Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 2202, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38485927

RESUMEN

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.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Tirosina/análogos & derivados , Animales , Ratones , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Tirosina/metabolismo , Proteínas Virales de Fusión , Infecciones por Virus Sincitial Respiratorio/prevención & control
2.
Vaccine ; 41 Suppl 2: S7-S40, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37422378

RESUMEN

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.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Lactante , Niño , Humanos , Preescolar , Anticuerpos Monoclonales/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Inmunización Pasiva
3.
Am J Vet Res ; 83(11): 1-9, 2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-36173761

RESUMEN

OBJECTIVES: To compare initial titers, duration, and residual clinical protection of passively transferred bovine respiratory syncytial virus (BRSV) nasal immunoglobulin (Ig) G-1 and IgA, and serum neutralizing (SN) antibodies. ANIMALS: 40 three-month-old beef steers born either to unvaccinated or vaccinated cows. PROCEDURES: During the last trimester of gestation, cows were assigned randomly to either vaccinated or unvaccinated groups. Calves were grouped on the basis of whether they nursed colostrum from unvaccinated dams (NO-VACC group; n = 20) versus dams vaccinated with 2 doses of an inactivated BRSV vaccine (VACC group; n = 20). At 3 months of age, calves were challenged with BRSV. Respiratory signs were scored. Nasal BRSV IgG-1 and IgA and SN antibodies were compared before and after the challenge. The presence of BRSV in nasal secretions was evaluated by reverse transcription-PCR assays. RESULTS: Respiratory scores after BRSV challenge were similar between treatment groups. Nasal BRSV IgG-1 and SN antibodies were significantly greater in VACC calves at 48 hours of life; however, by 3 months of age, titers had decayed in both groups. Nasal BRSV IgA titers were minimal after colostrum intake and before the BRSV challenge, and increased in both groups after the challenge. The NO-VACC group had a significantly greater probability of shedding BRSV compared with VACC calves. CLINICAL RELEVANCE: At 3 months of age, titers of passively transferred BRSV antibodies in VACC and NO-VACC calves had decayed to nonprotective levels. Calves born to vaccinated dams had a decreased probability of BRSV shedding; however, this was not related to differences in SN or nasal BRSV antibody titers.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Bovino , Embarazo , Femenino , Bovinos , Animales , Calostro , Enfermedades de los Bovinos/prevención & control , Anticuerpos Antivirales , Inmunoglobulina G , Inmunoglobulina A , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/veterinaria
4.
Front Immunol ; 11: 1701, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849597

RESUMEN

Respiratory syncytial virus (RSV) infections represent a major burden of disease in infants and are the second most prevalent cause of death worldwide. Human milk immunoglobulins provide protection against RSV. However, many infants depend on processed bovine milk-based nutrition, which lacks intact immunoglobulins. We investigated the potential of bovine antibodies to neutralize human RSV and facilitate-cell immune activation. We show cow's milk IgG (bIgG) and Intravenous Immunoglobulin (IVIG) have a similar RSV neutralization capacity, even though bIgG has a lower pre-F to post-F binding ratio compared to human IVIG, with the majority of bIgG binding to pre-F. RSV is better neutralized with human IVIG. Consequently, we enriched RSV specific T cells by culturing human PBMC with a mixture of RSV peptides, and used these T cells to study the effect of bIgG and IVIG on the activation of pre-F-pecific T cells. bIgG facilitated in vitro T cell activation in a similar manner as IVIG. Moreover, bIgG was able to mediate T cell activation and internalization of pathogens, which are prerequisites for inducing an adaptive viral response. Using in vivo mouse experiments, we showed that bIgG is able to bind the murine activating IgG Fc Receptors (FcγR), but not the inhibiting FcγRII. Intranasal administration of the monoclonal antibody palivizumab, but also of bIgG and IVIG prevented RSV infection in mice. The concentration of bIgG needed to prevent infection was ~5-fold higher compared to IVIG. In conclusion, the data presented here indicate that functionally active bIgG facilitates adaptive antiviral T cell responses and prevents RSV infection in vitro and in vivo.


