Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
medRxiv ; 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-37790382

ABSTRACT

All four serotypes of dengue virus (DENV) cause the full spectrum of disease. Therefore, vaccines must protect against all serotypes. To evaluate candidate vaccines, a human challenge model of dengue serotype 3 (rDEN30Δ30) was developed. All challenge virus recipients safely met the primary endpoint of viremia and secondary endpoints of rash and seroconversion to DENV-3.

2.
Vaccine ; 37(50): 7443-7450, 2019 11 28.
Article in English | MEDLINE | ID: mdl-27916410

ABSTRACT

On 1 February 2016, in the context of the ongoing Zika virus epidemic, the WHO declared that the recently reported clusters of microcephaly and other neurological disorders constituted a Public Health Emergency of International Concern (PHEIC). In response, WHO in collaboration with UNICEF and a working group of independent subject matter experts developed a Zika virus vaccine Target Product Profile (TPP) for use in an emergency, or in a future outbreak scenario. The drafting process of the Zika virus vaccine TPP included the opportunity for public comment, as well as consultation with epidemiologists, flavivirus vaccine subject matter experts, vaccine developers and global regulators to consider the regulatory expectations and potential emergency use pathways for a vaccine with the characteristics described in the TPP. This report summarizes an expert consultation held 6-7 June 2016 on the regulatory considerations for a Zika vaccine for emergency use.


Subject(s)
Disease Outbreaks/prevention & control , Viral Vaccines/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Emergencies , Humans , Public Health/methods , Referral and Consultation , World Health Organization
3.
Am J Trop Med Hyg ; 65(5): 405-13, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11716091

ABSTRACT

The recombinant dengue virus type-4 vaccine candidate 2AA30 was attenuated in rhesus monkeys due to an engineered 30-nucleotide deletion in the 3'-untranslated region of the viral genome. A clinical trial to evaluate the safety and immunogenicity of a single dose of 2Adelta30 was conducted with 20 adult human volunteers. The vaccine candidate was well tolerated and did not cause systemic illness in any of the 20 volunteers. Viremia was detectable in 14 volunteers at a mean level of 1.6 log10 plaque-forming units/ml of serum, although all 20 volunteers seroconverted with a seven-fold or greater increase in serum neutralizing antibody titer on day 28 post-vaccination (mean titer = 1:580). A mild, asymptomatic, macular rash developed in 10 volunteers, and a transient elevation in the serum level of alanine aminotransferase was noted in five volunteers. The low level of reactogenicity and high degree of immunogenicity of this vaccine candidate warrant its further evaluation and its use to create chimeric vaccine viruses expressing the structural genes of dengue virus types 1, 2, and 3.


Subject(s)
3' Untranslated Regions/physiology , Dengue Virus/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adult , Animals , Dengue Virus/genetics , Dengue Virus/physiology , Humans , Immunization , Macaca mulatta , Vaccines, Attenuated/immunology , Virus Replication
4.
Am J Trop Med Hyg ; 65(5): 414-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11716092

ABSTRACT

2Adelta30 is a live dengue-4 virus vaccine candidate with a 30-nucleotide deletion in its 3'-untranslated region. To assess the transmissibility of 2Adelta30 by mosquitoes, we compared its in vivo replication in mosquitoes with that of its wild type DEN-4 parent. Both the vaccine candidate and wild type virus were equally able to infect the mosquito Toxorhynchites splendens after intrathoracic inoculation. Relative to its wild type parent, 2Adelta30 was slightly restricted in its ability to infect the midgut of Aedes aegypti mosquitoes fed on an artificial blood meal and was even more restricted in its ability to disseminate from the midgut to the salivary glands. Thus, the 30-nucleotide deletion rendered the vaccine candidate more sensitive than its wild type parent to the mosquito midgut escape barrier. Most significantly, 2Adelta30 was not transmitted to 352 Ae. albopictus mosquitoes fed on 10 vaccinees, all of whom were infected with the vaccine candidate.


