Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Virol ; 82(18): 8965-77, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614629

ABSTRACT

Recombinant human parainfluenza virus type 1 (rHPIV1) was modified to create rHPIV1-P(C-), a virus in which expression of the C proteins (C', C, Y1, and Y2) was silenced without affecting the amino acid sequence of the P protein. Infectious rHPIV1-P(C-) was readily recovered from cDNA, indicating that the four C proteins were not essential for virus replication. Early during infection in vitro, rHPIV1-P(C-) replicated as efficiently as wild-type (wt) HPIV1, but its titer subsequently decreased coincident with the onset of an extensive cytopathic effect not observed with wt rHPIV1. rHPIV1-P(C-) infection, but not wt rHPIV1 infection, induced caspase 3 activation and nuclear fragmentation in LLC-MK2 cells, identifying the HPIV1 C proteins as inhibitors of apoptosis. In contrast to wt rHPIV1, rHPIV1-P(C-) and rHPIV1-C(F170S), a mutant encoding an F170S substitution in C, induced interferon (IFN) and did not inhibit IFN signaling in vitro. However, only rHPIV1-P(C-) induced apoptosis. Thus, the anti-IFN and antiapoptosis activities of HPIV1 were separable: both activities are disabled in rHPIV1-P(C-), whereas only the anti-IFN activity is disabled in rHPIV1-C(F170S). In African green monkeys (AGMs), rHPIV1-P(C-) was considerably more attenuated than rHPIV1-C(F170S), suggesting that disabling the anti-IFN and antiapoptotic activities of HPIV1 had additive effects on attenuation in vivo. Although rHPIV1-P(C-) protected against challenge with wt HPIV1, its highly restricted replication in AGMs and in primary human airway epithelial cell cultures suggests that it might be overattenuated for use as a vaccine. Thus, the C proteins of HPIV1 are nonessential but have anti-IFN and antiapoptosis activities required for virulence in primates.


Subject(s)
Apoptosis/drug effects , Interferons/antagonists & inhibitors , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 1, Human/pathogenicity , Viral Proteins/metabolism , Virus Replication/drug effects , Animals , Base Sequence , Caspase 3/metabolism , Cell Line , Chlorocebus aethiops , Enzyme Activation , Humans , Molecular Sequence Data , Parainfluenza Virus 1, Human/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Recombination, Genetic , Vero Cells , Viral Proteins/genetics
2.
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
3.
J Virol Methods ; 141(1): 30-3, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17210187

ABSTRACT

We describe a method for the generation of clinical grade, live-attenuated vaccines in Vero cells entirely from cDNA plasmids. The entire electroporation procedure can be completed in less than 15 minutes and this is a significant improvement over previous lipid or electroporation based transfection techniques that also involve a heat-shock step. Importantly, the virus preparations can be generated with a minimal use of animal product derived materials, an important consideration for a vaccine candidate that is to be tested in humans. Since it is likely that all live-attenuated parainfluenza virus and pneumovirus vaccines in the future will be generated using reverse genetics, this simplified method provides guidance on how this can be achieved.


Subject(s)
Clinical Trials as Topic , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Paramyxovirinae/immunology , Viral Vaccines/immunology , Animals , Chlorocebus aethiops , DNA, Complementary , Electroporation , Humans , Kinetics , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxovirinae/genetics , Plasmids , Recombination, Genetic , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Vaccines/genetics
4.
Vaccine ; 25(34): 6409-22, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17658669

ABSTRACT

Previously, we identified several attenuating mutations in the L polymerase protein of human parainfluenza virus type 2 (HPIV2) and genetically stabilized those mutations using reverse genetics [Nolan SM, Surman S, Amaro-Carambot E, Collins PL, Murphy BR, Skiadopoulos MH. Live-attenuated intranasal parainfluenza virus type 2 vaccine candidates developed by reverse genetics containing L polymerase protein mutations imported from heterologous paramyxoviruses. Vaccine 2005;39(23):4765-74]. Here we describe the discovery of an attenuating mutation at nucleotide 15 (15(T-->C)) in the 3' genomic promoter that was also present in the previously characterized mutants. We evaluated the properties of this promoter mutation alone and in various combinations with the L polymerase mutations. Amino acid substitutions at L protein positions 460 (460A or 460P) or 948 (948L), or deletion of amino acids 1724 and 1725 (Delta1724), each conferred a temperature sensitivity (ts) phenotype whereas the 15(T-->C) mutation did not. The 460A and 948L mutations each contributed to restricted replication in the lower respiratory tract of African green monkeys, but the Delta1724 mutation increased attenuation only in certain combinations with other mutations. We constructed two highly attenuated viruses, rV94(15C)/460A/948L and rV94(15C)/948L/Delta1724, that were immunogenic and protective against challenge with wild-type HPIV2 in African green monkeys and, therefore, appear to be suitable for evaluation in humans.


Subject(s)
Mutation , Parainfluenza Vaccines/immunology , Parainfluenza Virus 2, Human/immunology , Promoter Regions, Genetic , Vaccines, Synthetic/immunology , Viral Proteins/genetics , Animals , Cell Line , Cricetinae , Humans , Macaca mulatta , Mesocricetus , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 2, Human/physiology , Respiratory System/virology , Temperature , Vaccines, Attenuated/immunology , Virus Replication
5.
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
6.
Virology ; 345(2): 492-501, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16300813

ABSTRACT

We evaluated the individual contributions of the three surface glycoproteins of human metapneumovirus (HMPV), namely the fusion F, attachment G, and small hydrophobic SH proteins, to the induction of serum HMPV-binding antibodies, serum HMPV-neutralizing antibodies, and protective immunity. Using reverse genetics, each HMPV protein was expressed individually from an added gene in recombinant human parainfluenza virus type 1 (rHPIV1) and used to infect hamsters once or twice by the intranasal route. The F protein was highly immunogenic and protective, whereas G and SH were only weakly or negligibly immunogenic and protective, respectively. Thus, in contrast to other paramyxoviruses, the HMPV attachment G protein is not a major neutralization or protective antigen. Also, although the SH protein of HMPV is a virion protein that is much larger than its counterparts in previously studied paramyxoviruses, it does not appear to be a significant neutralization or protective antigen.


