Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Commun ; 15(1): 2202, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38485927

RESUMO

Viral fusion proteins facilitate cellular infection by fusing viral and cellular membranes, which involves dramatic transitions from their pre- to postfusion conformations. These proteins are among the most protective viral immunogens, but they are metastable which often makes them intractable as subunit vaccine targets. Adapting a natural enzymatic reaction, we harness the structural rigidity that targeted dityrosine crosslinks impart to covalently stabilize fusion proteins in their native conformations. We show that the prefusion conformation of respiratory syncytial virus fusion protein can be stabilized with two engineered dityrosine crosslinks (DT-preF), markedly improving its stability and shelf-life. Furthermore, it has 11X greater potency as compared with the DS-Cav1 stabilized prefusion F protein in immunogenicity studies and overcomes immunosenescence in mice with simply a high-dose formulation on alum.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Tirosina/análogos & derivados , Animais , Camundongos , Anticorpos Neutralizantes , Anticorpos Antivirais , Tirosina/metabolismo , Proteínas Virais de Fusão , Infecções por Vírus Respiratório Sincicial/prevenção & controle
2.
Vaccine ; 41 Suppl 2: S7-S40, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37422378

RESUMO

Respiratory syncytial virus (RSV) is the predominant cause of acute lower respiratory infection (ALRI) in young children worldwide, yet no licensed RSV vaccine exists to help prevent the millions of illnesses and hospitalizations and tens of thousands of young lives taken each year. Monoclonal antibody (mAb) prophylaxis exists for prevention of RSV in a small subset of very high-risk infants and young children, but the only currently licensed product is impractical, requiring multiple doses and expensive for the low-income settings where the RSV disease burden is greatest. A robust candidate pipeline exists to one day prevent RSV disease in infant and pediatric populations, and it focuses on two promising passive immunization approaches appropriate for low-income contexts: maternal RSV vaccines and long-acting infant mAbs. Licensure of one or more candidates is feasible over the next one to three years and, depending on final product characteristics, current economic models suggest both approaches are likely to be cost-effective. Strong coordination between maternal and child health programs and the Expanded Program on Immunization will be needed for effective, efficient, and equitable delivery of either intervention. This 'Vaccine Value Profile' (VVP) for RSV is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships and multi-lateral organizations, and in collaboration with stakeholders from the WHO headquarters. All contributors have extensive expertise on various elements of the RSV VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Infecções Respiratórias , Lactente , Criança , Humanos , Pré-Escolar , Anticorpos Monoclonais/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Imunização Passiva
3.
Sci Rep ; 12(1): 22552, 2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-36581658

RESUMO

Human respiratory syncytial virus (HRSV) is a major cause of severe lower respiratory tract disease in infants and the elderly, yet no safe, effective vaccine is commercially available. Closely related bovine RSV (BRSV) causes respiratory disease in young calves, with many similar features to those seen in HRSV. We previously showed that a Newcastle disease virus (NDV)-vectored vaccine expressing the F glycoprotein of HRSV reduced viral loads in lungs of mice and cotton rats and protected from HRSV. However, clinical signs and pathogenesis of disease in laboratory animals following HRSV infection differs from that observed in human infants. Thus, we examined whether a similar vaccine would protect neonatal calves from BRSV infection. Codon-optimized rNDV vaccine (rNDV-BRSV Fopt) was constructed and administered to colostrum-deprived calves. The rNDV-BRSV Fopt vaccine was well-tolerated and there was no evidence of vaccine-enhanced disease in the upper airways or lungs of these calves compared to the non-vaccinated calves. We found two intranasal doses reduces severity of gross and microscopic lesions and decreases viral load in the lungs. Furthermore, serum neutralizing antibodies were generated in vaccinated calves. Finally, reduced lung CXC chemokine levels were observed in vaccinated calves after BRSV challenge. In summary, we have shown that rNDV-BRSV Fopt vaccine is safe in colostrum-deprived calves, and is effective in reducing lung lesions, and decreasing viral load in upper respiratory tract and lungs after challenge.