Subject(s)
Aedes/virology , Dengue Virus/immunology , Dengue/transmission , Insect Vectors/virology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adult , Animals , Dengue Virus/physiology , Female , Humans , Vaccination , Vaccines, Attenuated/immunology , Virus Replication
5.
J Virol ; 74(15): 6821-31, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10888621

ABSTRACT

Recombinant human parainfluenza virus type 3 (PIV3) was used as a vector to express the major protective antigen of measles virus, the hemagglutinin (HA) glycoprotein, in order to create a bivalent PIV3-measles virus that can be administered intranasally. The measles virus HA open reading frame (ORF) was inserted as an additional transcriptional unit into the N-P, P-M, or HA-neuraminidase (HN)-L gene junction of wild-type PIV3 or into the N-P or P-M gene junction of an attenuated derivative of PIV3, termed rcp45L. The recombinant PIV3 (rPIV3) viruses bearing the HA inserts replicated more slowly in vitro than their parental viruses but reached comparable peak titers of >/=10(7.5) 50% tissue culture infective doses per ml. Each of the wild-type or cold-passaged 45L (cp45L) PIV3(HA) chimeric viruses replicated 5- to 10-fold less well than its respective parent virus in the upper respiratory tract of hamsters. Thus, insertion of the approximately 2-kb ORF itself conferred attenuation, and this attenuation was additive to that conferred by the cp45L mutations. The attenuated cp45L PIV3(HA) recombinants induced a high level of resistance to replication of PIV3 challenge virus in hamsters and induced very high levels of measles virus neutralizing antibodies (>1:8,000) that are well in excess of those known to be protective in humans. rPIV3s expressing the HA gene in the N-P or P-M junction induced about 400-fold more measles virus-neutralizing antibody than did the rPIV3 with the HA gene in the HN-L junction, indicating that the N-P or P-M junction appears to be the preferred insertion site. Previous studies indicated that the PIV3 cp45 virus, a more attenuated version of rcp45L, replicates efficiently in the respiratory tract of monkeys and is immunogenic and protective even when administered in the presence of very high titers of passively transferred PIV3 antibodies (A. P. Durbin, C. J. Cho, W. R. Elkins, L. S. Wyatt, B. Moss, and B. R. Murphy, J. Infect. Dis. 179:1345-1351, 1999). This suggests that this intranasally administered PIV3(HA) chimeric virus can be used to immunize infants with maternally acquired measles virus antibodies in whom the current parenterally administered live measles virus vaccine is ineffective.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles virus/immunology , Measles/prevention & control , Parainfluenza Virus 3, Human/genetics , Animals , Antibodies, Viral/blood , Base Sequence , Cells, Cultured , Cricetinae , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Humans , Infant , Measles virus/genetics , Mesocricetus , Molecular Sequence Data , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/physiology , Temperature , Vaccination , Vaccines, Synthetic/immunology , Virus Replication
6.
Virology ; 272(1): 225-34, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10873765

ABSTRACT

Recombinant parainfluenza virus 3 (rPIV3) is being developed as a vector to express foreign genes as a bivalent or multivalent live attenuated virus vaccine. In the present study, we examined the effect of inserted foreign sequence on virus replication in vitro and in vivo, focusing on the parameter of insert length. In one type of construct, foreign sequence of increasing length was flanked by PIV3 transcription signals and inserted as an additional gene unit (GU insert) between the HN and L genes, so that one additional mRNA would be made. In a second type of construct, foreign sequence was inserted into the downstream NCR (NCR insert) of the HN gene, so that the number of encoded mRNAs remained unchanged. In each case, the foreign sequence was designed to lack any significant open reading frame, which permitted an evaluation of the effect of insert length on replication independent of an effect of an expressed protein. The GU or NCR insert sizes ranged from 168 nucleotides (nt) to 3918 nt. rPIV3s containing GU insertions of up to 3918 nt in length, the largest size tested, were viable and replicated efficiently at permissive temperatures in vitro, but a reduction in plaque size was seen at 39 degrees C and 40 degrees C. The rPIV3 with a 3918-nt GU insertion was restricted in replication in the upper (fivefold) and lower (25-fold) respiratory tracts of hamsters. Although a 1908-nt GU insertion did not significantly modify replication of wild-type PIV3 in vitro or in vivo, its introduction significantly augmented the level of temperature sensitivity (ts) and attenuation (att) specified by three mutations in the L protein of a cold-passaged attenuated PIV3 vaccine virus. rPIV3s bearing a 3126- or 3894-nt NCR insertion exhibited in vitro and in vivo phenotypes like those of the rPIV3s bearing similar-sized GU insertions. These findings indicate that rPIV3s whose genome length has been increased by more than 3000 nt by either a GU or an NCR insertion exhibit an unexpected host-range phenotype, that is, efficient replication in vitro but restricted replication in hamsters, especially in the lower respiratory tract. Furthermore, these effects were greatly enhanced when the rPIV3 backbone contained other ts or att mutations. The implications of these findings for the use of single-stranded, negative-sense RNA viruses as vectors for vaccines are discussed.