Asunto(s)
Antivirales/farmacología , Inmunoglobulina G/farmacología , Activación de Linfocitos/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Antivirales/aislamiento & purificación , Bovinos , Línea Celular , Calostro/inmunología , Modelos Animales de Enfermedad , Epítopos , Femenino , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulinas Intravenosas/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Embarazo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/patogenicidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología
5.
Expert Opin Biol Ther ; 20(9): 1073-1082, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32264720

RESUMEN

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.


Asunto(s)
Productos Biológicos/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Antivirales/inmunología , Antivirales/uso terapéutico , Niño , Humanos , Palivizumab/inmunología , Palivizumab/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Ribavirina/uso terapéutico , Proteínas Virales de Fusión/inmunología
6.
Sci Rep ; 10(1): 3653, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32107411

RESUMEN

Respiratory syncytial virus (RSV) infection in mouse and human lung is associated with oxidative injury and pathogenic inflammation. RSV impairs antioxidant responses by increasing the degradation of transcription factor NRF2, which controls the expression of several antioxidant enzyme (AOE) genes, including catalase. Since catalase is a key enzyme for the dismutation of virus-mediated generation of hydrogen peroxide (H2O2) we developed a model of intranasal supplementation of polyethylene glycol-conjugated catalase (PG-CAT) for RSV-infected mice. The results of our study show that PG-CAT supplementation was able to increase specific enzymatic activity along with reduction in H2O2 in the airways and had a significant protective effect against RSV-induced clinical disease and airway pathology. PG-CAT treated mice showed amelioration in airway obstruction, reduction in neutrophil elastase and inflammation. Improved airway hyperresponsiveness was also observed in mice that received PG-CAT as a treatment post-viral inoculation. In addition, PG-CAT greatly reduced the concentration of inflammatory cytokines and chemokines, including IL-1, TNF-α, IL-9, CXCL1, CCL2, and CCL5 in the bronchoalveolar lavage fluid of RSV-infected mice, without increasing viral replication in the lung. In conclusion, catalase supplementation may represent a novel pharmacologic approach to be explored in human for prevention or treatment of respiratory infections caused by RSV.


Asunto(s)
Catalasa/farmacología , Pulmón/metabolismo , Polietilenglicoles/farmacología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología
7.
Vaccine ; 36(52): 8069-8078, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30389195

RESUMEN

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.


Asunto(s)
Palivizumab/farmacología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Aluminio/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Afinidad de Anticuerpos , Antivirales/farmacología , Farmacorresistencia Viral , Epítopos/inmunología , Femenino , Masculino , Mutación , Nanopartículas/administración & dosificación , Fosfatos/inmunología , Ratas , Virus Sincitial Respiratorio Humano/genética , Sigmodontinae , Vacunación
8.
Arch Dis Child ; 103(12): 1163-1167, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30217858

RESUMEN

OBJECTIVE: To evaluate the impact of new reimbursement decisions for palivizumab treatment on respiratory syncytial virus (RSV) hospitalisations and the concomitant number of palivizumab prescriptions for infants aged <2 years. DESIGN: We compared the RSV hospitalisation rates in infants before and after implementation of new limitations during three RSV seasons 2014-2017. SETTING: Population aged <2 years at the beginning of each RSV seasons extracted from regional health systems (Lazio region, 2016, 5 898 124 inhabitants and 47 595 births). PATIENTS: Out of 70 323 infants, 5895 (8.4%) premature babies (gestational age (GA) <37 weeks) were followed before-after Italian Medicines Agency (AIFA)-2016 limitations. INTERVENTION: In 2016, AIFA, following the American Academy of Pediatrics guidelines, decided to limit coverage of palivizumab prophylaxis (GA ≤29 weeks). MAIN OUTCOMES MEASURES: Trend of hospitalisations by months and rate of RSV before-after new restrictions were analysed. Palivizumab prescriptions and costs for National Health Service (NHS) were considered. RESULTS: In a population of 284 902 aged <2 years, the number of hospitalisations due to RSV infection was 1729. Following AIFA-2016 limitations, a reduction in the number of RSV infection-based hospitalisations from 6.3/1000 (95% CI 6.0 to 6.7) to 5.5/1000 (95% CI 5.0 to 5.9) was observed. Palivizumab showed a concomitant reduction of 48% in the number of prescriptions (saving €750 000 for the NHS). No differences of GA, age on admission or severity of RSV infection were observed. CONCLUSIONS: Implementation of the new palivizumab reimbursement criteria was not associated with an increase in the RSV hospitalisation rate for children aged <2 years despite a significant reduction in the number of palivizumab prescriptions.