Assuntos
Doenças dos Bovinos , Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Bovino , Vírus Sincicial Respiratório Humano , Feminino , Gravidez , Animais , Bovinos , Humanos , Idoso , Vírus da Doença de Newcastle , Colostro , Vacinas contra Vírus Sincicial Respiratório/genética , Anticorpos Antivirais , Doenças dos Bovinos/prevenção & controle
4.
Vaccine ; 40(37): 5459-5463, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-35941034

RESUMO

INTRODUCTION: Pregnant individuals are frequently excluded from clinical trials. Yet, inclusion of Pregnant individuals is of interest in vaccinology including during health crisis. Promotion of clinical trials by midwives may facilitate the decision making of Pregnant individuals. Attitudes of midwives about pregnant individuals participation in a vaccine clinical trial have been little explored. METHODS: We conducted an anonymous survey from the 11th of September to the 11th of November 2020. Primary endpoint was the willingness to encourage Pregnant individuals to participate in a hypothetical respiratory syncytial virus (RSV) vaccine clinical trial. RESULTS: Among 398 midwives who answered the questionnaire, 113 (28.3 %) were likely to encourage Pregnant individuals to participate in the vaccine clinical trial, this proportion ranged from 25 % in senior midwives to 34.5 % among the students. After adjustment on age, parenthood, previous personal attitudes of vaccine hesitancy, and psychological antecedents of vaccinations (5C-model), the only predictor of the promotion of the clinical trial was the experience of vaccine education (evaluated by a 20-point score) with an adjusted odds ratio of 1.09 (1.01-1.18, p = 0.027) for a one-point increase. Vaccine hesitancy and psychological antecedents of vaccinations were not associated with a lower promotion of pregnant individuals trial participation by midwives. CONCLUSION: Few respondents were likely to encourage Pregnant individuals to participate in a vaccine clinical trial. Midwives who considered themselves to have a good training about vaccines were more prone to encourage Pregnant individuals to participate in a RSV vaccine clinical trial.


Assuntos
Tocologia , Vacinas contra Vírus Sincicial Respiratório , Feminino , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Gravidez , Inquéritos e Questionários , Vacinação/psicologia
5.
Expert Opin Biol Ther ; 20(9): 1073-1082, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32264720

RESUMO

INTRODUCTION: Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in young children and a substantial contributor to respiratory tract disease throughout life. Despite RSV being a high priority for vaccine development, there is currently no safe and effective vaccine available. There are many challenges to developing an RSV vaccine and there are limited antiviral drugs or biologics available for the management of infection. In this article, we review the antiviral treatments, vaccination strategies along with alternative therapies for RSV. AREAS COVERED: This review is a summary of the current antiviral and RSV vaccination approaches noting strategies and alternative therapies that may prevent or decrease the disease severity in RSV susceptible populations. EXPERT OPINION: This review discusses anti-RSV strategies given that no safe and efficacious vaccines are available, and therapeutic treatments are limited. Various biologicals that target for RSV are considered for disease intervention, as it is likely that it may be necessary to develop separate vaccines or therapeutics for each at-risk population.


Assuntos
Produtos Biológicos/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Antivirais/imunologia , Antivirais/uso terapêutico , Criança , Humanos , Palivizumab/imunologia , Palivizumab/uso terapêutico , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Ribavirina/uso terapêutico , Proteínas Virais de Fusão/imunologia
6.
Vaccine ; 36(52): 8069-8078, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30389195