Subject(s)
HN Protein/genetics , Mutagenesis, Insertional/genetics , Nucleotides/genetics , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , Viral Proteins/genetics , Animals , Base Sequence , Cell Line , Cricetinae , Genetic Vectors/genetics , Genome, Viral , Mesocricetus , Molecular Sequence Data , Molecular Weight , Phenotype , RNA, Viral/genetics , Respiratory System/virology , Respirovirus Infections/virology , Temperature , Viral Plaque Assay , Viral Vaccines/genetics , Virus Replication/genetics
7.
Vaccine ; 18(22): 2462-9, 2000 May 08.
Article in English | MEDLINE | ID: mdl-10738104

ABSTRACT

Human parainfluenza virus (HPIV) types-1, -2, and -3 are significant causes of both upper and lower respiratory tract disease in infants and children. Although there are two live attenuated vaccines for the prevention of HPIV-3 disease in phase 1 clinical trials, vaccines are not currently available for prevention of HPIV-1 or -2 disease. Our laboratory is developing candidate vaccines for the prevention of HPIV-1, -2, and -3 disease, and a suitable nonhuman primate model is needed for evaluation of these vaccine candidates prior to administration to humans. We evaluated the replication of HPIV-1 and -2 in six different species of nonhuman primates and found both viruses to replicate most efficiently in African green monkeys and chimpanzees. We then compared the replication of HPIV-3 in African green monkeys to that in rhesus macaques, which we have used previously, and found that HPIV-3 replicated to higher titer in African green monkeys. In summary, African green monkeys provide a very useful nonhuman primate for the evaluation of HPIV-1, -2, and -3 vaccine candidates, especially for the evaluation of various combinations of these PIV vaccines and for vaccine strategies that employ sequential immunization.


Subject(s)
Chlorocebus aethiops/immunology , Paramyxoviridae Infections/etiology , Paramyxoviridae Infections/immunology , Animals , Aotidae , Disease Models, Animal , Humans , Macaca mulatta , Macaca nemestrina , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 2, Human/physiology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/physiology , Paramyxoviridae Infections/prevention & control , Saimiri , Species Specificity , Viral Vaccines/isolation & purification , Virus Replication
8.
J Virol ; 74(7): 3188-95, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10708435

ABSTRACT

The shipping fever (SF) and Kansas (Ka) strains of bovine parainfluenza virus type 3 (BPIV3) are restricted in their replication in rhesus monkeys 100- to 1,000-fold compared to human parainfluenza virus type 3 (HPIV3), and the Ka strain also was shown to be attenuated in humans. To initiate an investigation of the genetic basis of the attenuation of BPIV3 in primates, we produced viable chimeric HPIV3 recombinants containing the nucleoprotein (N) open reading frame (ORF) from either BPIV3 Ka or SF in place of the HPIV3 N ORF. These chimeric recombinants were designated cKa-N and cSF-N, respectively. Remarkably, cKa-N and cSF-N grew to titers comparable to those of their HPIV3 and BPIV3 parents in LLC-MK2 monkey kidney and Madin-Darby bovine kidney cells. Thus, the heterologous nature of the N protein did not impede replication in vitro. However, cKa-N and cSF-N were each restricted in replication in rhesus monkeys to a similar extent as Ka and SF, respectively. This identified the BPIV3 N protein as a determinant of the host range restriction of BPIV3 in primates. These chimeras thus combine the antigenic determinants of HPIV3 with the host range restriction and attenuation phenotype of BPIV3. Despite their restricted replication in rhesus monkeys, the chimeric viruses induced a level of resistance to HPIV3 challenge in these animals which was indistinguishable from that conferred by immunization with HPIV3. The infectivity, attenuation, and immunogenicity of these BPIV3/HPIV3 chimeras suggest that the modified Jennerian approach described in the present report represents a novel method to design vaccines to protect against HPIV3-induced disease in humans.