Asunto(s)
Antivirales/economía , Utilización de Medicamentos/tendencias , Hospitalización/tendencias , Reembolso de Seguro de Salud , Palivizumab/economía , Pautas de la Práctica en Medicina/tendencias , Infecciones por Virus Sincitial Respiratorio/prevención & control , Antivirales/uso terapéutico , Costos de los Medicamentos/tendencias , Utilización de Medicamentos/economía , Femenino , Hospitalización/economía , Humanos , Lactante , Recién Nacido , Italia/epidemiología , Masculino , Programas Nacionales de Salud/economía , Programas Nacionales de Salud/normas , Evaluación de Resultado en la Atención de Salud , Palivizumab/uso terapéutico , Guías de Práctica Clínica como Asunto , Pautas de la Práctica en Medicina/economía , Prevalencia , Infecciones por Virus Sincitial Respiratorio/economía , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/terapia
9.
Vaccine ; 36(20): 2876-2885, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29599087

RESUMEN

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.


Asunto(s)
Administración Intranasal , Chlorocebus aethiops , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Sigmodontinae , Animales , Evaluación Preclínica de Medicamentos/métodos , Femenino , Modelos Animales
10.
Viral Immunol ; 31(2): 133-141, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29323621

RESUMEN

Human parainfluenza viruses (family Paramyxoviridae), human metapneumovirus, and respiratory syncytial virus (family Pneumoviridae) infect most infants and children within the first few years of life and are the etiologic agents for many serious acute respiratory illnesses. These virus infections are also associated with long-term diseases that impact quality of life, including asthma. Despite over a half-century of vaccine research, development, and clinical trials, no vaccine has been licensed to date for the paramyxoviruses or pneumoviruses for the youngest infants. In this study, we describe the recent reclassification of paramyxoviruses and pneumoviruses into distinct families by the International Committee on the Taxonomy of Viruses. We also discuss some past unsuccessful vaccine trials and some currently preferred vaccine strategies. Finally, we discuss hurdles that must be overcome to support successful respiratory virus vaccine development for the youngest children.


Asunto(s)
Descubrimiento de Drogas/tendencias , Infecciones por Paramyxoviridae/prevención & control , Paramyxovirinae/inmunología , Pneumovirinae/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas Virales/inmunología , Vacunas Virales/aislamiento & purificación , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Infecciones por Paramyxoviridae/epidemiología , Paramyxovirinae/clasificación , Pneumovirinae/clasificación , Infecciones por Virus Sincitial Respiratorio/epidemiología
11.
Am J Epidemiol ; 187(7): 1490-1500, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351636

RESUMEN

We sought to determine the real-world effectiveness of respiratory syncytial virus (RSV) immunoprophylaxis in a population-based cohort to inform policy. The study population included infants born during 1996-2008 and enrolled in the Kaiser Permanente Northern California integrated health-care delivery system. During the RSV season (November-March), the date of RSV immunoprophylaxis administration and the following 30 days were defined as RSV immunoprophylaxis protected period(s), and all other days were defined as unprotected period(s). Numbers of bronchiolitis hospitalizations were determined using International Classification of Diseases, Ninth Revision, codes during RSV season. We used a proportional hazards model to estimate risk of bronchiolitis hospitalization when comparing infants' protected period(s) with unprotected period(s). Infants who had ever received RSV immunoprophylaxis had a 32% decreased risk of bronchiolitis hospitalization (adjusted hazard ratio = 0.68, 95% confidence interval: 0.46, 1.00) when protected periods were compared with unprotected periods. Infants with chronic lung disease (CLD) had a 52% decreased risk of bronchiolitis hospitalization (adjusted hazard ratio = 0.48, 95% confidence interval: 0.25, 0.94) when protected periods were compared with unprotected periods. Under the new 2014 American Academy of Pediatrics (AAP) guidelines, 48% of infants eligible for RSV immunoprophylaxis on the basis of AAP guidelines in place at birth would no longer be eligible, but nearly all infants with CLD would remain eligible. RSV immunoprophylaxis is effective in decreasing hospitalization. This association is greatest for infants with CLD, a group still recommended for receipt of RSV immunoprophylaxis under the new AAP guidelines.