RESUMO

Human respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in newborns, young children, elderly, and immune-compromised. The RSV fusion (F) glycoprotein is a major focus of vaccine development and the target of palivizumab (Synagis®) which is licensed as an immuno-prophylactic for use in newborn children at high risk of infection. However, clinical use of a narrowly targeted monoclonal antibodies leads to the generation of escape mutant strains that are fully resistant to neutralization by the antibody. Herein, we evaluated the RSV F nanoparticle vaccine (RSV F vaccine), produced as near-full-length, pre-fusogenic F trimers that form stable protein-detergent nanoparticles. The RSV F vaccine induces polyclonal antibodies that bind to antigenic site II as well as other epitopes known to be broadly neutralizing. Cotton rats immunized with the RSV F vaccine produced antibodies that were both neutralizing and protected against wild-type RSV infection, as well as against a palivizumab-resistant mutant virus. Use of aluminum phosphate adjuvant with the RSV F vaccine increased site II antibody avidity 100 to 1000-fold, which correlated with enhanced protection against challenge. The breadth of the vaccine-induced antibody response was demonstrated using competitive binding with monoclonal antibodies targeting antigenic sites Ø, II, IV, and VIII found on pre-fusion and post-fusion conformations of RSV F. In summary, we found the RSV F vaccine induced antibodies that bind to conserved epitopes including those defined as pre-fusion F specific; that use of adjuvant increased antibody avidity that correlated with enhanced protection in the cotton rat challenge model; and the polyclonal, high-avidity antibodies neutralized and protected against both wild-type and palivizumab-resistant mutant virus. These data support the ongoing clinical development of the aluminum phosphate adjuvanted RSV F nanoparticle vaccine.


Assuntos
Palivizumab/farmacologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Proteínas Virais de Fusão/imunologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alumínio/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Afinidade de Anticorpos , Antivirais/farmacologia , Farmacorresistência Viral , Epitopos/imunologia , Feminino , Masculino , Mutação , Nanopartículas/administração & dosagem , Fosfatos/imunologia , Ratos , Vírus Sincicial Respiratório Humano/genética , Sigmodontinae , Vacinação
7.
Vaccine ; 36(20): 2876-2885, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29599087

RESUMO

Respiratory syncytial virus (RSV) is the most common viral cause of bronchiolitis and pneumonia in children twelve months of age or younger and a significant cause of lower respiratory disease in older adults. As various clinical and preclinical candidates advance, cotton rats (Sigmodon hispidus) and non-human primates (NHP) continue to play a valuable role in RSV vaccine development, since both animals are semi-permissive to human RSV (HRSV). However, appropriate utilization of the models is critical to avoid mis-interpretation of the preclinical findings. Using a multimodality imaging approach; a fluorescence based optical imaging technique for the cotton rat and a nuclear medicine based positron emission tomography (PET) imaging technique for monkeys, we demonstrate that many common practices for intranasal immunization in both species result in inoculum delivery to the lower respiratory tract, which can result in poor translation of outcomes from the preclinical to the clinical setting. Using these technologies we define a method to limit the distribution of intranasally administered vaccines solely to the upper airway of each species, which includes volume restrictions in combination with injectable anesthesia. We show using our newly defined methods for strict intranasal immunization that these methods impact the immune responses and efficacy observed when compared to vaccination methods resulting in distribution to both the upper and lower respiratory tracts. These data emphasize the importance of well-characterized immunization methods in the preclinical assessment of intranasally delivered vaccine candidates.


Assuntos
Administração Intranasal , Chlorocebus aethiops , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Sigmodontinae , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Modelos Animais
8.
Viral Immunol ; 31(2): 195-203, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29336703

RESUMO

Correlates of protection (CoPs) can play a significant role in vaccine development by assisting the selection of vaccine candidates for clinical trials, supporting clinical trial design and implementation, and simplifying tests of vaccine modifications. Because of this important role in vaccine development, it is essential that CoPs be defined by well-designed immunogenicity and efficacy studies, with attention paid to benefits and limitations. The respiratory syncytial virus (RSV) field is unique in that a great deal of information about the humoral response is available from basic research and clinical studies. Polyclonal and monoclonal antibodies have been used routinely in the clinic to protect vulnerable infants from infection, providing a wealth of information about correlations between neutralizing antibodies and disease prevention. Considerations for the establishment of future CoPs to support RSV vaccine development in different populations are therefore discussed.


Assuntos
Biomarcadores , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas contra Vírus Sincicial Respiratório/isolamento & purificação , Vírus Sincicial Respiratório Humano/imunologia , Humanos
9.
Hum Vaccin Immunother ; 14(1): 59-66, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-28933663

RESUMO

Peptide antigens are combined with an adjuvant in order to increase immunogenicity in vivo. The immunogenicity and safety of a RSV vaccine formulated in a novel oil-based platform, DepoVax™ (DPX), was compared to an alum formulation. A peptide B cell epitope derived from RSV small hydrophobic ectodomain (SHe) served as the antigen. Both vaccines induced SHe-specific antibodies after immunization of mice. A single dose of the DPX-based formulation resulted in anti-SHe titres for up to 20 weeks. Boosting with Alum-SHe, but not with DPX-SHe, led to unexpected clinical signs such as decreased activity, cyanosis and drop in body temperature in mice but not in rabbits. The severity of adverse reactions correlated with magnitude of SHe-specific IgG immune responses and decreased complement component 3 plasma levels, indicating a type III hypersensitivity reaction. By RP-HPLC analysis, we found that only 8-20% of the antigen was found to be adsorbed to alum in vitro, indicating that this antigen is likely released systemically upon injection in vivo. Clinical signs were not observed in rabbits, indicating the response correlates with peptide dose relative to size of animal. These results suggest that peptide antigens targeted to produce B cell mediated response may result in increased incidence of type III hypersensitivity reactions when delivered in non-depot forming vaccines. The DPX formulation induced strong antibody titres to the antigen without causing adverse events, likely due to the strength of the depot in vivo, and demonstrates the potential safety and immunogenicity of this platform for B cell peptide antigens.


Assuntos
Adjuvantes Imunológicos/efeitos adversos , Epitopos de Linfócito B/imunologia , Doenças do Complexo Imune/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Adjuvantes Imunológicos/química , Compostos de Alúmen/efeitos adversos , Compostos de Alúmen/química , Animais , Preparações de Ação Retardada/efeitos adversos , Preparações de Ação Retardada/química , Avaliação Pré-Clínica de Medicamentos , Feminino , Doenças do Complexo Imune/epidemiologia , Imunogenicidade da Vacina , Incidência , Camundongos , Óleos/efeitos adversos , Óleos/química , Coelhos , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/química , Vacinação/métodos , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/química , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia
10.
Vaccine ; 35(32): 3951-3958, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28624306

RESUMO

Maternal immunization directed to control RSV infection in newborns and infants is an appealing vaccination strategy currently under development. In this work we have modeled maternal vaccination against RSV in cotton rats (CR) to answer two fundamental questions on maternal vaccine safety. We tested (i), whether a known, unsafe RSV vaccine (i.e., FI-RSV Lot 100 vaccine) induces vaccine enhanced disease in the presence of passively transferred, RSV maternal immunity, and (ii) whether the same FI-RSV vaccine could induce vaccine enhanced disease in CR litters when used to immunize their RSV-primed mothers. Our data show that FI-RSV immunization of pups with subsequent RSV infection results in vaccine-enhanced disease independent of whether the pups were born to RSV-seropositive or RSV-seronegative mothers, and that FI-RSV immunization of RSV-seropositive mothers does not present a health risk to either the mother or the infant. Our study also raises a novel concern regarding infant immunization, namely that "safe" RSV vaccines (e.g., live RSV administered intramuscularly) may induce vaccine-enhanced disease in RSV-infected pups born to seropositive mothers. Finally, we describe for the first time a sharp decrease in RSV neutralizing antibody titers in immunized seropositive CR at the time of delivery. This decline may reflect maternal immune suppression, potentially pinpointing a window of increased vulnerability to RSV infection that could be alleviated by effective immunization of expectant mothers.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Injeções Intramusculares , Gravidez , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Sigmodontinae
11.
Hum Vaccin Immunother ; 13(7): 1586-1597, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28272978

RESUMO

The development of safe and potent vaccines for human respiratory syncytial virus (HRSV) is still a challenge for researchers worldwide. DNA-based immunization is currently a promising approach that has been used to generate human vaccines for different age groups. In this study, novel HRSV DNA vaccine candidates were generated and preclinically tested in BALB/c mice. Three different versions of the codon-optimized HRSV fusion (F) gene were individually cloned into the pPOE vector. The new recombinant vectors either express full-length (pPOE-F), secretory (pPOE-TF), or M282-90 linked (pPOE-FM2) forms of the F protein. Distinctive expression of the F protein was identified in HEp-2 cells transfected with the different recombinant vectors using ELISA and immunofluorescence. Mice immunization verified the potential for recombinant vectors to elicit significant levels of neutralizing antibodies and CD8+ T-cell lymphocytes. pPOE-TF showed higher levels of gene expression in cell culture and better induction of the humoral and cellular immune responses. Following virus challenge, mice that had been immunized with the recombinant vectors were able to control virus replication and displayed lower inflammation compared with mice immunized with empty pPOE vector or formalin-inactivated HRSV vaccine. Moreover, pulmonary cytokine profiles of mice immunized with the 3 recombinant vectors were similar to those of the mock infected group. In conclusion, recombinant pPOE vectors are promising HRSV vaccine candidates in terms of their safety, immunogenicity and protective efficiency. These data encourage further evaluation in phase I clinical trials.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Camundongos Endogâmicos BALB C , Potexvirus , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/isolamento & purificação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/isolamento & purificação , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
12.
Expert Rev Vaccines ; 15(1): 53-68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26506139

RESUMO

Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections causing pneumonia and bronchiolitis in infants. RSV also causes serious illness in elderly populations, immunocompromised patients and individuals with pulmonary or cardiac problems. The significant morbidity and mortality associated with RSV infection have prompted interest in RSV vaccine development. In the 1960s, a formalin-inactivated vaccine trial failed to protect children, and indeed enhanced pathology when naturally infected later with RSV. Hence, an alternative approach to traditional killed virus vaccines, which can induce protective immunity without serious adverse events, is desired. Several strategies have been explored in attempts to produce effective vaccine candidates including gene-based and subunit vaccines. Subunit-based vaccine approaches have shown promising efficacy in animal studies and several have reached clinical trials. The current stage of development of subunit-based vaccines against RSV is reviewed in this article.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Ensaios Clínicos como Assunto , Descoberta de Drogas/tendências , Avaliação Pré-Clínica de Medicamentos , Humanos , Infecções por Vírus Respiratório Sincicial/epidemiologia , Vacinas contra Vírus Sincicial Respiratório/isolamento & purificação , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/isolamento & purificação
13.
Vaccine ; 33(47): 6488-92, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26319066

RESUMO

BACKGROUND: Protection of newborns and young infants against RSV disease via maternal immunization mediated by transplacental transfer of antibodies is under evaluation in third-trimester pregnant women with the RSV recombinant F nanoparticle vaccine (RSV F vaccine). Since the hemichorial placental architecture in guinea pigs and humans is similar, the guinea pig model was employed to assess RSV F vaccine immunogenicity in pregnant sows and to compare RSV-specific maternal antibody levels in their pups. METHODS: Thirty (30) presumptive pregnant guinea pigs were immunized on gestational day 25 and 46 with placebo (PBS), 30µg RSV F, or 30µg RSV F+400µg aluminum phosphate. Sera at delivery/birth (sows/pups) and 15 and 30 days post-partum (pups) were analyzed for the presence of anti-F IgG, palivizumab-competitive antibody (PCA) and RSV/A microneutralization (MN). RESULTS: The rates of pregnancy and stillbirth were similar between controls and vaccinees. The vaccine induced high levels of anti-F IgG, PCA and MN in sows, with the highest levels observed in adjuvanted vaccinees. Placental transfer to pups was proportional to the maternal antibody levels, with concentration effects observed for all immune measures. CONCLUSIONS: The RSV F vaccine was safe and immunogenic in pregnant guinea pigs and supported robust transplacental antibody transfer to their pups. Relative concentration of antibodies in the pups was observed even in the presence of high levels of maternal antibody. Guinea pigs may be an important safety and immunogenicity model for preclinical assessment of candidate vaccines for maternal immunization.


