Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
PLoS One ; 15(2): e0228572, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32045432

RESUMO

Human respiratory syncytial virus (RSV) and parainfluenza virus type 3 (HPIV3) are among the most common viral causes of childhood bronchiolitis and pneumonia worldwide, and lack effective antiviral drugs or vaccines. Recombinant (r) HPIV3 was modified to express the RSV fusion (F) glycoprotein, the major RSV neutralization and protective antigen, providing a live intranasal bivalent HPIV3/RSV vaccine candidate. This extends previous studies using a chimeric bovine-human PIV3 vector (rB/HPIV3). One advantage is that rHPIV3 expresses all of the HPIV3 antigens compared to only two for rB/HPIV3. In addition, the use of rHPIV3 as vector should avoid excessive attenuation following addition of the modified RSV F gene, which may occur with rB/HPIV3. To enhance its immunogenicity, RSV F was modified (i) to increase the stability of the prefusion (pre-F) conformation and (ii) by replacement of its transmembrane (TM) and cytoplasmic tail (CT) domains with those of HPIV3 F (H3TMCT) to increase incorporation in the vector virion. RSV F (+/- H3TMCT) was expressed from the first (F/preN) or the second (F/N-P) gene position of rHPIV3. The H3TMCT modification dramatically increased packaging of RSV F into the vector virion and, in hamsters, resulted in significant increases in the titer of high-quality serum RSV-neutralizing antibodies, in addition to the increase conferred by pre-F stabilization. Only F-H3TMCT/preN replication was significantly attenuated in the nasal turbinates by the RSV F insert. F-H3TMCT/preN, F/N-P, and F-H3TMCT/N-P provided complete protection against wt RSV challenge. F-H3TMCT/N-P exhibited the most stable and highest expression of RSV F, providing impetus for its further development.


Assuntos
Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/imunologia , Vacinas contra Vírus Sincicial Respiratório/genética , Proteínas Virais de Fusão/genética , Montagem de Vírus , Administração Intranasal , Animais , Chlorocebus aethiops , Cricetinae , Feminino , Humanos , Imunogenicidade da Vacina , Macaca mulatta , Mesocricetus , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/fisiologia , Estabilidade Proteica , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Células Vero , Proteínas Virais de Fusão/metabolismo
2.
Antiviral Res ; 162: 54-60, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550799

RESUMO

Human parainfluenza virus 3 (PIV3) and respiratory syncytial virus (RSV) are major causative agents of serious respiratory tract illness in newborns and infants. Maternal vaccination could be a promising approach to provide immediate protection against severe PIV3 and RSV infection in young infants. Previously, we demonstrated that maternal immunization with a subunit vaccine consisting of the RSV fusion (F) protein formulated with TriAdj, an adjuvant consisting of poly(I:C), immune defense regulatory peptide and polyphosphazene, protects newborn lambs from RSV. In the present study we evaluated the protective efficacy of a novel bivalent RSV-PIV3 vaccine candidate, FRipScHN/TriAdj, as a maternal vaccine against PIV3 infection in a neonatal lamb model. This vaccine consists of the pre-fusion form of the RSV F protein linked to the haemagglutinin-neuraminidase (HN) of PIV3, formulated with TriAdj. First, we successfully established PIV3 infection in neonatal lambs. Lambs infected with human PIV3 showed gross pathology, bronchointerstitial pneumonia and viral replication in the lungs. Subsequently, ewes were immunized with FRipScHN/TriAdj. RSV FRipSc- and PIV3 HN-specific antibodies with virus-neutralizing activity were detected in both the serum and the colostrum of the vaccinated ewes. The newborn lambs had RSV- and PIV3- neutralizing antibodies in their serum, which demonstrates that maternal antibodies were transferred to the neonates. At three days of age, the newborn lambs received an intrapulmonary challenge with PIV3. The lung pathology and virus production were significantly reduced in lambs that had received PIV3-specific maternal antibodies compared to lambs born to non-vaccinated ewes. These results suggest that maternal vaccination with a bivalent FRipScHN/TriAdj vaccine might be an effective method to provide protection against both PIV3 and RSV in neonates.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antivirais/sangue , Imunidade Materno-Adquirida , Vacinas contra Parainfluenza/administração & dosagem , Infecções por Respirovirus/veterinária , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/sangue , Feminino , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Gravidez , Vírus Sinciciais Respiratórios/genética , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/prevenção & controle , Ovinos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/imunologia
3.
Paediatr Drugs ; 19(6): 523-531, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28808938

