Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Pediatrics ; 153(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38738290

ABSTRACT

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.


Subject(s)
Parainfluenza Virus 3, Human , Humans , Female , Male , Child, Preschool , Infant , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/genetics , Metapneumovirus/immunology , Metapneumovirus/genetics , Single-Blind Method , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/immunology , Antibodies, Viral/blood , Parainfluenza Vaccines/immunology , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Immunogenicity, Vaccine , RNA, Messenger
2.
PLoS One ; 15(2): e0228572, 2020.
Article in English | MEDLINE | ID: mdl-32045432

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/immunology , Respiratory Syncytial Virus Vaccines/genetics , Viral Fusion Proteins/genetics , Virus Assembly , Administration, Intranasal , Animals , Chlorocebus aethiops , Cricetinae , Female , Humans , Immunogenicity, Vaccine , Macaca mulatta , Mesocricetus , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , Protein Stability , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Vero Cells , Viral Fusion Proteins/metabolism
3.
Antiviral Res ; 162: 54-60, 2019 02.
Article in English | MEDLINE | ID: mdl-30550799

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/blood , Immunity, Maternally-Acquired , Parainfluenza Vaccines/administration & dosage , Respirovirus Infections/veterinary , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Female , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Pregnancy , Respiratory Syncytial Viruses/genetics , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Sheep , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/immunology
4.
Pediatr Rev ; 35(6): 217-27; quiz 228, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24891595

ABSTRACT

• 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).


Subject(s)
Influenza Vaccines/administration & dosage , Influenza, Human/diagnosis , Influenza, Human/prevention & control , Parainfluenza Vaccines/administration & dosage , Paramyxoviridae Infections/diagnosis , Paramyxoviridae Infections/prevention & control , Antiviral Agents/therapeutic use , Child , Cross-Sectional Studies , Diagnosis, Differential , Humans , Influenza, Human/epidemiology , Paramyxoviridae Infections/epidemiology
5.
Vaccine ; 31(48): 5706-12, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24103895

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines/adverse effects , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Vaccination/adverse effects , Vaccination/methods , Antibodies, Viral/blood , Child, Preschool , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Infant , Male , Nasal Cavity/virology , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/genetics , Placebos/administration & dosage , Respirovirus Infections/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
Vaccine ; 31(52): 6239-46, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24144478

ABSTRACT

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.


Subject(s)
HN Protein/immunology , Influenza Vaccines/immunology , Orthomyxoviridae/immunology , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Animals , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/blood , Antibodies, Viral/analysis , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/immunology , Cricetinae , Female , HN Protein/genetics , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin G/blood , Immunoglobulin M/analysis , Immunoglobulin M/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mesocricetus , Orthomyxoviridae/genetics , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/genetics , Serum/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
7.
Vaccine ; 30(26): 3975-81, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22178099

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Vaccination/methods , Administration, Intranasal , Adult , Antibodies, Viral/blood , Child, Preschool , Hemagglutination Inhibition Tests , Humans , Infant , Parainfluenza Vaccines/adverse effects , Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/genetics , Vaccination/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virus Replication , Virus Shedding
8.
Pediatr Infect Dis J ; 30(10): e186-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21829138

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines/adverse effects , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Administration, Intranasal , Antibodies, Viral/blood , Child, Preschool , DNA, Complementary/genetics , DNA, Viral/genetics , Double-Blind Method , Humans , Infant , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/genetics , Placebos/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virus Shedding
9.
Expert Rev Respir Med ; 5(4): 515-26, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21859271

ABSTRACT

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.


Subject(s)
Drug Design , Parainfluenza Vaccines/administration & dosage , Respirovirus Infections/prevention & control , Respirovirus/immunology , Acute Disease , Administration, Intranasal , Aerosols , Child, Preschool , Humans , Infant , Parainfluenza Vaccines/chemistry , Parainfluenza Vaccines/genetics , Respirovirus/genetics , Respirovirus Infections/epidemiology , Respirovirus Infections/immunology , Treatment Outcome , Vaccines, Attenuated/administration & dosage
10.
Vaccine ; 29(40): 7042-8, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21782874

ABSTRACT

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.


Subject(s)
Immune Tolerance/immunology , Parainfluenza Vaccines/administration & dosage , Parainfluenza Virus 3, Human/immunology , Antibodies, Viral/immunology , Double-Blind Method , Female , Genotype , Humans , Infant , Male , Parainfluenza Vaccines/adverse effects , Parainfluenza Vaccines/immunology , Phenotype , Placebos , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Virus Shedding/immunology
11.
Pediatr Infect Dis J ; 28(7): 655-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19483659

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/prevention & control , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Administration, Intranasal , Child , Child, Preschool , Feces/virology , Female , Human Experimentation , Humans , Infant , Male , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/adverse effects , Placebos/administration & dosage , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Virus Shedding
12.
Vaccine ; 26(50): 6373-82, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18822334

ABSTRACT

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.