Subject(s)
Nucleocapsid/genetics , Parainfluenza Virus 3, Human/genetics , Respirovirus/genetics , Animals , Base Sequence , Cell Line , DNA Primers , Macaca mulatta , Open Reading Frames , Parainfluenza Virus 3, Human/physiology , Recombination, Genetic , Respirovirus/physiology , Virus Replication
9.
Vaccine ; 18(14): 1359-66, 2000 Jan 31.
Article in English | MEDLINE | ID: mdl-10618533

ABSTRACT

Using a reverse genetics system for PIV3, we previously recovered recombinant chimeric PIV3-PIV1 virus bearing the major protective antigens of PIV1, the hemaglutinin-neuraminidase and fusion proteins, on a background of PIV3 genes bearing temperature sensitive (ts) and attenuating mutations in the L gene. Immunization of hamsters with this virus, designated rPIV3-1.cp45L, induced a high level of resistance to replication of wild type (wt) PIV1 and, surprisingly, also induced a moderate amount of restriction of the replication of PIV3 challenge virus. This suggested that some immunity is conferred by the internal PIV3 proteins shared by the two viruses. In the present study, we found that the immunity to PIV3 conferred by infection with rPIV3-1.cp45L is short-lived and completely disappeared four months after immunization, whereas resistance to replication of PIV3 induced by prior infection with PIV3 remains high even after an interval of four months. Since a live attenuated PIV1 vaccine such as the chimeric rPIV3-1.cp45L virus will likely be given to infants after a live attenuated PIV3 vaccine in a sequential immunization schedule, we examined the immunogenicity and efficacy of rPIV3-1.cp45L against PIV1 challenge in animals with and without prior immunity to PIV3. rPIV3-1.cp45L efficiently infected hamsters previously infected with wt or attenuated PIV3, but there was approximately a five-fold reduction in replication of rPIV3-1. cp45L virus in the PIV3-immune animals. This reduction in replication of rPIV3-1.cp45L in PIV3-immune animals was accompanied by a significant decrease in efficacy against PIV1 challenge. However, rPIV3-1.cp45L immunization of PIV3-immune animals induced a vigorous serum antibody response to PIV1 and reduced replication of PIV1 challenge virus 1000-fold in the lower respiratory tract and 25 to 200-fold in the upper respiratory tract. This study demonstrated that the recombinant chimeric rPIV3-1.cp45L candidate vaccine can induce immunity to PIV1 even in animals immune to PIV3. This establishes the feasibility of employing a sequential immunization schedule in which a recombinant chimeric rPIV3-1.cp45L vaccine is given following a live attenuated PIV3 vaccine.


Subject(s)
Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Viral Vaccines/therapeutic use , Animals , Cricetinae , Evaluation Studies as Topic , Hemagglutinins/immunology , Immunization , Mesocricetus , Neuraminidase/immunology , Parainfluenza Virus 2, Human/immunology , Recombinant Fusion Proteins/immunology , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/immunology
10.
Virology ; 261(2): 319-30, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10497117

ABSTRACT

Human parainfluenza virus type 3 (HPIV3) is a single-stranded negative-sense RNA virus belonging to the Respirovirus genus of the Paramyxoviridae family in the order Mononegavirales. The P gene encodes at least four proteins, including the C protein, which is expressed from an open reading frame (ORF) that overlaps the P ORF, and the D protein, which is encoded when the P ORF is fused to the D ORF by transcriptional editing. The P mRNA also contains a third ORF for the V protein, although it is unclear how or whether this ORF is accessed. We have used recombinant DNA technology to recover five mutant viruses that either interrupt or alter the C, D, and V ORFs. In one mutant virus, rC-KO, expression of the C protein was abrogated by changing the start codon from methionine to threonine and introducing two stop codons at amino acid positions 7 and 26 of the C ORF. In a second mutant virus, rF164S, a point mutation was introduced into the C ORF changing amino acid position 164 from phenylalanine (F) to serine (S), which corresponds to the F170S mutation described in the C protein of Sendai virus (Itoh et al., J. Gen. Virol. 78, 3207-3215). rC-KO was significantly attenuated in vitro and in vivo (rodents and primates), whereas rF164S was attenuated only in vivo. Interestingly, the rF164S mutant was more attenuated in the upper than in the lower respiratory tract of hamsters and monkeys. This pattern is the converse of that seen with temperature-sensitive attenuating mutations, and thus inclusion of this novel mutation in a recombinant live-attenuated vaccine candidate might prove useful in reducing residual virulence in the upper respiratory tract. Both rC-KO and rF164S conferred protection against challenge with wild-type HPIV3. In three other viruses, the D and V ORFs were interrupted singly or in combination. Although interruption of the D and V ORFs individually did not affect virus replication in vitro or in vivo, interruption of both together attenuated replication in vivo. These results indicate that the C, D, and V proteins of HPIV3 each has a role in virus replication in vitro, in vivo, or both, and define mutations that might be useful for the development of a vaccine against HPIV3.