RESUMO

Childhood vaccination has played a critical role in the reduction of morbidity and mortality from communicable diseases, including specific respiratory pathogens. Acute lower respiratory infection (ALRI) of both bacterial and viral aetiology continues to impact global child health. Key bacterial pathogens including Streptococcus pneumoniae and Haemophilus influenza type b are specifically targeted with current vaccination programmes, while at present there are less effective strategies for the prevention of viral disease. Influenza vaccines, including both live attenuated intranasal vaccines and inactivated influenza vaccines, are limited by seasonal strain variation and unsustained immunity. Research into the development of a universal influenza vaccine is ongoing; potential targets are the conserved regions of the virus such as the M2e antigen and hemagglutinin stalk. Respiratory syncytial virus (RSV) and parainfluenza virus 3 (PIV3) are the viral pathogens most commonly causing ALRI in children, particularly the infant population. Currently, no vaccine exists for either virus. Over the last decade, promising advances have been made. Protection of neonates via maternal RSV immunisation is being assessed in a phase III clinical trial, with many other candidates for RSV and PIV3 at less advanced stages of development.


Assuntos
Infecções Respiratórias/prevenção & controle , Vacinas Virais , Administração Intranasal , Criança , Humanos , Vacinas contra Influenza , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Vacinas contra Parainfluenza , Vacinas contra Vírus Sincicial Respiratório , Infecções Respiratórias/virologia , Vacinação , Vacinas Atenuadas
4.
J Vet Intern Med ; 30(1): 164-6, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26692461

RESUMO

BACKGROUND: Canine adenovirus 2, parainfluenza, and Bordetella bronchiseptica cause respiratory disease in dogs, and each has a modified live intranasal vaccine available. Molecular diagnostic assays to amplify specific nucleic acids are available for each of these agents. If positive molecular diagnostic assay results are common after vaccination, the positive predictive value of the diagnostic assays for disease would be decreased. OBJECTIVE: To determine the impact of administration of commercially available modified live topical adenovirus 2, B. bronchiseptica, and parainfluenza vaccine has on the results of a commercially available PCR panel. ANIMALS: Eight puppies from a research breeding facility negative for these pathogens. METHODS: Blinded prospective pilot study. Puppies were vaccinated with a single dose of modified live topical adenovirus 2, B. bronchiseptica, and parainfluenza and parenteral dose of adenovirus 2, canine distemper virus, and parvovirus. Nasal and pharyngeal swabs were collected on multiple days and submitted for PCR assay. RESULTS: Nucleic acids of all 3 organisms contained in the topical vaccine were detected from both samples multiple times through 28 days after vaccination with higher numbers of positive samples detected between days 3 and 10 after vaccination. CONCLUSIONS AND CLINICAL IMPORTANCE: Vaccine status should be considered when interpreting respiratory agent PCR results if modified live vaccines have been used. Development of quantitative PCR and wild-type sequencing are necessary to improve positive predictive value of these assays by distinguishing vaccinate from natural infection.


Assuntos
Adenoviridae , Vacinas Bacterianas/imunologia , Bordetella bronchiseptica , Doenças do Cão/prevenção & controle , Vacinas contra Parainfluenza/imunologia , Vacinas Virais/imunologia , Infecções por Adenoviridae/prevenção & controle , Infecções por Adenoviridae/veterinária , Administração Tópica , Animais , Infecções por Bordetella/prevenção & controle , DNA Bacteriano/genética , Vírus da Cinomose Canina/genética , Doenças do Cão/microbiologia , Doenças do Cão/virologia , Cães , Parvovirus/genética , Projetos Piloto , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/veterinária , RNA Viral/genética , Vacinação , Vacinas Atenuadas
5.
J Pediatric Infect Dis Soc ; 4(4): e143-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26582883

RESUMO

We conducted a phase I clinical trial (clinicaltrials.gov identifier, NCT00641017) of the experimental live-attenuated human parainfluenza virus type 1 (HPIV-1) vaccine rHPIV-1/84/del 170/942A sequentially in 3 groups: adults, HPIV-1-seropositive children, and HPIV-1-seronegative children, the target population for vaccination. rHPIV-1/84/del 170/942A was appropriately restricted in replication in adults and HPIV-1-seropositive children but was overattenuated (ie, insufficiently infectious and immunogenic) for HPIV-1-seronegative children.