Asunto(s)
Bronquiolitis Viral/prevención & control , Hospitalización/estadística & datos numéricos , Inmunización/estadística & datos numéricos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Antivirales/uso terapéutico , Bronquiolitis Viral/epidemiología , Bronquiolitis Viral/virología , California/epidemiología , Estudios de Cohortes , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/virología , Factores de Riesgo , Estaciones del Año , Resultado del Tratamiento
12.
Viral Immunol ; 31(2): 195-203, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29336703

RESUMEN

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.


Asunto(s)
Biomarcadores , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/aislamiento & purificación , Virus Sincitial Respiratorio Humano/inmunología , Humanos
13.
Hum Vaccin Immunother ; 14(1): 59-66, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-28933663

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/efectos adversos , Epítopos de Linfocito B/inmunología , Enfermedades del Complejo Inmune/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Adyuvantes Inmunológicos/química , Compuestos de Alumbre/efectos adversos , Compuestos de Alumbre/química , Animales , Preparaciones de Acción Retardada/efectos adversos , Preparaciones de Acción Retardada/química , Evaluación Preclínica de Medicamentos , Femenino , Enfermedades del Complejo Inmune/epidemiología , Inmunogenicidad Vacunal , Incidencia , Ratones , Aceites/efectos adversos , Aceites/química , Conejos , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/química , Vacunación/métodos , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/química , Vacunas Sintéticas/inmunología
14.
Vaccine ; 35(32): 3951-3958, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28624306

RESUMEN

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.


Asunto(s)
Evaluación Preclínica de Medicamentos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Inyecciones Intramusculares , Embarazo , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Sigmodontinae
15.
Hum Vaccin Immunother ; 13(7): 1586-1597, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28272978

RESUMEN

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.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Ratones Endogámicos BALB C , Potexvirus , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/aislamiento & purificación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/aislamiento & purificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
16.
Mucosal Immunol ; 10(6): 1569-1580, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28295020

RESUMEN

Regulation of respiratory mucosal immunity by microbial-derived metabolites has been a proposed mechanism that may provide airway protection. Here we examine the effect of oral Lactobacillus johnsonii supplementation on metabolic and immune response dynamics during respiratory syncytial virus (RSV) infection. L. johnsonii supplementation reduced airway T helper type 2 cytokines and dendritic cell (DC) function, increased regulatory T cells, and was associated with a reprogrammed circulating metabolic environment, including docosahexanoic acid (DHA) enrichment. RSV-infected bone marrow-derived DCs (BMDCs) from L. johnsonii-supplemented mice had altered cytokine secretion, reduced expression of co-stimulatory molecules, and modified CD4+ T-cell cytokines. This was replicated upon co-incubation of wild-type BMDCs with either plasma from L. johnsonii-supplemented mice or DHA. Finally, airway transfer of BMDCs from L. johnsonii-supplemented mice or with wild-type derived BMDCs pretreated with plasma from L. johnsonii-supplemented mice reduced airway pathological responses to infection in recipient animals. Thus L. johnsonii supplementation mediates airway mucosal protection via immunomodulatory metabolites and altered immune function.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Lactobacillus johnsonii/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Linfocitos T Reguladores/metabolismo , Células Th2/metabolismo , Animales , Células de la Médula Ósea/virología , Línea Celular , Microambiente Celular , Reprogramación Celular , Citocinas/metabolismo , Células Dendríticas/virología , Suplementos Dietéticos , Ácidos Docosahexaenoicos/metabolismo , Inmunomodulación , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/prevención & control , Linfocitos T Reguladores/inmunología , Células Th2/inmunología
17.
BMC Pregnancy Childbirth ; 16(1): 309, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27737646

RESUMEN

BACKGROUND: Early infancy is a high-risk period for severe acute respiratory infection (ARI), particularly in low-income countries with resource-limited health systems. Lower respiratory tract infection (LRTI) is commonly preceded by upper respiratory infection (URTI), and often caused by respiratory syncytial virus (RSV), influenza and other common community-acquired viral pathogens. Vitamin D status is a candidate modifiable early-life determinant of the host antiviral immune response and thus may influence the risk of ARI-associated morbidity in high-risk populations. METHODS/DESIGN: In the Maternal Vitamin D for Infant Growth (MDIG) study in Dhaka, Bangladesh (NCT01924013), 1300 pregnant women are randomized to one of five groups: placebo, 4200 IU/week, 16,800 IU/week, or 28,000 IU/week from 2nd trimester to delivery plus placebo from 0-6 months postpartum; or, 28,000 IU/week prenatal and until 6-months postpartum. In the Maternal Vitamin D for ARI in Infancy (MDARI) sub-study nested within the MDIG trial, trained personnel conduct weekly postnatal home visits to inquire about ARI symptoms and conduct a standardized clinical assessment. Supplementary home visits between surveillance visits are conducted when caregivers make phone notifications of new infant symptoms. Mid-turbinate nasal swab samples are obtained from infants who meet standardized clinical ARI criteria. Specimens are tested by polymerase chain reaction (PCR) for 8 viruses (influenza A/B, parainfluenza 1/2/3, RSV, adenovirus, and human metapneumovirus), and nasal carriage density of Streptococcus pneumoniae. The primary outcome is the incidence rate of microbiologically-positive viral ARI, using incidence rate ratios to estimate between-group differences. We hypothesize that among infants 0-6 months of age, the incidence of microbiologically-confirmed viral ARI will be significantly lower in infants whose mothers received high-dose prenatal/postpartum vitamin D supplements versus placebo. Secondary outcomes include incidence of ARI associated with specific pathogens (influenza A or B, RSV), clinical ARI, and density of pneumococcal carriage. DISCUSSION: If shown to reduce the risk of viral ARI in infancy, integration of maternal prenatal/postpartum vitamin D supplementation into antenatal care programs in South Asia may be a feasible primary preventive strategy to reduce the burden of ARI-associated morbidity and mortality in young infants. TRIAL REGISTRATION: NCT02388516 , registered March 9, 2015.


Asunto(s)
Suplementos Dietéticos , Enfermedades del Recién Nacido/prevención & control , Lactancia , Atención Prenatal/métodos , Infecciones del Sistema Respiratorio/prevención & control , Vitamina D/administración & dosificación , Vitaminas/administración & dosificación , Enfermedad Aguda , Adulto , Bangladesh , Lactancia Materna , Protocolos Clínicos , Femenino , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/virología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Embarazo , Estudios Prospectivos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Infecciones del Sistema Respiratorio/virología , Resultado del Tratamiento
18.
Sci Rep ; 6: 35851, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27767097

RESUMEN

The roots of Scutellaria baicalensis has been used as a remedy for inflammatory and infective diseases for thousands of years. We evaluated the antiviral activity against respiratory syncytial virus (RSV) infection, the leading cause of childhood infection and hospitalization. By fractionation and chromatographic analysis, we determined that baicalin was responsible for the antiviral activity of S. baicalensis against RSV infection. The concentration for 50% inhibition (IC50) of RSV infection was determined at 19.9 ± 1.8 µM, while the 50% cytotoxic concentration (CC50) was measured at 370 ± 10 µM. We then used a mouse model of RSV infection to further demonstrate baicalin antiviral effect. RSV infection caused significant lung injury and proinflammatory response, including CD4 and CD8 T lymphocyte infiltration. Baicalin treatment resulted in reduction of T lymphocyte infiltration and gene expression of proinflammatory factors, while the treatment moderately reduced RSV titers recovered from the lung tissues. T lymphocyte infiltration and cytotoxic T lymphocyte modulated tissue damage has been identified critical factors of RSV disease. The study therefore demonstrates that baicalin subjugates RSV disease through antiviral and anti-inflammatory effect.