Subject(s)
Parainfluenza Vaccines , Parainfluenza Virus 3, Human/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines , Respirovirus Infections/prevention & control , Vaccines, Attenuated , Adolescent , Adult , Animals , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Double-Blind Method , Female , Genetic Vectors , Humans , Male , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/genetics , Rats , Rats, Sprague-Dawley , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sigmodontinae , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Vero Cells , Young Adult
13.
Vet Ther ; 9(4): 257-62, 2008.
Article in English | MEDLINE | ID: mdl-19177330

ABSTRACT

Three groups of healthy dogs with low antibody titers to Bordetella bronchiseptica (Bb), canine parainfluenza virus (CPI), and canine adenovirus type 2 (CAV-2) were used in this study. One group was vaccinated with a single dose of monovalent attenuated Bb vaccine and one group with a trivalent vaccine containing attenuated Bb, CPI, and CAV-2; dogs were vaccinated intranasally with a single dose of the respective vaccines. The third group served as unvaccinated controls. All vaccinated dogs subsequently developed serum antibody titers to Bb that persisted for at least 1 year. Following Bb challenge 1 year after vaccination, all vaccinated dogs, regardless of group, showed significantly fewer clinical signs and shed significantly fewer challenge organisms than unvaccinated controls. These results demonstrate that intranasal administration of a single dose of monovalent attenuated Bb vaccine or trivalent vaccine containing attenuated Bb, CPI, and CAV-2 provides 1 year of protection against Bb.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bordetella Infections/veterinary , Bordetella bronchiseptica/immunology , Dog Diseases/prevention & control , Viral Vaccines/administration & dosage , Adenoviridae Infections/prevention & control , Adenoviridae Infections/veterinary , Adenoviruses, Canine/immunology , Administration, Intranasal , Animals , Animals, Newborn , Antibodies, Viral/blood , Bordetella Infections/prevention & control , Dogs , Female , Male , Parainfluenza Vaccines/administration & dosage , Parainfluenza Virus 2, Human/immunology , Random Allocation , Rubulavirus Infections/prevention & control , Rubulavirus Infections/veterinary , Vaccines, Attenuated
14.
Virol J ; 4: 67, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17605811

ABSTRACT

BACKGROUND: Two recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) mutant viruses have been developed, using a reverse genetics system, for evaluation as potential intranasal vaccine candidates. These rHPIV1 vaccine candidates have two non-temperature sensitive (non-ts) attenuating (att) mutations primarily in the P/C gene, namely CR84GHNT553A (two point mutations used together as a set) and CDelta170 (a short deletion mutation), and two ts att mutations in the L gene, namely LY942A (a point mutation), and LDelta1710-11 (a short deletion), the last of which has not been previously described. The latter three mutations were specifically designed for increased genetic and phenotypic stability. These mutations were evaluated on the HPIV1 backbone, both individually and in combination, for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). RESULTS: The rHPIV1 mutant bearing the novel LDelta1710-11 mutation was highly ts and attenuated in AGMs and was immunogenic and efficacious against HPIV1 wt challenge. The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates were highly ts, with shut-off temperatures of 38 degrees C and 35 degrees C, respectively, and were highly attenuated in AGMs. Immunization with rHPIV1-CR84G/Delta170HNT553ALY942A protected against HPIV1 wt challenge in both the upper and lower respiratory tracts. In contrast, rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 was not protective in AGMs due to over-attenuation, but it is expected to replicate more efficiently and be more immunogenic in the natural human host. CONCLUSION: The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates are clearly highly attenuated in AGMs and clinical trials are planned to address safety and immunogenicity in humans.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, DNA/immunology , Viral Proteins/genetics , Administration, Intranasal , Animals , Attachment Sites, Microbiological/genetics , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mutation , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Phosphoproteins/genetics , Phosphoproteins/immunology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vero Cells , Viral Proteins/immunology , Virus Replication
15.
Vaccine ; 24(14): 2674-84, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16364511

ABSTRACT

The P/C gene of human parainfluenza virus type 1 (HPIV1) encodes a nested set of related accessory C proteins, C'/C/Y1/Y2, which have been shown in other paramyxoviruses to have a role in evasion of the type I interferon (IFN) response following virus infection. We previously demonstrated that a set of two amino acid substitutions, CR84G/HNT553A, and a separate amino acid substitution, CF170S, are independently attenuating for HPIV1 in African green monkeys (AGMs). However, in each case the attenuation (att) phenotype is vulnerable to reversion by a single nucleotide change back to wild type. Using reverse genetics, recombinant HPIV1 (rHPIV1) vaccine candidates were generated that were designed for increased genetic and phenotypic stability by: (i) creating a two-amino acid deletion and substitution at the site of the CF170S mutation, yielding CDelta170; (ii) introducing a six amino acid deletion in the N-terminal region of C, CDelta10-15; and (iii) combining these stable deletion mutations with the att CR84G/HNT553A mutation. The resulting rHPIV1 vaccine candidates were evaluated for attenuation in hamsters and AGMs and for immunogenicity and protective efficacy in AGMs. The CDelta10-15 mutation was attenuating in hamsters but not in AGMs, and likely will be of limited value for an HPIV1 vaccine. Conversely, the CR84G/HNT553A mutation set was attenuating in AGMs but not in hamsters. Thus, these two mutations demonstrated reciprocal host range phenotypes involving different regions of C. The CDelta170 mutation conferred a significant level of attenuation in hamsters and AGMs that closely resembled that of CF170S and will be of particular utility for vaccine development because it involves a deletion of six nucleotides rendering it highly refractory to reversion. The combination of the CR84G/HNT553A mutation set and the CDelta170 deletion mutation yielded a virus, rCR84G/Delta170 HNT553A, that exhibited a satisfactory level of attenuation in hamsters and AGMs and was immunogenic and highly protective against HPIV1 wt challenge. This virus will be evaluated clinically as a live intranasal HPIV1 vaccine, one that can be further attenuated as necessary by the introduction of additional stabilized att mutations previously developed in the L protein.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, Attenuated/immunology , Animals , Cell Line , Cricetinae , Humans , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/pathogenicity , Point Mutation , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics
16.
Vaccine ; 23(38): 4631-46, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-15951066

ABSTRACT

A set of recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) vaccine candidates was evaluated for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). Temperature sensitive (ts) and non-ts attenuating (att) mutations in the P/C and L genes were introduced individually or in various combinations into rHPIV1, including the C(R84G) and HN(T553A) mutations identified in the present work and the C(F170S), L(Y942A), and L(L992C) mutations identified previously. The rHPIV1 vaccine candidates exhibited a spectrum of attenuation in AGMs. One genetically and phenotypically stable vaccine candidate, rC(R84G/F170S)L(Y942A/L992C), was attenuated and efficacious in AGMs and is a promising live attenuated intranasal HPIV1 vaccine candidate suitable for clinical evaluation.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Viral/blood , Disease Models, Animal , Humans , Macaca mulatta , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/pathogenicity , Point Mutation , Respirovirus Infections/prevention & control , Temperature , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Proteins/immunology
17.
Vaccine ; 23(39): 4765-74, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15964103

ABSTRACT

Live-attenuated recombinant human parainfluenza virus type 2 (rHPIV2) vaccine candidates were created using reverse genetics by importing known attenuating mutations in the L polymerase protein from heterologous paramyxoviruses into the homologous sites of the HPIV2 L protein. Four recombinants (rF460L, rY948H, rL1566I, and rS1724I) were recovered and three were attenuated for replication in hamsters. The genetic stability of the imported mutations at three of the four sites was enhanced by use of alternative codons or by deletion of a pair of amino acids. rHPIV2s bearing these modified mutations exhibited enhanced attenuation. The genetically stabilized mutations conferring a high level of attenuation will be useful in generating a live-attenuated virus vaccine for HPIV2.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 2, Human/immunology , Paramyxovirinae/genetics , Vaccines, Synthetic/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Cell Line , Codon , Cricetinae , Humans , Mesocricetus , Molecular Sequence Data , Mutation , Parainfluenza Vaccines/administration & dosage , Parainfluenza Virus 2, Human/physiology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virus Replication
18.
J Virol ; 79(11): 6674-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15890905

ABSTRACT

Influenza and human parainfluenza virus infections are of both medical and economical importance. Currently, inactivated vaccines provide suboptimal protection against influenza, and vaccines for human parainfluenza virus infection are not available, underscoring the need for new vaccines against these respiratory diseases. Furthermore, to reduce the burden of vaccination, the development of multivalent vaccines is highly desirable. Thus, to devise a single vaccine that would elicit immune responses against both influenza and parainfluenza viruses, we used reverse genetics to generate an influenza A virus that possesses the coding region for the hemagglutinin/neuraminidase ectodomain of parainfluenza virus instead of the influenza virus neuraminidase. The recombinant virus grew efficiently in eggs but was attenuated in mice. When intranasally immunized with the recombinant vaccine, all mice developed antibodies against both influenza and parainfluenza viruses and survived an otherwise lethal challenge with either of these viruses. This live bivalent vaccine has obvious advantages over combination vaccines, and its method of generation could, in principle, be applied in the development of a "cocktail" vaccine with efficacy against several different infectious diseases.


Subject(s)
Influenza Vaccines/administration & dosage , Parainfluenza Vaccines/administration & dosage , Animals , Cell Line , Chick Embryo , Dogs , Female , Genetic Engineering , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Humans , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/pathogenicity , Influenza Vaccines/genetics , Influenza Vaccines/isolation & purification , Mice , Mice, Inbred BALB C , Neuraminidase/genetics , Neuraminidase/immunology , Parainfluenza Vaccines/genetics , Parainfluenza Vaccines/isolation & purification , Sendai virus/genetics , Sendai virus/immunology , Sendai virus/pathogenicity , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/isolation & purification , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/isolation & purification , Virulence/genetics , Virulence/immunology
19.
J Infect Dis ; 191(7): 1116-22, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15747247

ABSTRACT

BACKGROUND: A phase 2 trial was conducted to assess in young infants the safety, tolerability, infectivity, and immunogenicity of multiple doses of an intranasal vaccine using bovine parainfluenza virus type 3 (bPIV3). METHODS: One hundred ninety-two healthy 2-month-old infants were randomized 1 : 1 : 1 to receive 1x10(5) median tissue culture infective dose (TCID(50)) bPIV3 vaccine, 1x10(6) TCID(50) bPIV3 vaccine, or placebo at 2, 4, 6, and 12-15 months of age. Safety information was collected by use of diary sheets and telephone interviews. Nasal wash and serum specimens were collected for assessment of infectivity and immunogenicity. RESULTS: The safety profiles of both dosages of bPIV3 were similar to that of placebo, with the exception of fever with temperature of >/=38.1 degrees C after dose 2 only, occurring in 34% of the 1x10(5) TCID(50) group, 35% of the 1x10(6) TCID(50) group, and 12% of the placebo group (P<.01). No vaccine-related serious adverse events were reported. The cumulative vaccine infectivity (isolation of bPIV3 and/or bPIV3 seroconversion) after dose 3 was similar in the 2 vaccine groups (87% in the 1x10(5) TCID(50) group and 77% in the 1x10(6) TCID(50) group) (P=.46). Seroconversion rates after dose 3, assessed by means of hemagglutination inhibition assay, after adjustment for decrease in maternal antibody titers, were 67% in the 1x10(5) TCID(50) group, 57% in the 1x10(6) TCID(50) group, and 12% in the placebo group (P<.01). Isolation of bPIV3 was common after dose 1, dose 2, or dose 3, but only 1 of 51 participants in the vaccine groups had bPIV3 isolated after dose 4. CONCLUSIONS: Multiple doses of bPIV3 vaccine were well tolerated and immunogenic in young infants.


Subject(s)
Antibodies, Viral/blood , Parainfluenza Vaccines/adverse effects , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Bovine/immunology , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Administration, Intranasal , Hemagglutination Inhibition Tests , Humans , Infant , Nasal Lavage Fluid/immunology , Nasal Lavage Fluid/virology , Parainfluenza Vaccines/administration & dosage , United States
20.
Vet Ther ; 5(3): 173-86, 2004.
Article in English | MEDLINE | ID: mdl-15578450

ABSTRACT

The results of this study confirmed that dogs vaccinated subcutaneously with a commercially available multivalent vaccine containing modified-live canine distemper virus, canine adenovirus type 2, canine parvovirus type 2b, and canine parainfluenza virus antigens were protected against sequential experimental challenge 55 to 57 months after initial vaccination given at 7 to 8 weeks of age. All 10 vaccinates were protected against clinical diseases and mortality following parvovirus and infectious canine hepatitis experimental infections. All vaccinates were protected against mortality and 90% against clinical disease following distemper challenge. These data support at least a 4-year duration of immunity for these three "core" fractions in the combination vaccine.


Subject(s)
Dog Diseases/prevention & control , Viral Vaccines , Virus Diseases/veterinary , Adenoviruses, Canine/immunology , Animals , Antibodies, Viral/biosynthesis , Distemper/prevention & control , Distemper Virus, Canine/immunology , Dog Diseases/immunology , Dogs , Female , Hepatitis, Infectious Canine/prevention & control , Male , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/immunology , Parvoviridae Infections/prevention & control , Parvoviridae Infections/veterinary , Parvovirus, Canine/immunology , Parvovirus, Canine/pathogenicity , Random Allocation , Time Factors , Treatment Outcome , Vaccination/veterinary , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Virulence , Virus Diseases/immunology , Virus Diseases/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...