Assuntos
Vacinas contra Parainfluenza/uso terapêutico , Infecções por Paramyxoviridae/prevenção & controle , Adulto , Anticorpos Antivirais/sangue , Pré-Escolar , Método Duplo-Cego , Humanos , Lactente , Vírus da Parainfluenza 1 Humana , Infecções por Paramyxoviridae/epidemiologia , Vacinas Atenuadas/uso terapêutico
6.
Pediatr Rev ; 35(6): 217-27; quiz 228, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24891595

RESUMO

• On the basis of strong epidemiologic evidence, influenza and parainfluenza viruses are responsible for significant morbidity and mortality in young infants and children and in persons with chronic medical conditions. (1)(4)(26)(27)(35). • On the basis of research evidence, influenza vaccines are effective in preventing disease in high-risk individuals. (8)(17)(18). • On the basis of strong research evidence, influenza vaccines are safe in young infants and children 6 months or older. (8)(15).• On the basis of research evidence, the use of corticosteroids and epinephrine is beneficial in the treatment of laryngotracheitis caused by parainfluenza viruses. (44)(45)(46)(47). • Strong evidence supports the use of influenza vaccines in pregnant mothers as a strategy to prevent disease in infants younger than 6 months. (17)(18)(19).


Assuntos
Vacinas contra Influenza/administração & dosagem , Influenza Humana/diagnóstico , Influenza Humana/prevenção & controle , Vacinas contra Parainfluenza/administração & dosagem , Infecções por Paramyxoviridae/diagnóstico , Infecções por Paramyxoviridae/prevenção & controle , Antivirais/uso terapêutico , Criança , Estudos Transversais , Diagnóstico Diferencial , Humanos , Influenza Humana/epidemiologia , Infecções por Paramyxoviridae/epidemiologia
7.
Vaccine ; 31(52): 6239-46, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24144478

RESUMO

Influenza virus and human parainfluenza virus (HPIV) are major etiologic agents of acute respiratory illness in young children. Inactivated and live attenuated influenza vaccines are approved in several countries, yet no vaccine is licensed for HPIV. We previously showed that a replication-incompetent PB2-knockout (PB2-KO) virus that possesses a reporter gene in the coding region of the PB2 segment can serve as a platform for a bivalent vaccine. To develop a bivalent vaccine against influenza and parainfluenza virus, here, we generated a PB2-KO virus possessing the hemagglutinin-neuraminidase (HN) glycoprotein of HPIV type 3 (HPIV3), a major surface antigen of HPIV, in its PB2 segment. We confirmed that this virus replicated only in PB2-expressing cells and expressed HN. We then examined the efficacy of this virus as a bivalent vaccine in a hamster model. High levels of virus-specific IgG antibodies in sera and IgA, IgG, and IgM antibodies in bronchoalveolar lavage fluids against both influenza virus and HPIV3 were detected from hamsters immunized with this virus. The neutralizing capability of these serum antibodies was also confirmed. Moreover, the immunized hamsters were completely protected from virus challenge with influenza virus or HPIV3. These results indicate that PB2-KO virus expressing the HN of HPIV3 has the potential to be a novel bivalent vaccine against influenza and human parainfluenza viruses.


Assuntos
Proteína HN/imunologia , Vacinas contra Influenza/imunologia , Orthomyxoviridae/imunologia , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Animais , Anticorpos Neutralizantes/análise , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Líquido da Lavagem Broncoalveolar/imunologia , Cricetinae , Feminino , Proteína HN/genética , Imunoglobulina A/análise , Imunoglobulina G/análise , Imunoglobulina G/sangue , Imunoglobulina M/análise , Imunoglobulina M/sangue , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Mesocricetus , Orthomyxoviridae/genética , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/genética , Soro/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
8.
Vaccine ; 31(48): 5706-12, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24103895

RESUMO

BACKGROUND: Human parainfluenza virus type 3 (HPIV3) is a common cause of upper and lower respiratory tract illness in infants and young children. Live-attenuated cold-adapted HPIV3 vaccines have been evaluated in infants but a suitable interval for administration of a second dose of vaccine has not been defined. METHODS: HPIV3-seronegative children between the ages of 6 and 36 months were randomized 2:1 in a blinded study to receive two doses of 105 TCID50 (50% tissue culture infectious dose) of live-attenuated, recombinant cold-passaged human PIV3 vaccine (rHPIV3cp45) or placebo 6 months apart. Serum antibody levels were assessed prior to and approximately 4-6 weeks after each dose. Vaccine virus infectivity, defined as detection of vaccine-HPIV3 in nasal wash and/or a≥4-fold rise in serum antibody titer, and reactogenicity were assessed on days 3, 7, and 14 following immunization. RESULTS: Forty HPIV3-seronegative children (median age 13 months; range 6-35 months) were enrolled; 27 (68%) received vaccine and 13 (32%) received placebo. Infectivity was detected in 25 (96%) of 26 evaluable vaccinees following doses 1 and 9 of 26 subject (35%) following dose 2. Among those who shed virus, the median duration of viral shedding was 12 days (range 6-15 days) after dose 1 and 6 days (range 3-8 days) after dose 2, with a mean peak log10 viral titer of 3.4 PFU/mL (SD: 1.0) after dose 1 compared to 1.5 PFU/mL (SD: 0.92) after dose 2. Overall, reactogenicity was mild, with no difference in rates of fever and upper respiratory infection symptoms between vaccine and placebo groups. CONCLUSION: rHPIV3cp45 was immunogenic and well-tolerated in seronegative young children. A second dose administered 6 months after the initial dose was restricted in those previously infected with vaccine virus; however, the second dose boosted antibody responses and induced antibody responses in two previously uninfected children.


Assuntos
Vacinas contra Parainfluenza/efeitos adversos , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Vacinação/efeitos adversos , Vacinação/métodos , Anticorpos Antivirais/sangue , Pré-Escolar , Método Duplo-Cego , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Lactente , Masculino , Cavidade Nasal/virologia , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/genética , Placebos/administração & dosagem , Infecções por Respirovirus/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
9.
Voen Med Zh ; 334(4): 27-31, 2013 Apr.
Artigo em Russo | MEDLINE | ID: mdl-24000610

RESUMO

Research is performed for investigation of cultivation peculiarities of virus parainfluenza-3 on different cell cultures and optimization of methods of virus infection of these cultures with PI-3 virus in the process of development of vaccines. Influence of a seed lot on the formation rate of a cellular monolayer of continuous cell cultures PT-80, LEK, KSTand T-1 is defined, it is established that the seed lot of 250,000 cells in 1 ml provides optimum concentration for formation of a full monolayer within the time limits of 48-72 hours. The most sensitive cell cultures (PT-80) for cultivation of parainfluenza-3 virus are selected; an optimal CCID of virus PI-3(0,1 TCD/ml), whereby we can observe the maximal accumulation of a virus within 2-4 day after infecting cell cultures, is defined.


Assuntos
Vacinas contra Parainfluenza , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Infecções por Respirovirus/metabolismo , Animais , Bovinos , Linhagem Celular , Humanos , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/prevenção & controle
10.
J Dairy Sci ; 96(9): 5826-35, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23810586