Subject(s)
Genes, Viral , Open Reading Frames/genetics , Parainfluenza Virus 3, Human/physiology , Parainfluenza Virus 3, Human/pathogenicity , Respirovirus Infections/virology , Animals , Cricetinae , Humans , Mutation , Primates , Recombination, Genetic , Virulence/genetics , Virus Replication/genetics
11.
J Infect Dis ; 179(6): 1345-51, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10228053

ABSTRACT

Two parainfluenza virus type 3 (PIV3) vaccine candidates-cp45, a live attenuated derivative of the JS wild type (wt), and a replication-defective vaccinia virus recombinant expressing the hemagglutinin-neuraminidase or fusion glycoprotein of human PIV3 (modified vaccinia virus Ankara [MVA]/PIV3 recombinants)-were evaluated in rhesus monkeys to determine whether successful immunization could be achieved in the presence of passively transferred PIV3 antibodies. The cp45 virus, administered intranasally (in) and intratracheally (it) in the presence of high levels of PIV3 antibodies, replicated efficiently in the nasopharynx and protected against challenge with wt human PIV3. The MVA recombinants, administered in, it, and intramuscularly in the absence of passive antibody, conferred protection against replication of PIV3 wt challenge virus, but this was largely abrogated when immunization occurred in the presence of passive antibodies. Because immunization for the prevention of HPIV3 disease must occur in early infancy when maternal antibodies are present, the live attenuated cp45 virus continues to be a promising vaccine candidate for this age group.


Subject(s)
Antigens, Viral/therapeutic use , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Vaccination , Viral Vaccines/therapeutic use , Animals , Antibodies, Viral/blood , Antibodies, Viral/therapeutic use , Immunization, Passive , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Respiratory System/virology , Vaccines, Synthetic/therapeutic use , Virus Replication
12.
Vaccine ; 17(9-10): 1100-8, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10195620

ABSTRACT

The recovery of wild type and attenuated human parainfluenza type 3 (PIV3) recombinant viruses has made possible a new strategy to rapidly generate a live-attenuated vaccine virus fof PIV1. We previously replaced the coding sequences for the hemagglutinin-neuraminidase (HN) and fusion (F) proteins of PIV3 with those of PIV1 in the PIV3 antigenomic cDNA. This was used to recover a fully-viable, recombinant chimeric PIV3-PIV1 virus, termed rPIV3-1, which bears the major protective antigens of PIV1 and is wild type-like with regard to growth in cell culture and in hamsters [Tao T, Durbin AP, Whitehead SS, Davoodi F, Collins PL, Murphy BR. Recovery of a fully viable chimeric human parainfluenza virus (PIV) type 3 in which the hemagglutinin-neuraminidase and fusion glycoprotein have been replaced by those of PIV type 1. J Virol 1998;72:2955-2961]. Here we report the recovery of a derivative of rPIV3-1 carrying the three temperature-sensitive and attenuating amino acid coding changes found in the L gene of the live-attenuated cp45 PIV3 candidate vaccine virus. This virus, termed rPIV3-1.cp45L, is temperature-sensitive with a shut-off temperature of 38 degrees C, which is similar to that of the recombinant rPIV3cp45L, which possesses the same three mutations. rPIV3-1.cp45L is attenuated in the respiratory tract of hamsters to the same extent as rPIV3cp45L. Infection of hamsters with rPIV3-1.cp45L generated a moderate level of hemagglutination-inhibiting antibodies against wild type PIV1 and induced complete resistance to challenge with wild type PIV1. This demonstrates that this novel attenuated chimeric virus is capable of inducing a highly effective immune response against PIV1. It confirms previous observations that the surface glycoproteins of parainfluenza viruses are sufficient to induce a high level of resistance to homologous virus challenge. Unexpectedly, infection with recombinant chimeric virus rPIV3-1.cp45L or rPIV3-1, each bearing the surface glycoprotein genes of PIV1 and the internal genes of PIV3, also induced a moderate level of resistance to replication of wild type PIV3 challenge virus. This indicates that the internal genes of PIV3 can independently induce protective immunity against PIV3 in rodents, albeit a lower level of resistance than that induced by the surface glycoproteins. Thus, a reverse genetics system for PIV3 has been used successfully to produce a live attenuated PIV1 vaccine candidate that is attenuated and protective in experimental infection in hamsters.


Subject(s)
Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 3, Human/immunology , Vaccines, Attenuated/immunology , Viral Fusion Proteins/immunology , Animals , Cell Line , Cricetinae , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization , Mesocricetus , Parainfluenza Virus 1, Human/physiology , Receptors, Virus/genetics , Receptors, Virus/immunology , Respiratory System/virology , Surface Properties , Temperature , Virus Replication
13.
J Virol ; 73(2): 1374-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9882342

ABSTRACT

The live-attenuated human parainfluenza virus 3 (PIV3) cold-passage 45 (cp45) candidate vaccine was shown previously to be safe, immunogenic, and phenotypically stable in seronegative human infants. Previous findings indicated that each of the three amino acid substitutions in the L polymerase protein of cp45 independently confers the temperature-sensitive (ts) and attenuation (att) phenotypes but not the cold-adaptation (ca) phenotype (29). cp45 contains 12 additional potentially important point mutations in other proteins (N, C, M, F, and hemagglutinin-neuraminidase [HN]) or in cis-acting sequences (the leader region and the transcription gene start [GS] signal of the N gene), and their contribution to these phenotypes was undefined. To further characterize the genetic basis for the ts, ca, and att phenotypes of this promising vaccine candidate, we constructed, using a reverse genetics system, a recombinant cp45 virus that contained all 15 cp45-specific mutations mentioned above, and found that it was essentially indistinguishable from the biologically derived cp45 on the basis of plaque size, level of temperature sensitivity, cold adaptation, level of replication in the upper and lower respiratory tract of hamsters, and ability to protect hamsters from subsequent wild-type PIV3 challenge. We then constructed recombinant viruses containing the cp45 mutations in individual proteins as well as several combinations of mutations. Analysis of these recombinant viruses revealed that multiple cp45 mutations distributed throughout the genome contribute to the ts, ca, and att phenotypes. In addition to the mutations in the L gene, at least one other mutation in the 3' N region (i.e., including the leader, N GS, and N coding changes) contributes to the ts phenotype. A recombinant virus containing all the cp45 mutations except those in L was more ts than cp45, illustrating the complex nature of this phenotype. The ca phenotype of cp45 also is a complex composite phenotype, reflecting contributions of at least three separate genetic elements, namely, mutations within the 3' N region, the L protein, and the C-M-F-HN region. The att phenotype is a composite of both ts and non-ts mutations. Attenuating ts mutations are located in the L protein, and non-ts attenuating mutations are located in the C and F proteins. The presence of multiple ts and non-ts attenuating mutations in cp45 likely contributes to the high level of attenuation and phenotypic stability of this promising vaccine candidate.


Subject(s)
Mutation , Parainfluenza Virus 3, Human/genetics , Viral Vaccines/genetics , Adaptation, Biological , Animals , Cell Line , Cold Temperature , Cricetinae , Humans , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/immunology , Phenotype , Vaccines, Attenuated , Viral Plaque Assay , Viral Vaccines/immunology
14.
Vaccine ; 16(13): 1324-30, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9682397

ABSTRACT

Immunization of rhesus monkeys with modified vaccinia Ankara (MVA) recombinants expressing the haemagglutinin-neuraminidase (HN) or fusion (F) glycoproteins of human parainfluenza virus type 3 (HPIV3) was compared with an intranasally-administered live, attenuated HPIV3 vaccine candidate, the cp45 derivative of the JS strain of wildtype HPIV3. The MVA recombinants, when given parenterally (i.m.) or as a parenteral-local (i.m. and i.t.) combination, induced an antibody response comparable to that of cp45 and protected the upper and lower respiratory tracts of the rhesus monkeys against challenge with wildtype HPIV3. When given by the i.n. route alone, the MVA/PIV3 recombinants induced a serum antibody response that was comparable to that of cp45 and induced resistance in the lower respiratory tract. Despite the ability of the intranasally-administered MVA/PIV3 recombinants to stimulate a good serological response and to protect the lower respiratory tract, they unexpectedly failed to induce a significant level of resistance in the upper respiratory tract. The live, attenuated virus vaccine candidate induced almost complete resistance in both the upper and lower tracts. The data thus identify two vaccine candidates that can protect both the upper and lower respiratory tracts of rhesus monkey, parenterally-administered MVA/PIV3 and intranasally-administered cp45. Further studies with these vaccines in non-human primates and humans should identify the relative merits of these immunogens for use in the very young infant.


Subject(s)
Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/biosynthesis , Chick Embryo , HN Protein/immunology , Injections, Intramuscular , Macaca mulatta , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Attenuated/standards , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/standards , Vaccinia virus/immunology , Viral Fusion Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/standards
15.
J Virol ; 72(4): 2955-61, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9525616

ABSTRACT

The recent recovery of human parainfluenza virus type 3 (PIV3) from cDNA, together with the availability of a promising, highly characterized live attenuated PIV3 vaccine virus, suggested a novel strategy for the rapid development of comparable recombinant vaccine viruses for human PIV1 and PIV2. The strategy, illustrated here for PIV1, is to create chimeric viruses in which the two protective antigens, the hemagglutinin-neuraminidase (HN) and fusion (F) envelope glycoproteins, of an attenuated PIV3 variant are replaced by those of PIV1 or PIV2. As a first step, this has been achieved by using recombinant wild-type (wt) PIV3 as the recipient for PIV1 HN and F, engineered so that each PIV1 open reading frame is flanked by the existing PIV3 nontranslated regions and transcription signals. This yielded a viable chimeric recombinant virus, designated rPIV3-1, that encodes the PIV1 HN and F glycoproteins in the background of the wt PIV3 internal proteins. There were three noteworthy findings. First, in contrast to recently reported glycoprotein replacement chimeras of vesicular somatitis virus or measles virus, the PIV3-1 chimera replicates in LLC-MK2 cells and in the respiratory tract of hamsters as efficiently as its PIV1 and PIV3 parents. This is remarkable because the HN and F glycoproteins share only 43 and 47%, respectively, overall amino acid sequence identity between serotypes. In particular, the cytoplasmic tails share only 9 to 11% identity, suggesting that their presumed role in virion morphogenesis does not involve sequence-specific contacts. Second, rPIV3-1 was found to possess biological properties derived from each of its parent viruses. Specifically, it requires trypsin for efficient plaque formation in tissue culture, like its PIV1 parent but unlike PIV3. On the other hand, it causes an extensive cytopathic effect (CPE) in LLC-MK2 cultures which resembles that of its PIV3 parent but differs from that of its noncytopathic PIV1 parent. This latter finding indicates that the genetic basis for the CPE of PIV3 in tissue culture lies outside regions encoding the HN or F glycoprotein. Third, it should now be possible to rapidly develop a live attenuated PIV1 vaccine by the staged introduction of known, characterized attenuating mutations present in a live attenuated PIV3 vaccine candidate into the PIV3-1 cDNA followed by recovery of attenuated derivatives of rPIV3-1.


Subject(s)
Glycoproteins/genetics , HN Protein/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , Viral Fusion Proteins/genetics , Animals , Antigens, Viral/genetics , Base Sequence , Cell Line , Cloning, Molecular , Cricetinae , DNA, Viral , Glycoproteins/physiology , HN Protein/physiology , Humans , Mesocricetus , Molecular Sequence Data , RNA, Viral , Reassortant Viruses/genetics , Reassortant Viruses/physiology , Tumor Cells, Cultured , Viral Fusion Proteins/physiology , Virus Replication
16.
J Virol ; 72(3): 1762-8, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499025

ABSTRACT

Studies were initiated to define the genetic basis of the temperature-sensitive (ts), cold adaptation (ca), and attenuation (att) phenotypes of the human parainfluenza virus type 3 (PIV3) cp45 live attenuated vaccine candidate. Genetic data had previously suggested that the L polymerase protein of cp45, which contains three amino acid substitutions at positions 942, 992, and 1558, contributed to its temperature sensitivity (R. Ray, M. S. Galinski, B. R. Heminway, K. Meyer, F. K. Newman, and R. B. Belshe, J. Virol. 70:580-584, 1996; A. Stokes, E. L. Tierney, C. M. Sarris, B. R. Murphy, and S. L. Hall, Virus Res. 30:43-52, 1993). To study the individual and aggregate contributions that these amino acid substitutions make to the ts, att, and ca phenotypes of cp45, seven PIV3 recombinant viruses (three single, three double, and one triple mutant) representing all possible combinations of the three amino acid substitutions were recovered from full-length antigenomic cDNA and analyzed for their ts, att, and ca phenotypes. None of the seven mutant recombinant PIVs was cold adapted. The substitutions at L protein amino acid positions 992 and 1558 each specified a 105-fold reduction in plaque formation in cell culture at 40 degrees C, whereas the substitution at position 942 specified a 300-fold reduction. Thus, each of the three mutations contributes individually to the ts phenotype. The triple recombinant which possesses an L protein with all three mutations was almost as temperature sensitive as cp45, indicating that these mutations are the major contributors to the ts phenotype of cp45. The three individual mutations in the L protein each contributed to restricted replication in the upper or lower respiratory tract of hamsters, and this likely contributes to the observed stability of the ts and att phenotypes of cp45 during replication in vivo. Importantly, the recombinant virus possessing L protein with all three mutations was as restricted in replication as was the cp45 mutant in both the upper and lower respiratory tracts of hamsters, indicating that the L gene of the cp45 virus is a major attenuating component of this candidate vaccine.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Parainfluenza Virus 3, Human/enzymology , Vaccines, Attenuated , Viral Proteins/metabolism , Viral Vaccines , Animals , Cell Line , Cricetinae , DNA-Directed RNA Polymerases/genetics , Humans , Macaca mulatta , Mesocricetus , Mutagenesis, Site-Directed , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/growth & development , Phenotype , Temperature , Tumor Cells, Cultured , Viral Plaque Assay , Viral Proteins/genetics
17.
Virology ; 235(2): 323-32, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9281512

ABSTRACT

Infectious HPIV3 was produced by the intracellular coexpression of four plasmid-borne cDNAs. These separately encoded a complete HPIV3 genome (negative-sense), the HPIV3 nucleocapsid protein N, the phosphoprotein P, and the polymerase protein L. The cDNA-encoded HPIV3 genome differed from the JS wildtype (wt) strain of HPIV3 used in its construction by seven point mutations: four of these are silent mutations in the HN or L gene coding regions that serve as markers of a cDNA-derived virus, two were introduced to create an amino acid substitution that ablates an epitope recognized by the HN-specific monoclonal neutralizing antibody 423/6, and the remaining point mutation results in an incidental amino acid substitution in the HN protein at amino acid position 263. The four plasmids were transfected into HEp-2 cell monolayers and their expression was driven by T7 RNA polymerase supplied by a vaccinia virus recombinant. The titer of virus present in the harvested transfection supernatant was low (<5 PFU/ml), and the recovered recombinant virus (rJS) retained each of the seven mutations present in the cDNA from which it was derived. Despite the introduced and incidental mutations, rJS retained the wt phenotypes as regards replication at elevated temperature in vitro and efficient replication in the upper and lower respiratory tract of hamsters. rJS was also recovered from a cDNA encoding a complete antigenome (positive-sense) with slightly greater efficiency than from the negative-sense construct. The ability to generate infectious HPIV3 from cDNA should greatly enhance our ability to develop new live-attenuated parainfluenza virus vaccines, including chimeric PIV1 and PIV2 vaccines, and to understand the genetic basis of attenuation of PIV3 candidate vaccines.


Subject(s)
DNA, Viral/physiology , Nucleoproteins , Parainfluenza Virus 3, Human/genetics , Animals , Base Sequence , Cells, Cultured , Cricetinae , DNA, Complementary , Genome, Viral , Humans , Mutation , Nucleocapsid Proteins , Parainfluenza Virus 3, Human/pathogenicity , Respiratory System/virology , Temperature , Viral Core Proteins/genetics , Viral Proteins/genetics
18.
Virology ; 234(1): 74-83, 1997 Jul 21.
Article in English | MEDLINE | ID: mdl-9234948

ABSTRACT

A reconstituted transcription and RNA replication system for human parainfluenza virus type 3 (HPIV3) was developed using components expressed intracellularly from transfected plasmids driven by T7 RNA polymerase supplied by a vaccinia virus recombinant. The system is based on a negative-sense analog of HPIV3 genomic RNA in which the viral genes were deleted and replaced with that encoding bacterial chloramphenicol acetyl transferase (CAT). The N, P, and L proteins expressed from cotransfected plasmids were necessary and sufficient to direct efficient transcription and RNA replication. Transcription yielded subgenomic polyadenylated mRNA, which was isolated by oligo(dT) chromatography. RNA replication yielded a mini-antigenome and progeny minigenome, which were shown to be encapsidated based on resistance to digestion with micrococcal nuclease. A panel of cDNAs was constructed to encode minigenomes which differed in length by single-nucleotide increments. Transcription and RNA replication in the reconstituted system were most efficient for the minigenome whose length was an even multiple of six. Both RNA replication and transcription appeared to be governed by the rule. However, minigenomes whose lengths were one nucleotide greater than or less than an even multiple of six also were very active, especially in RNA replication, indicating that the rule was not absolute.


Subject(s)
Genome, Viral , Nucleoproteins , Parainfluenza Virus 3, Human/genetics , RNA, Viral/biosynthesis , Transcription, Genetic , Chloramphenicol O-Acetyltransferase/genetics , Genes, Reporter , Humans , Nucleocapsid Proteins , Phosphoproteins/genetics , Viral Core Proteins/genetics , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...