Assuntos
Anticorpos Antivirais/sangue , Imunidade Materno-Adquirida , Exposição Materna , Vacinas contra Vírus Sincicial Respiratório/imunologia , Proteínas Virais de Fusão/imunologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alumínio/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Feminino , Cobaias , Imunoensaio , Imunoglobulina G/sangue , Fosfatos/administração & dosagem , Placebos/administração & dosagem , Gravidez , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos
14.
J Interferon Cytokine Res ; 34(11): 902-14, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25051168

RESUMO

Formalin-inactivated respiratory syncytial virus (FI-RSV) immunization is known to cause severe pulmonary inflammatory disease after subsequent RSV infection. Ginseng has been used in humans for thousands of years due to its potential health benefits. We investigated whether ginseng would have immune modulating effects on RSV infection in mice previously immunized with FI-RSV. Oral administration of mice with ginseng increased IgG2a isotype antibody responses to FI-RSV immunization, indicating T-helper type 1 (Th1) immune responses. Ginseng-treated mice that were nonimmunized or previously immunized with FI-RSV showed improved protection against RSV challenge compared with control mice without ginseng treatment. Ginseng-mediated improved clinical outcomes after live RSV infection were evidenced by diminished weight losses, decreased interleukin-4 cytokine production but increased interferon-γ production, modulation of CD3 T-cell populations toward a Th1 response, and reduced inflammatory response. Ginseng-mediated protective host immune modulation against RSV pulmonary inflammation was observed in different strains of wild-type and mutant mice. These results indicate that ginseng can modulate host immune responses to FI-RSV immunization and RSV infection, resulting in protective effects against pulmonary inflammatory disease.


Assuntos
Pneumopatias/prevenção & controle , Panax/imunologia , Extratos Vegetais/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vírus Sinciciais Respiratórios/imunologia , Animais , Feminino , Formaldeído/química , Células Hep G2 , Humanos , Imunização/efeitos adversos , Imunoglobulina G/sangue , Imunomodulação , Interferon gama/metabolismo , Interleucina-4/metabolismo , Pneumopatias/imunologia , Pneumopatias/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Extratos Vegetais/imunologia , Raízes de Plantas/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sinciciais Respiratórios/química , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Vacinas de Produtos Inativados/efeitos adversos
15.
Vaccine ; 31(3): 524-32, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23153449

RESUMO

OBJECTIVE: We performed a Phase 1 randomized, observer-blinded, placebo-controlled trial to evaluate the safety and immunogenicity of a recombinant respiratory syncytial virus (RSV) fusion (F) protein nanoparticle vaccine. METHODS: Six formulations with (5, 15, 30 and 60 µg) and without (30 and 60 µg) aluminum phosphate (AdjuPhos) were administered intramuscularly on day 0 and 30 in a dose escalating fashion to healthy adults 18-49 years of age. Solicited and unsolicited events were collected through day 210. Immunogenicity measures taken at day 0, 30 and 60 included RSV A and B microneutralization, anti-F IgG, antigenic site II peptide and palivizumab competitive antibodies. RESULTS: The vaccine was well-tolerated, with no evident dose-related toxicity or attributable SAEs. At day 60 both RSV A and B microneutralization was significantly increased in vaccinees versus placebo. Across all vaccinees there was a 7- to 19-fold increase in the anti-F IgG and a 7- to 24-fold increase in the antigenic site II binding and palivizumab competitive antibodies. CONCLUSIONS: The RSV F nanoparticle vaccine candidate was well tolerated without dose-related increases in adverse events. Measures of immunity indicate that neutralization, anti-RSV F IgG titers and palivizumab competing antibodies were induced at levels that have been associated with decreased risk of hospitalization. NCT01290419.