Asunto(s)
Antivirales/uso terapéutico , Flavonoides/uso terapéutico , Lesión Pulmonar/prevención & control , Infecciones por Virus Sincitial Respiratorio/prevención & control , Scutellaria baicalensis/química , Animales , Antivirales/química , Antivirales/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Flavonoides/química , Flavonoides/farmacología , Humanos , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/fisiología , Scutellaria baicalensis/metabolismo , Internalización del Virus/efectos de los fármacos , Replicación Viral
19.
Vet Immunol Immunopathol ; 178: 88-98, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27496747

RESUMEN

Our objective was to evaluate the effect of an injectable trace mineral (ITM) supplement containing zinc, manganese, selenium, and copper on the humoral and cell mediated immune (CMI) responses to vaccine antigens in dairy calves receiving a modified-live viral (MLV) vaccine containing BVDV, BHV1, PI3V and BRSV. A total of 30 dairy calves (3.5 months of age) were administered a priming dose of the MLV vaccine containing BHV1, BVDV1 & 2, BRSV, PI3V, and an attenuated-live Mannheimia-Pasteurella bacterin subcutaneously (SQ). Calves were randomly assigned to 1 of 2 groups: (1) administration of ITM SQ (ITM, n=15) or (2) injection of sterile saline SQ (Control; n=15). Three weeks later, calves received a booster of the same vaccine combination SQ, and a second administration of ITM, or sterile saline, according to the treatment group. Blood samples were collected on days 0, 7, 14, 21, 28, 42, 56, and 90 post-vaccination for determination of antibody titer, viral recall antigen-induced IFN-γ production, and viral antigen-induced proliferation by peripheral blood mononuclear cells (PBMC). Administration of ITM concurrently with MLV vaccination resulted in higher antibody titers to BVDV1 on day 28 after priming vaccination compared to the control group (P=0.03). Calves treated with ITM showed an earlier enhancement in PBMC proliferation to BVDV1 following vaccination compared to the control group. Proliferation of PBMC after BVDV stimulation tended to be higher on day 14 after priming vaccination in calves treated with ITM than in the control group (P=0.08). Calves that received ITM showed higher PBMC proliferation to BRSV stimulation on day 7 after priming vaccination compared to the control group (P=0.01). Moreover, calves in the ITM group also had an enhanced production IFN-γ by PBMC after stimulation with BRSV on day 21 after priming vaccination compared to day 0 (P<0.01). In conclusion, administration of ITM concurrently with MLV vaccination in dairy calves resulted in increased antibody titer to BVDV1, and greater PBMC proliferation to BVDV1 and BRSV recall stimulation compared to the control group, suggesting that ITM might represent a promising tool to enhance the humoral and CMI responses to MLV vaccines in cattle.


Asunto(s)
Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/prevención & control , Virus de la Diarrea Viral Bovina/inmunología , Herpesvirus Bovino 1/inmunología , Virus Sincitial Respiratorio Bovino/inmunología , Oligoelementos/administración & dosificación , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Diarrea Mucosa Bovina Viral/inmunología , Diarrea Mucosa Bovina Viral/prevención & control , Bovinos , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/veterinaria , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Masculino , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/veterinaria , Vacunas Atenuadas/administración & dosificación
20.
Expert Rev Vaccines ; 15(1): 53-68, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26506139

RESUMEN

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.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Ensayos Clínicos como Asunto , Descubrimiento de Drogas/tendencias , Evaluación Preclínica de Medicamentos , Humanos , Infecciones por Virus Sincitial Respiratorio/epidemiología , Vacunas contra Virus Sincitial Respiratorio/aislamiento & purificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA