN

An effective immune response against viral infections depends on the activation of cytotoxic T cells that can clear infection by killing virus-infected cells. Proper activation of these T cells depends on professional antigen-presenting cells, such as dendritic cells (DCs). In this review, we will discuss the potential of peptide-based vaccines for prevention and treatment of viral diseases. We will describe features of an effective response against both acute and chronic infections, such as an appropriate magnitude, breadth, and quality and discuss requirements for inducing such an effective antiviral immune response. We will address modifications that affect presentation of vaccine components by DCs, including choice of antigen, adjuvants, and formulation. Furthermore, we will describe differences in design between preventive and therapeutic peptide-based vaccines. The ultimate goal in the design of preventive vaccines is to develop a universal vaccine that cross-protects against multiple strains of the virus. For therapeutic vaccines, cross-protection is of less importance, but enhancing existing T cell responses is essential. Although peptide vaccination is successful in inducing responses in human papillomavirus (HPV) infected patients, there are still several challenges such as choosing the right target epitopes, choosing safe adjuvants that improve immunogenicity of these epitopes, and steering the immune response in the desired direction. We will conclude with an overview of the current status of peptide vaccination, hurdles to overcome, and prospects for the future.

18.
Vaccine ; 29(32): 5153-62, 2011 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-21624413

RESUMEN

The H5N1-clade 1 influenza vaccine strain NIBRG-14 produces exceptionally low amounts of antigen, a problem recently encountered also for initial pandemic H1N1-2009 vaccine seeds. Here, we report on a strategy that may contribute to overcome this obstacle. Influenza vaccine viruses usually consist of two segments coding for the antigenic HA and NA proteins of a wild-type strain and the six residual internal gene segments of the vaccine donor strain A/PR/8/34 (PR8). To enhance the antigen yield from H5N1 vaccine virus we generated by reverse genetics a set of PR8-based reassortant viruses expressing the HA and NA segments of the prototypic strain A/Vietnam/1203/2004 and additional replacements of the internal M or PB1 genes of PR8. The reassortants were compared to the parental PR8 and H5N1 viruses in terms of growth in embryonated chicken eggs and the amount of incorporated antigenic HA protein. Compared to NIBRG-14, three out of six viruses displayed an increased replication in embryonated chicken eggs and higher HA content that was also maintained after ether/detergent extraction of virions. Electron microscopic analysis showed that the reassortment hardly affected particle shape and size. Two selected H5N1 reassortant viruses were investigated concerning their pathogenicity in ferrets and found to behave as low pathogenic as the PR8 donor strain. In conclusion, this study shows that replication and antigen content of PR8-derived H5N1 influenza vaccine viruses can be improved by incorporation of heterologous internal gene segments without compromising their attenuated character.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas Sintéticas/inmunología , Animales , Embrión de Pollo , Pollos , Femenino , Hurones/inmunología , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Microscopía Electrónica , Virus Reordenados/genética , Virus Reordenados/inmunología
19.
J Liposome Res ; 17(3-4): 173-82, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18027237

RESUMEN

Virosomes derived from influenza virus are reconstituted viral envelopes, which retain the receptor-binding and cell entry properties of the native virus, but lack the viral genetic material. These virosomes are of interest because of their potential use as vaccines or cellular delivery systems. However, in aqueous dispersion influenza virosomes have a relatively poor stability. Although freeze-drying of the virosomes could improve their stability, a lyoprotectant is required to preserve the structure and function of the virosomes during the lyophilization process as well as during subsequent storage of the dry powder formulation. In this study, inulin, a medium-chain oligosaccharide, was identified as an effective stabilizer of influenza virosomes. When inulin was added to an aqueous virosomal dispersion, the vesicular structure of the virosomes, with spike proteins protruding from the virosomal surface, as well as their membrane fusion activity were completely preserved during freeze-drying. When the freeze-drying process was performed from dispersions lacking a lyoprotectant, both structure and fusogenic properties of the virosomes were lost. Moreover, it was shown that the immunogenicity of inulin-stabilized virosomes was preserved. For example, dry powder formulations of virosomes retained HA potency for at least 12 weeks at 20 degrees C. Virosomes with encapsulated pDNA encoding for the eGFP reporter gene were also found to be stabilized by inulin. The fusion capacity and the transfection efficacy (determined in BHK-21 cells) could be preserved for 12 weeks during storage at 4 degrees C. It is concluded that freeze-drying in the presence of inulin as a lyoprotectant completely preserves the structure and function of influenza virosomes.


Asunto(s)
Liofilización , Liposomas , Orthomyxoviridae , Animales , Línea Celular , Cricetinae , Genes Reporteros
20.
Eur J Pharm Sci ; 32(1): 33-44, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17628452

RESUMEN

Influenza virosomes are reconstituted influenza virus envelopes that may be used as vaccines or as carrier systems for cellular delivery of therapeutic molecules. Here we present a procedure to generate influenza virosomes as a stable dry-powder formulation by freeze-drying (lyophilization) using an amorphous inulin matrix as a stabilizer. In the presence of inulin the structural integrity and fusogenic activity of virosomes were fully preserved during freeze-drying. For example, the immunological properties of the virosomes, i.e. the HA potency in vitro and the immunogenic potential in vivo, were maintained during lyophilization in the presence of inulin. In addition, compared to virosomes dispersed in buffer, inulin-formulated virosomes showed substantially prolonged preservation of the HA potency upon storage. Also the capacity of virosomes to mediate cellular delivery of macromolecules was maintained during lyophilization in the presence of inulin and upon subsequent storage. Specifically, when dispersed in buffer, virosomes with encapsulated plasmid DNA lost their transfection activity completely within 6 weeks, whereas their transfection activity was fully preserved for at least 12 weeks after incorporation in an inulin matrix. Thus, in the presence of inulin as a stabilizing agent, the shelf-life of influenza virosomes with and without encapsulated macromolecules was considerably prolonged. Formulation of influenza virosomes as a dry-powder is advantageous for storage and transport and offers the possibility to develop needle-free dosage forms, e.g. for oral, nasal, pulmonal, or dermal delivery.


Asunto(s)
Virus de la Influenza A/química , Inulina/química , Vacunas de Virosoma/inmunología , Virosomas/química , Cloruro de Amonio/farmacocinética , Animales , Formación de Anticuerpos/inmunología , Línea Celular , Centrifugación por Gradiente de Densidad , Cricetinae , Estabilidad de Medicamentos , Almacenaje de Medicamentos/métodos , Membrana Eritrocítica/inmunología , Membrana Eritrocítica/metabolismo , Femenino , Liofilización/métodos , Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Pruebas de Inhibición de Hemaglutinación , Virus de la Influenza A/inmunología , Virus de la Influenza A/ultraestructura , Fusión de Membrana , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Plásmidos/química , Plásmidos/genética , Transfección , Vacunas de Virosoma/administración & dosificación , Vacunas de Virosoma/química , Virosomas/genética , Virosomas/inmunología , Internalización del Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...