Assuntos
Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/imunologia , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Adolescente , Adulto , Compostos de Alumínio/administração & dosagem , Compostos de Alumínio/efeitos adversos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Biotecnologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Fosfatos/administração & dosagem , Fosfatos/efeitos adversos , Placebos/administração & dosagem , Placebos/efeitos adversos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/genética , Células Sf9 , Método Simples-Cego , Tecnologia Farmacêutica , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virossomais/administração & dosagem , Vacinas Virossomais/efeitos adversos , Vacinas Virossomais/genética , Vacinas Virossomais/imunologia , Adulto Jovem
16.
Virol J ; 7: 20, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20102602

RESUMO

BACKGROUND: Infectivity of retroviruses such as HIV-1 and MuLV can be abrogated by compounds targeting zinc finger motif in viral nucleocapsid protein (NC), involved in controlling the processivity of reverse transcription and virus infectivity. Although a member of a different viral family (Pneumoviridae), respiratory syncytial virus (RSV) contains a zinc finger protein M2-1 also involved in control of viral polymerase processivity. Given the functional similarity between the two proteins, it was possible that zinc finger-reactive compounds inactivating retroviruses would have a similar effect against RSV by targeting RSV M2-1 protein. Moreover, inactivation of RSV through modification of an internal protein could yield a safer whole virus vaccine than that produced by RSV inactivation with formalin which modifies surface proteins. RESULTS: Three compounds were evaluated for their ability to reduce RSV infectivity: 2,2'-dithiodipyridine (AT-2), tetraethylthiuram disulfide and tetramethylthiuram disulfide. All three were capable of inactivating RSV, with AT-2 being the most potent. The mechanism of action of AT-2 was analyzed and it was found that AT-2 treatment indeed results in the modification of RSV M2-1. Altered intramolecular disulfide bond formation in M2-1 protein of AT-2-treated RSV virions might have been responsible for abrogation of RSV infectivity. AT-2-inactivated RSV was found to be moderately immunogenic in the cotton rats S.hispidus and did not cause a vaccine-enhancement seen in animals vaccinated with formalin-inactivated RSV. Increasing immunogenicity of AT-2-inactivated RSV by adjuvant (Ribi), however, led to vaccine-enhanced disease. CONCLUSIONS: This work presents evidence that compounds that inactivate retroviruses by targeting the zinc finger motif in their nucleocapsid proteins are also effective against RSV. AT-2-inactivated RSV vaccine is not strongly immunogenic in the absence of adjuvants. In the adjuvanted form, however, vaccine induces immunopathologic response. The mere preservation of surface antigens of RSV, therefore may not be sufficient to produce a highly-efficacious inactivated virus vaccine that does not lead to an atypical disease.


Assuntos
Antivirais/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Dedos de Zinco , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/metabolismo , 2,2'-Dipiridil/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antivirais/metabolismo , Esqueleto da Parede Celular/administração & dosagem , Fatores Corda/administração & dosagem , Dissulfetos/metabolismo , Dissulfetos/farmacologia , Dissulfiram/metabolismo , Dissulfiram/farmacologia , Lipídeo A/administração & dosagem , Lipídeo A/análogos & derivados , Ligação Proteica , Ratos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/fisiologia , Sigmodontinae/virologia , Tiram/metabolismo , Tiram/farmacologia , Vacinas Atenuadas/imunologia , Proteínas Virais/metabolismo
17.
Vaccine ; 27(42): 5913-9, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19651171

RESUMO

We studied the safety and immunogenicity of a Respiratory Syncytial Virus (RSV)-A vaccine containing subunit antigens F, G and M in older persons, and its effect on influenza vaccine immunogenicity. In a dose-ranging, placebo-controlled, blinded trial 561 adults > or =65 years of age at five Canadian sites were randomized to one intramuscular injection of either 100, 50 or 25 microg RSV-A-alum vaccine or 100 microg non-adjuvanted RSV-A vaccine, or alum-placebo. All participants were offered inactivated influenza vaccine on day 32. Immunization was well tolerated and reactogenicity was similar between the RSV and influenza vaccines and the alum-placebo. Only the 100 microg non-adjuvanted RSV vaccine achieved the primary immunogenicity outcome of eliciting a > or =4-fold rise in neutralizing antibody (NA) titres against RSV-A in > or =50% of participants at day 32. Geometric mean titres against RSV-A and -B at all points were comparable in 100 microg adjuvanted and non-adjuvanted groups. At day 32, a > or =4-fold haemagluttinin inhibition (HI) antibody response or HI > or =40 to Influenza (A-H3N2) was seen in >74% of participants; no difference was seen between groups. A subunit non-alum-containing RSV-A vaccine was well tolerated in a large population > or =65 years and did not interfere with influenza vaccine immunogenicity. This RSV-A-based vaccine demonstrated NA rise which could provide seasonal protection against severe RSV illnesses from RSV-A or -B and warrants further testing to determine its efficacy in the prevention of clinical illness in elderly persons.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alumínio/imunologia , Fosfatos/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Relação Dose-Resposta Imunológica , Feminino , Testes de Inibição da Hemaglutinação , Humanos , Masculino , Testes de Neutralização , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vacinas de Subunidades Antigênicas/imunologia
18.
Vaccine ; 27(23): 3045-52, 2009 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-19428918

RESUMO

Agonists for TLR7, TLR8, and TLR9 have been shown to enhance vaccine immunogenicity. We evaluated the impact of TLR activation on RSV disease in a murine model by administering TLR7/8 and TLR9 agonists during FI-RSV immunization or RSV infection. CpG administered during immunization reduced disease following challenge as evidenced by decreased lung pathology, illness, and cytokines. In marked contrast, TLR7/8 agonist had little impact. To evaluate potential therapeutic use, TLR agonists were administered during primary infection. Although type 2 cytokine responses decreased and type 1 cytokines and MIP-1-alpha/beta increased, both TLR7/8 and TLR9 agonists increased clinical symptoms and pulmonary inflammation when administered during primary infection. Thus, TLR9-induced signaling during FI-RSV immunization reduced vaccine-enhanced disease whereas immunostimulatory properties of TLR agonists enhanced disease severity when used during RSV infection. Immunomodulation elicited by TLR9 agonist confirms the adjuvant potential of TLR agonists during RSV immunization. However, in contrast to work done with HIV-1 vaccines, the inability of TLR7/8 agonist to boost type 1 vaccine-induced RSV immunity demonstrates pathogen-TLR specificity. These data reveal that the timing of administration of immunomodulatory agents is critical. Furthermore, these data underscore that amplification of anti-viral immune responses may result in immunopathology rather than immune-mediated protection.


Assuntos
Glicoproteínas de Membrana/agonistas , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Vírus Sinciciais Respiratórios/patogenicidade , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas , Adjuvantes Imunológicos/uso terapêutico , Animais , Formação de Anticorpos , Linhagem Celular , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Imidazóis/farmacologia , Imunidade Celular , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Virulência
19.
Expert Rev Vaccines ; 6(2): 169-82, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17408367

RESUMO

Pneumoviruses, which are viruses of the family Paramyxoviridae, subfamily Pneumovirinae, are pathogens that infect the respiratory tract of their host species. The human pneumovirus pathogen, human respiratory syncytial virus (RSV), has counterparts that infect cows (bovine RSV), sheep (ovine RSV), goats (caprine RSV) and rodents (pneumonia virus of mice). Each pneumovirus is host specific and results in a spectrum of disease, ranging from mild upper-respiratory illness to severe bronchiolitis and pneumonia with significant morbidity and mortality. Given the public health burden caused by human RSV and the concomitant agricultural impact of bovine RSV, these two viruses are considered as prime targets for the development of safe and effective vaccines. In this review, we describe the strategies used to develop vaccines against human and bovine RSV and introduce the pneumonia virus mouse model as a novel and invaluable tool for preclinical studies and new vaccine strategies.


Assuntos
Doenças dos Bovinos/prevenção & controle , Infecções por Pneumovirus/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Bovino/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vacinação , Adjuvantes Imunológicos/uso terapêutico , Animais , Bovinos , Doenças dos Bovinos/imunologia , Química Farmacêutica , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Camundongos , Vírus da Pneumonia Murina/imunologia , Infecções por Pneumovirus/imunologia , Infecções por Pneumovirus/veterinária , Infecções por Pneumovirus/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/veterinária , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinação/métodos , Vacinação/tendências , Vacinas Atenuadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA