Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(25): e2316376121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38861603

RESUMEN

Human parainfluenza virus type 3 (HPIV3) is a major pediatric respiratory pathogen lacking available vaccines or antiviral drugs. We generated live-attenuated HPIV3 vaccine candidates by codon-pair deoptimization (CPD). HPIV3 open reading frames (ORFs) encoding the nucleoprotein (N), phosphoprotein (P), matrix (M), fusion (F), hemagglutinin-neuraminidase (HN), and polymerase (L) were modified singly or in combination to generate 12 viruses designated Min-N, Min-P, Min-M, Min-FHN, Min-L, Min-NP, Min-NPM, Min-NPL, Min-PM, Min-PFHN, Min-MFHN, and Min-PMFHN. CPD of N or L severely reduced growth in vitro and was not further evaluated. CPD of P or M was associated with increased and decreased interferon (IFN) response in vitro, respectively, but had little effect on virus replication. In Vero cells, CPD of F and HN delayed virus replication, but final titers were comparable to wild-type (wt) HPIV3. In human lung epithelial A549 cells, CPD F and HN induced a stronger IFN response, viral titers were reduced 100-fold, and the expression of F and HN proteins was significantly reduced without affecting N or P or the relative packaging of proteins into virions. Following intranasal infection in hamsters, replication in the nasal turbinates and lungs tended to be the most reduced for viruses bearing CPD F and HN, with maximum reductions of approximately 10-fold. Despite decreased in vivo replication (and lower expression of CPD F and HN in vitro), all viruses induced titers of serum HPIV3-neutralizing antibodies similar to wt and provided complete protection against HPIV3 challenge. In summary, CPD of HPIV3 yielded promising vaccine candidates suitable for further development.


Asunto(s)
Codón , Virus de la Parainfluenza 3 Humana , Vacunas Atenuadas , Replicación Viral , Animales , Virus de la Parainfluenza 3 Humana/inmunología , Virus de la Parainfluenza 3 Humana/genética , Humanos , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/genética , Codón/genética , Cricetinae , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/prevención & control , Infecciones por Respirovirus/virología , Chlorocebus aethiops , Células Vero , Sistemas de Lectura Abierta/genética , Mesocricetus , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Vacunas Virales/inmunología , Vacunas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/genética , Vacunas contra la Parainfluenza/inmunología , Vacunas contra la Parainfluenza/genética
2.
Pediatrics ; 153(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38738290

RESUMEN

OBJECTIVES: Human metapneumovirus (hMPV) and parainfluenza virus type 3 (PIV3) are common respiratory illnesses in children. The safety and immunogenicity of an investigational mRNA-based vaccine, mRNA-1653, encoding membrane-anchored fusion proteins of hMPV and PIV3, was evaluated in hMPV/PIV3-seropositive children. METHODS: In this phase 1b randomized, observer-blind, placebo-controlled, dose-ranging study, hMPV/PIV3-seropositive children were enrolled sequentially into 2 dose levels of mRNA-1653 administered 2 months apart; children aged 12 to 36 months were randomized (1:1) to receive 10-µg of mRNA-1653 or placebo and children aged 12 to 59 months were randomized (3:1) to receive 30-µg of mRNA-1653 or placebo. RESULTS: Overall, 27 participants aged 18 to 55 months were randomized; 15 participants received 10-µg of mRNA-1653 (n = 8) or placebo (n = 7), whereas 12 participants received 30-µg of mRNA-1653 (n = 9) or placebo (n = 3). mRNA-1653 was well-tolerated at both dose levels. The only reported solicited local adverse reaction was tenderness at injection site; solicited systemic adverse reactions included grade 1 or 2 chills, irritability, loss of appetite, and sleepiness. A single 10-µg or 30-µg mRNA-1653 injection increased hMPV and PIV3 neutralizing antibody titers (geometric mean fold-rise ratio over baseline: hMPV-A = 2.9-6.1; hMPV-B = 6.2-13.2; PIV3 = 2.8-3.0) and preF and postF binding antibody concentrations (geometric mean fold-rise ratio: hMPV preF = 5.3-6.1; postF = 4.6-6.5 and PIV3 preF = 13.9-14.2; postF = 11.0-12.1); a second injection did not further increase antibody levels in these seropositive children. Binding antibody responses were generally preF biased. CONCLUSIONS: mRNA-1653 was well-tolerated and boosted hMPV and PIV3 antibody levels in seropositive children aged 12 to 59 months, supporting the continued development of mRNA-1653 or its components for the prevention of hMPV and PIV3.


Asunto(s)
Virus de la Parainfluenza 3 Humana , Humanos , Femenino , Masculino , Preescolar , Lactante , Virus de la Parainfluenza 3 Humana/inmunología , Virus de la Parainfluenza 3 Humana/genética , Metapneumovirus/inmunología , Metapneumovirus/genética , Método Simple Ciego , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/inmunología , Anticuerpos Antivirales/sangre , Vacunas contra la Parainfluenza/inmunología , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Inmunogenicidad Vacunal , ARN Mensajero
3.
PLoS One ; 15(2): e0228572, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32045432

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 3 Humana/inmunología , Vacunas contra Virus Sincitial Respiratorio/genética , Proteínas Virales de Fusión/genética , Ensamble de Virus , Administración Intranasal , Animales , Chlorocebus aethiops , Cricetinae , Femenino , Humanos , Inmunogenicidad Vacunal , Macaca mulatta , Mesocricetus , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 3 Humana/fisiología , Estabilidad Proteica , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Células Vero , Proteínas Virales de Fusión/metabolismo
4.
Antiviral Res ; 162: 54-60, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30550799

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Antivirales/sangre , Inmunidad Materno-Adquirida , Vacunas contra la Parainfluenza/administración & dosificación , Infecciones por Respirovirus/veterinaria , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/sangre , Femenino , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Embarazo , Virus Sincitiales Respiratorios/genética , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/prevención & control , Ovinos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/inmunología
5.
J Vet Intern Med ; 30(1): 164-6, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26692461

RESUMEN

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.


Asunto(s)
Adenoviridae , Vacunas Bacterianas/inmunología , Bordetella bronchiseptica , Enfermedades de los Perros/prevención & control , Vacunas contra la Parainfluenza/inmunología , Vacunas Virales/inmunología , Infecciones por Adenoviridae/prevención & control , Infecciones por Adenoviridae/veterinaria , Administración Tópica , Animales , Infecciones por Bordetella/prevención & control , ADN Bacteriano/genética , Virus del Moquillo Canino/genética , Enfermedades de los Perros/microbiología , Enfermedades de los Perros/virología , Perros , Parvovirus/genética , Proyectos Piloto , Reacción en Cadena de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa/veterinaria , ARN Viral/genética , Vacunación , Vacunas Atenuadas
6.
Vaccine ; 31(48): 5706-12, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24103895

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/efectos adversos , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Respirovirus/prevención & control , Vacunación/efectos adversos , Vacunación/métodos , Anticuerpos Antivirales/sangre , Preescolar , Método Doble Ciego , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Lactante , Masculino , Cavidad Nasal/virología , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 3 Humana/genética , Placebos/administración & dosificación , Infecciones por Respirovirus/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
7.
Vaccine ; 31(52): 6239-46, 2013 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24144478

RESUMEN

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.


Asunto(s)
Proteína HN/inmunología , Vacunas contra la Influenza/inmunología , Orthomyxoviridae/inmunología , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Animales , Anticuerpos Neutralizantes/análisis , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Líquido del Lavado Bronquioalveolar/inmunología , Cricetinae , Femenino , Proteína HN/genética , Inmunoglobulina A/análisis , Inmunoglobulina G/análisis , Inmunoglobulina G/sangre , Inmunoglobulina M/análisis , Inmunoglobulina M/sangre , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Mesocricetus , Orthomyxoviridae/genética , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 3 Humana/genética , Suero/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
8.
J Dairy Sci ; 96(9): 5826-35, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23810586

RESUMEN

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.


Asunto(s)
Bovinos/crecimiento & desarrollo , Ácidos Grasos/farmacología , Alimentación Animal , Fenómenos Fisiológicos Nutricionales de los Animales/efectos de los fármacos , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/inmunología , Butiratos/farmacología , Bovinos/inmunología , Virus de la Diarrea Viral Bovina Tipo 1/inmunología , Dieta/veterinaria , Suplementos Dietéticos , Femenino , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/fisiología , Masculino , Vacunas contra la Parainfluenza/inmunología , Vacunas contra la Parainfluenza/farmacología , Virus de la Parainfluenza 3 Bovina/inmunología , Vacunas Virales/inmunología , Vacunas Virales/farmacología , Ácido alfa-Linolénico/farmacología
9.
Vaccine ; 30(26): 3975-81, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22178099

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Vacunación/métodos , Administración Intranasal , Adulto , Anticuerpos Antivirales/sangre , Preescolar , Pruebas de Inhibición de Hemaglutinación , Humanos , Lactante , Vacunas contra la Parainfluenza/efectos adversos , Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 3 Humana/genética , Vacunación/efectos adversos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Replicación Viral , Esparcimiento de Virus
10.
Pediatr Infect Dis J ; 30(10): e186-91, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21829138

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/efectos adversos , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Administración Intranasal , Anticuerpos Antivirales/sangre , Preescolar , ADN Complementario/genética , ADN Viral/genética , Método Doble Ciego , Humanos , Lactante , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 3 Humana/genética , Placebos/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Esparcimiento de Virus
11.
Vaccine ; 29(40): 7042-8, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21782874

RESUMEN

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.


Asunto(s)
Tolerancia Inmunológica/inmunología , Vacunas contra la Parainfluenza/administración & dosificación , Virus de la Parainfluenza 3 Humana/inmunología , Anticuerpos Antivirales/inmunología , Método Doble Ciego , Femenino , Genotipo , Humanos , Lactante , Masculino , Vacunas contra la Parainfluenza/efectos adversos , Vacunas contra la Parainfluenza/inmunología , Fenotipo , Placebos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Esparcimiento de Virus/inmunología
12.
Rev Argent Microbiol ; 42(3): 189-92, 2010.
Artículo en Español | MEDLINE | ID: mdl-21186672

RESUMEN

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.


Asunto(s)
Moquillo/prevención & control , Visón/inmunología , Morbillivirus/inmunología , Vacunas contra la Parainfluenza/inmunología , Animales , Argentina
13.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19857454

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 1 Humana/inmunología , Virus de la Parainfluenza 1 Humana/patogenicidad , Fosfoproteínas/genética , Eliminación de Secuencia , Proteínas Virales/genética , Animales , Anticuerpos Antivirales/sangre , Apoptosis , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Humanos , Interferón Tipo I/biosíntesis , Datos de Secuencia Molecular , Vacunas contra la Parainfluenza/genética , Virus de la Parainfluenza 1 Humana/genética , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Carga Viral , Virulencia , Replicación Viral
14.
Pediatr Infect Dis J ; 28(7): 655-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19483659

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Administración Intranasal , Niño , Preescolar , Heces/virología , Femenino , Experimentación Humana , Humanos , Lactante , Masculino , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/efectos adversos , Placebos/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Esparcimiento de Virus
15.
Pediatr Infect Dis J ; 27(10 Suppl): S123-5, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18820572

RESUMEN

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.


Asunto(s)
Vacunas contra la Parainfluenza , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Respirovirus/prevención & control , Adulto , Niño , Frío , Crup/prevención & control , Humanos , Lactante , Mutación , Vacunas contra la Parainfluenza/genética , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 1 Humana/inmunología , Virus de la Parainfluenza 2 Humana/inmunología , Virus de la Parainfluenza 3 Bovina/genética , Virus de la Parainfluenza 3 Bovina/inmunología , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 4 Humana/inmunología , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Infecciones por Rubulavirus/inmunología , Infecciones por Rubulavirus/prevención & control , Infecciones por Rubulavirus/virología
16.
Pediatr Infect Dis J ; 27(10 Suppl): S126-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18820573

RESUMEN

Respiratory syncytial virus (RSV) and human parainfluenza viruses (hPIVs) are the most important causes of hospitalization for viral respiratory tract diseases in infants and young children. Unfortunately, there are currently no licensed vaccines for prevention of these infections. Researchers at St. Jude Children's Research Hospital are now developing Sendai virus (SV), a natural respiratory pathogen of mice, as a Jennerian vaccine for hPIV-1, and as a vaccine backbone for the prevention of RSV and other hPIVs. Unmodified SV is currently being tested in the clinic. Thus far, the vaccine has been well tolerated. Preclinical studies also continue and have demonstrated that intranasal vaccinations with recombinant SV expressing an RSV antigen are sufficient to activate high-magnitude RSV-specific neutralizing B- and T-cell activities in a cotton rat system. Furthermore, vaccinated animals are completely protected against RSV challenges. As clinical safety studies progress, St. Jude Children's Research Hospital researchers are also working to formulate a SV-based cocktail vaccine designed to prevent several hPIV and RSV infections in humans.


Asunto(s)
Vacunas contra la Parainfluenza , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio , Virus Sendai/inmunología , Vacunas Virales , Adulto , Animales , Humanos , Inmunidad Innata , Lactante , Vacunas contra la Parainfluenza/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sendai/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
17.
Vaccine ; 26(50): 6373-82, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-18822334

RESUMEN

MEDI-534 is a bivalent live attenuated vaccine candidate against human respiratory syncytial virus (hRSV) and human parainfluenza virus type 3 (hPIV3) that was previously shown to be immunogenic and to protect rodents and African green monkeys from wild-type (wt) hRSV challenge. We performed further preclinical evaluations to address the safety of MEDI-534 prior to human testing. MEDI-534 did not predispose rodents to enhanced RSV disease following wt-RSV challenge, and the tissue tropism of the chimeric virus was confined to the respiratory tract. Representative clinical trial material did not produce toxicity in rats. In adults, MEDI-534 was highly restricted in replication, did not boost RSV and PIV3 antibody titers, and produced no medically significant vaccine-related adverse events thereby warranting further evaluation in pediatric populations.


Asunto(s)
Vacunas contra la Parainfluenza , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio , Infecciones por Respirovirus/prevención & control , Vacunas Atenuadas , Adolescente , Adulto , Animales , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Vectores Genéticos , Humanos , Masculino , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/genética , Ratas , Ratas Sprague-Dawley , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Sigmodontinae , Resultado del Tratamiento , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Células Vero , Adulto Joven
18.
Vaccine ; 26(27-28): 3480-8, 2008 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-18499307

RESUMEN

The human parainfluenza viruses (hPIVs) and respiratory syncytial virus (RSV) are the leading causes of serious respiratory illness in the human pediatric population. Despite decades of research, there are currently no licensed vaccines for either the hPIV or RSV pathogens. Here we describe the testing of hPIV-3 and RSV candidate vaccines using Sendai virus (SeV, murine PIV-1) as a vector. SeV was selected as the vaccine backbone, because it has been shown to elicit robust and durable immune activities in animal studies, and has already advanced to human safety trials as a xenogenic vaccine for hPIV-1. Two new SeV-based hPIV-3 vaccine candidates were first generated by inserting either the fusion (F) gene or hemagglutinin-neuraminidase (HN) gene from hPIV-3 into SeV. The resultant rSeV-hPIV3-F and rSeV-hPIV3-HN vaccines expressed their inserted hPIV-3 genes upon infection. The inoculation of either vaccine into cotton rats elicited binding and neutralizing antibody activities, as well as interferon-gamma-producing T cells. Vaccination of cotton rats resulted in protection against subsequent challenges with either homologous or heterologous hPIV-3. Furthermore, vaccination of cotton rats with a mixture of rSeV-hPIV3-HN and a previously described recombinant SeV expressing the F protein of RSV resulted in protection against three different challenge viruses: hPIV-3, hPIV-1 and RSV. Results encourage the continued development of the candidate recombinant SeV vaccines to combat serious respiratory infections of children.


Asunto(s)
Proteína HN/inmunología , Vacunas contra la Parainfluenza/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Infecciones por Respirovirus/prevención & control , Virus Sendai/genética , Proteínas Virales de Fusión/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Femenino , Vectores Genéticos , Proteína HN/genética , Pulmón/virología , Datos de Secuencia Molecular , Vacunas contra la Parainfluenza/genética , Ratas , Vacunas contra Virus Sincitial Respiratorio/genética , Sigmodontinae , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética
19.
Scand J Immunol ; 66(6): 645-53, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17944814

RESUMEN

No licensed vaccines are available to protect against parainfluenza virus type 3 (PIV3), a significant health risk for infants. In search of a safe vaccine, we used an alphavirus-based chimeric vector, consisting of Sindbis virus (SIN) structural proteins and Venezuelan equine encephalitis virus (VEE) replicon RNA, expressing the PIV3 hemagglutinin-neuraminidase (HN) glycoprotein (VEE/SIN-HN). We compared different routes of intramuscular (i.m.), intranasal (i.n.), or combined i.n. and i.m. immunizations with VEE/SIN-HN in hamsters. Six months after the final immunization, all hamsters were protected against live PIV3 i.n. challenge in nasal turbinates and lungs. This protection appeared to correlate with antibodies in serum, nasal turbinates and lungs. This is the first report demonstrating mucosal protection against PIV3 for an extended time following immunizations with an RNA replicon delivery system.


Asunto(s)
Alphavirus/inmunología , Membrana Mucosa/inmunología , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , ARN Viral/inmunología , Replicón/inmunología , Administración Intranasal , Alphavirus/genética , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Cricetinae , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/inmunología , Humanos , Inmunización , Inyecciones Intramusculares , Virus de la Parainfluenza 3 Humana/crecimiento & desarrollo , ARN Viral/genética , Replicón/genética , Virus Sindbis/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...