RESUMO

Typical fatty acid profiles of milk and milk replacer (MR) differ. Calf MR in the United States are made from animal fat, which are low in short- and medium-chain fatty acids and linolenic acid. Two 56-d trials compared a control MR containing 27% crude protein and formulated with 3 fat and fatty acid compositions. The 3 MR treatments were (1) only animal fat totaling 17% fat (CON), (2) animal fat supplemented with butyrate, medium-chain fatty acids, and linolenic acid using a commercial product (1.25% NeoTec4 MR; Provimi North America, Brookville, OH) totaling 17% fat (fatty acid-supplemented; FA-S), and (3) milk fat totaling 33% fat (MF). The MR were fed at 660 g of dry matter from d 0 to 42 and weaned. Starter (20% crude protein) and water were fed ad libitum for 56 d. Trial 1 utilized Holstein calves (24 female, 24 male) during summer months and trial 2 utilized Holstein calves (48 male) during fall months. Calves (41±1 kg of initial body weight; 2 to 3d of age) were sourced from a single farm and housed in a naturally ventilated nursery without added heat. Calves were in individual pens with straw bedding. Calf was the experimental unit. Data for each trial were analyzed as a completely randomized design with a 3 (MR treatment) × 2 (sex) factorial arrangement of treatments in trial 1 with repeated measures and as a completely randomized design with 3 MR treatments in trial 2 with repeated measures. Preplanned contrast statements of treatments CON versus FA-S and CON versus MF were used to separate means. We found no interactions of MR treatment by sex. Calf average daily gain, hip width change, and feed efficiency differed (CONFA-S). Titers to bovine respiratory parainfluenza-3 and bovine virus diarrhea type 1 (vaccinations to these pathogens were on d 7 and 28) in serum samples taken on d 49 and 56 differed (CONFA-S; CONFA-S; CON>MF). Calves fed FA-S and MF had improved growth and feed efficiency compared with calves fed CON, whereas calves fed FA-S also had improved measurements related to health and immunity.


Assuntos
Bovinos/crescimento & desenvolvimento , Ácidos Graxos/farmacologia , Ração Animal , Fenômenos Fisiológicos da Nutrição Animal/efeitos dos fármacos , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/imunologia , Butiratos/farmacologia , Bovinos/imunologia , Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Dieta/veterinária , Suplementos Nutricionais , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunidade Humoral/fisiologia , Masculino , Vacinas contra Parainfluenza/imunologia , Vacinas contra Parainfluenza/farmacologia , Vírus da Parainfluenza 3 Bovina/imunologia , Vacinas Virais/imunologia , Vacinas Virais/farmacologia , Ácido alfa-Linolênico/farmacologia
12.
Vaccine ; 30(26): 3975-81, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22178099

RESUMO

Human parainfluenza virus type 3 (HPIV3) is an important cause of lower respiratory tract illness in children, yet a licensed vaccine or antiviral drug is not available. We evaluated the safety, tolerability, infectivity, and immunogenicity of two intranasal, live-attenuated HPIV3 vaccines, designated rHPIV3-N(B) and rB/HPIV3, that were cDNA-derived chimeras of HPIV3 and bovine PIV3 (BPIV3). These were evaluated in adults, HPIV3 seropositive children, and HPIV3 seronegative children. A total of 112 subjects participated in these studies. Both rB/HPIV3 and rHPIV3-N(B) were highly restricted in replication in adults and seropositive children but readily infected seronegative children, who shed mean peak virus titers of 10(2.8) vs. 10(3.7)pfu/mL, respectively. Although rB/HPIV3 was more restricted in replication in seronegative children than rHPIV3-N(B), it induced significantly higher titers of hemagglutination inhibition (HAI) antibodies against HPIV3. Taken together, these data suggest that the rB/HPIV3 vaccine is the preferred candidate for further clinical development.


Assuntos
Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vacinação/métodos , Administração Intranasal , Adulto , Anticorpos Antivirais/sangue , Pré-Escolar , Testes de Inibição da Hemaglutinação , Humanos , Lactente , Vacinas contra Parainfluenza/efeitos adversos , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/genética , Vacinação/efeitos adversos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Replicação Viral , Eliminação de Partículas Virais
13.
Expert Rev Respir Med ; 5(4): 515-26, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21859271

RESUMO

In children under 5 years of age, human parainfluenza viruses (HPIVs) as a group are the second most common etiology of acute respiratory illness leading to hospitalization, surpassed only by respiratory syncytial virus but ahead of influenza viruses. Using reverse genetics systems for HPIV serotypes 1, 2 and 3 (HPIV1, 2 and 3), several live-attenuated HPIVs have been generated and evaluated as intranasal vaccines in adults and in children. Two vaccines against HPIV3 were found to be well tolerated, infectious and immunogenic in Phase I trials in HPIV3-seronegative infants and children and should progress to proof-of-concept trials. Vaccines against HPIV1 and HPIV2 are less advanced and have just entered pediatric trials.


Assuntos
Desenho de Fármacos , Vacinas contra Parainfluenza/administração & dosagem , Infecções por Respirovirus/prevenção & controle , Respirovirus/imunologia , Doença Aguda , Administração Intranasal , Aerossóis , Pré-Escolar , Humanos , Lactente , Vacinas contra Parainfluenza/química , Vacinas contra Parainfluenza/genética , Respirovirus/genética , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/imunologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem
14.
Pediatr Infect Dis J ; 30(10): e186-91, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21829138

RESUMO

BACKGROUND: Human parainfluenza virus type 3 (HPIV3) is an important yet underappreciated cause of lower respiratory tract illness in children, and a licensed vaccine is not yet available. METHODS: A live-attenuated investigational HPIV3 vaccine virus designated rcp45 was derived from cDNA by using reverse genetics. rcp45 is genetically similar to the biologically derived cp45 vaccine virus and contains all of the known attenuating mutations of cp45, but has the advantage of a short, well-characterized passage history. We evaluated the tolerability, infectivity, and immunogenicity of 2 intranasal doses of rcp45 administered 4 to 10 weeks apart in a placebo-controlled, double-blind trial. A total of 45 infants and children between 6 and 36 months of age participated in this study. Tolerability and antibody responses to vaccine or placebo were assessed in all recipients. Infectivity was assessed by quantitation of vaccine virus shedding in a subset of vaccinated children. RESULTS: rcp45 was well tolerated and highly infectious in HPIV3-seronegative children. A second dose of vaccine administered 4 to 10 weeks after the first dose was restricted in replication and did not boost serum antibody responses. The stability of 9 cp45 mutations, including the 6 major attenuating mutations, was examined and confirmed for viral isolates from 10 children. CONCLUSIONS: The level of attenuation and immunogenicity of cDNA-derived rcp45 is comparable to what was previously observed with the biologically derived cp45 vaccine, and preliminary data suggest that the attenuating mutations in this vaccine virus are genetically stable. Continued clinical development of rcp45 is warranted.


Assuntos
Vacinas contra Parainfluenza/efeitos adversos , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Administração Intranasal , Anticorpos Antivirais/sangue , Pré-Escolar , DNA Complementar/genética , DNA Viral/genética , Método Duplo-Cego , Humanos , Lactente , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/genética , Placebos/administração & dosagem , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Eliminação de Partículas Virais
15.
Vaccine ; 29(40): 7042-8, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21782874

RESUMO

OBJECTIVE: To evaluate the safety, tolerability, immunogenicity, and viral shedding profiles of a recombinant, live, attenuated human parainfluenza virus type 3 (HPIV3) vaccine, rHPIV3cp45, in healthy HPIV3-seronegative infants 6 to <12 months of age. METHODS: In this double-blind, multicenter study, subjects were randomized 2:1 to receive a 10(5)TCID(50) dose of rHPIV3cp45 (n=20) or placebo (n=10) at enrollment and at 2 and 4 months after the first dose. Blood for evaluation of antibody to HPIV3 was collected at baseline and approximately 1 month after each dose. Solicited adverse events (SEs) and unsolicited adverse events (AEs) were collected on days 0-28 after each dose. Nasal wash samples for vaccine virus shedding were collected 3 times after each dose (7-10, 12-18, and 28-34 days post dose) and at unscheduled illness visits. Subjects were followed for 180 days after the last dose. RESULTS: Vaccine virus was shed by 85% of vaccine recipients after dose 1, by 1 subject after dose 2, and was not shed by any subject after dose 3. The highest titer of shed virus was detected on day 7 after dose 1. The attenuation phenotype and the genotype of the vaccine virus were stable in shed virus. Seroresponse (≥ 4-fold rise in HPIV3 antibody from baseline) occurred in 61% of subjects after dose 1 and in 77% after dose 3. Either seroresponse or shedding occurred in 95% of vaccine subjects. Adverse events were similar in vaccine and placebo recipients. CONCLUSION: The safety, shedding, and immunogenicity profiles of rHPIV3cp45 in HPIV3-seronegative infants 6 to <12 months of age support further development of this vaccine.


Assuntos
Tolerância Imunológica/imunologia , Vacinas contra Parainfluenza/administração & dosagem , Vírus da Parainfluenza 3 Humana/imunologia , Anticorpos Antivirais/imunologia , Método Duplo-Cego , Feminino , Genótipo , Humanos , Lactente , Masculino , Vacinas contra Parainfluenza/efeitos adversos , Vacinas contra Parainfluenza/imunologia , Fenótipo , Placebos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Eliminação de Partículas Virais/imunologia
16.
Rev Argent Microbiol ; 42(3): 189-92, 2010.
Artigo em Espanhol | MEDLINE | ID: mdl-21186672

RESUMO

Distemper virus causes a disease affecting minks with respiratory, gastrointestinal, neurological and skin symptoms and showing high morbidity and mortality, mainly among puppies. It is controlled through immunization, using vaccines that are supplied for mink use. The aim of this work was to determine the seroneutralization titer against the distemper virus at a mink farm in Argentina. The antibody kinetics obtained after vaccination in 27 adult animals, as well as the duration of colostrum-transferred antibodies in 10 puppies were determined. All vaccinated adult minks showed protective titers up to at least 3 months after vaccination, and 37.5% significantly reduced their antibody levels, 12 months after vaccination. Only 20% of the puppies showed protective levels of colostrum-transferred antibodies at the age of 7 weeks, while non-detectable levels of antibodies were found when puppies reached 11 weeks old. Vaccination performed in these puppies at the age of 13 weeks, elicited protective seroneutralization titers. These results show that vaccination induces a satisfactory humoral immune response in our environment, and support the convenience of vaccinating dams annually before the beginning of the breeding season. The vaccination plan in puppies is also discussed.


Assuntos
Cinomose/prevenção & controle , Vison/imunologia , Morbillivirus/imunologia , Vacinas contra Parainfluenza/imunologia , Animais , Argentina
17.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19857454

RESUMO

A novel recombinant human parainfluenza virus type 1 (rHPIV1), rHPIV1-C+P, in which the overlapping open reading frames of the C and P genes were separated in order to introduce mutations into the C gene without affecting P, was generated. Infectious rHPIV1-C+P was readily recovered and replicated as efficiently as HPIV1 wild type (wt) in vitro and in African green monkeys (AGMs). rHPIV1-C+P expressed increased levels of C protein and, surprisingly, activated the type I IFN and apoptosis responses more strongly than HPIV1 wt. rHPIV1-C+P provided a useful backbone for recovering an attenuated P/C gene mutation (Delta 84-85), which was previously unrecoverable, likely due to detrimental effects of the deletion on the P protein. rHPIV1-C(Delta 84-85)+P and an additional mutant, rHPIV1-C(Delta 169-170)+P, were found to replicate to similar titers in vitro and to activate the type I IFN and apoptosis responses to a similar degree as rHPIV1-C+P. rHPIV1-C(Delta 84-85)+P was found to be highly attenuated in AGMs, and all viruses were immunogenic and effective in protecting AGMs against challenge with HPIV1 wt. rHPIV1-C(Delta 84-85)+P will be investigated as a potential live-attenuated vaccine candidate for HPIV1.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Fosfoproteínas/genética , Deleção de Sequência , Proteínas Virais/genética , Animais , Anticorpos Antivirais/sangue , Apoptose , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Humanos , Interferon Tipo I/biossíntese , Dados de Sequência Molecular , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Carga Viral , Virulência , Replicação Viral
18.
Pediatr Infect Dis J ; 28(7): 655-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19483659

RESUMO

A live, attenuated respiratory syncytial virus and parainfluenza virus type 3 vaccine was evaluated in healthy respiratory syncytial virus/parainfluenza virus type 3 seropositive children aged 1 to 9 years. Three cohorts of 40 children were randomized 1:1 to receive 10, 10, or 10 median tissue culture infectious dose50 MEDI-534 vaccine or placebo. The vaccine's safety profile was similar to placebo, no viral shedding was detected, and the vaccine was minimally immunogenic.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Administração Intranasal , Criança , Pré-Escolar , Fezes/virologia , Feminino , Experimentação Humana , Humanos , Lactente , Masculino , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/efeitos adversos , Placebos/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Eliminação de Partículas Virais
19.
Vaccine ; 27(12): 1848-57, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19200447

RESUMO

The human parainfluenza viruses (hPIVs) and respiratory syncytial viruses (RSVs) are the leading causes of hospitalizations due to respiratory viral disease in infants and young children, but no vaccines are yet available. Here we describe the use of recombinant Sendai viruses (rSeVs) as candidate vaccine vectors for these respiratory viruses in a cotton rat model. Two new Sendai virus (SeV)-based hPIV-2 vaccine constructs were generated by inserting the fusion (F) gene or the hemagglutinin-neuraminidase (HN) gene from hPIV-2 into the rSeV genome. The inoculation of either vaccine into cotton rats elicited neutralizing antibodies toward both homologous and heterologous hPIV-2 virus isolates. The vaccines elicited robust and durable antibodies toward hPIV-2, and cotton rats immunized with individual or mixed vaccines were fully protected against hPIV-2 infections of the lower respiratory tract. The immune responses toward a single inoculation with rSeV vaccines were long-lasting and cotton rats were protected against viral challenge for as long as 11 months after vaccination. One inoculation with a mixture of the hPIV-2-HN-expressing construct and two additional rSeVs (expressing the F protein of RSV and the HN protein of hPIV-3) resulted in protection against challenge viruses hPIV-1, hPIV-2, hPIV-3, and RSV. Results identify SeV vectors as promising vaccine candidates for four different paramyxoviruses, each responsible for serious respiratory infections in children.


Assuntos
Vacinas contra Parainfluenza/uso terapêutico , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sendai/genética , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Vetores Genéticos , Humanos , Testes de Neutralização , Sigmodontinae , Vacinas Sintéticas/imunologia
20.
Pediatr Infect Dis J ; 27(10 Suppl): S123-5, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18820572

RESUMO

Parainfluenza viruses (PIV) have been generally disregarded as pathogens in spite of their importance in pediatric lower respiratory illness. Because PIVs account for 17% of hospitalized illness associated virus isolation, the development of PIV vaccine would be a major advance in preventing lower respiratory tract infection in infants and young children. We will review in detail several PIV vaccine candidates and recent newer approaches to PIV vaccine development. Intranasally administered bovine PIV3 (bPIV3) vaccine and cold-adapted PIV3 vaccine have been evaluated throughout the pediatric age spectrum. BPIV3 does not give a robust response to the heterotypic human strain although seroconversion rate to bPIV3 is 57-65%. However, bPIV3 vaccine is being used as an attenuated backbone for insertion of human PIV3 hemagglutinin-neuraminidase and fusion (F) proteins and a surface protein, F, of respiratory syncytial virus. The effectiveness of this vaccine against both PIV3 and RSV challenge has been demonstrated in African green monkeys. The cold-adapted PIV3 vaccine has been extensively evaluated and is safe and immunogenic in seronegative children with a seroconversion rate of 79%. These promising candidates deserve to enter into efficacy trials both for their ability to prevent PIV3 disease and as a model of protection against respiratory illness by mucosal vaccination.


Assuntos
Vacinas contra Parainfluenza , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Adulto , Criança , Temperatura Baixa , Crupe/prevenção & controle , Humanos , Lactente , Mutação , Vacinas contra Parainfluenza/genética , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 3 Bovina/genética , Vírus da Parainfluenza 3 Bovina/imunologia , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 4 Humana/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/virologia , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/prevenção & controle , Infecções por Rubulavirus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...