Subject(s)
Antibodies, Viral/blood , Membrane Glycoproteins/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Viral Proteins/immunology , Animals , Base Sequence , Cricetinae , Genetic Vectors , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesocricetus , Metapneumovirus/genetics , Metapneumovirus/metabolism , Metapneumovirus/pathogenicity , Molecular Sequence Data , Neutralization Tests , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/metabolism , Paramyxoviridae Infections/prevention & control , Recombination, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Virology ; 352(1): 61-73, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16750233

ABSTRACT

Recombinant human parainfluenza virus type 1 (HPIV1) and mutants containing point and deletion (Delta) mutations in the P/C gene (r-CDelta10-15HNT553A, r-CR84G, r-CF170S and r-CDelta170), which have previously been evaluated as HPIV1 vaccine candidates, were evaluated for their effect on the type I interferon (IFN) response in vitro. HPIV1 wt infection inhibited the IFN response by inhibiting IFN regulatory factor-3 (IRF-3) activation and IFN production in A549 cells and IFN signaling in Vero cells. In contrast, r-CR84G, r-CF170S and r-CDelta170 were defective for inhibition of IRF-3 activation and IFN production and r-CF170S and r-CDelta170 did not inhibit IFN signaling. Thus, HPIV1 antagonizes the IFN response at both the level of induction and signaling, and antagonism at both levels was disrupted by mutations in the P/C gene. Because CF170S affects C and not P, the anti-IFN function can be attributed to the C proteins. These data, in the context of previous in vivo studies, suggest that the loss of antagonism of the IFN response at both the level of induction and signaling, observed with the P/C mutants, r-CF170S and r-CDelta170, was necessary for significant attenuation in African green monkeys (AGMs).


Subject(s)
Interferon Type I/metabolism , Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/pathogenicity , Phosphoproteins/genetics , Vaccines, Attenuated/immunology , Viral Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Humans , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Phosphoproteins/immunology , Point Mutation , Signal Transduction , Vaccines, Attenuated/genetics , Vero Cells , Viral Proteins/immunology
8.
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
9.
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
10.
Virology ; 297(1): 136-52, 2002 May 25.
Article in English | MEDLINE | ID: mdl-12083844

ABSTRACT

The level of replication and immunogenicity of recombinant parainfluenza virus type 3 (rHPIV3) bearing one, two, or three gene insertions expressing foreign protective antigens was examined. cDNA-derived recombinant HPIV3s bearing genes encoding the open reading frames (ORFs) of the hemagglutinin-neuraminidase (HN) of HPIV1, the HN of HPIV2, or the hemagglutinin (HA) of measles virus replicated efficiently in vitro, including the largest recombinant, which had three gene unit insertions and which was almost 23 kb in length, 50% longer than unmodified HPIV3. Several viruses were recovered from cDNAs whose genome length was not a multiple of six nucleotides and these contained nucleotide insertions that corrected the length to be a multiple of 6, confirming that the "rule of six" applies to HPIV3. Using a hemagglutination inhibition assay, we determined that the HPIV1 HN expressed by recombinant HPIV3 was incorporated into HPIV3 virions, whereas using this assay incorporation of the HPIV2 HN could not be detected. HPIV3 virions bearing HPIV1 HN were not neutralized by HPIV1 antiserum but were readily neutralized by antibodies to the HPIV3 HN or fusion protein (F). Viruses with inserts were restricted for replication in the respiratory tract of hamsters, and the level of restriction was a function of the total number of genes inserted, the nature of the insert, and the position of the inserted gene in the gene order. A single insert of HPIV2 HN or measles virus HA reduced the in vivo replication of rHPIV3 up to 25-fold, whereas the HPIV1 HN insert decreased replication almost 1000-fold. This indicates that the HPIV1 HN insert has an attenuating effect in addition to that of the extra gene insert itself, presumably because it is incorporated into the virus particle. Viruses containing two inserts were generally more attenuated than those with a single insert, and viruses with three inserts were over-attenuated for replication in hamsters. Inserts between the N and P genes were slightly more attenuating than those between the P and the M genes. A recombinant HPIV3 bearing both the HPIV1 and the HPIV2 HN genes (r1HN 2HN) was attenuated, immunogenic, and protected immunized hamsters from challenge with HPIV1, HPIV2, and HPIV3. Thus, it is possible to use a single HPIV vector expressing two foreign gene inserts to protect infants and young children from the severe lower respiratory tract disease caused by the three major human PIV pathogens.


Subject(s)
Genetic Vectors/physiology , Parainfluenza Virus 3, Human/physiology , Viral Vaccines , Animals , Antibodies, Viral/analysis , Cell Line , Cricetinae , Drug Design , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/immunology , HN Protein/genetics , HN Protein/immunology , HN Protein/metabolism , Hemagglutination Tests , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Hemagglutinins, Viral/metabolism , Measles virus/genetics , Measles virus/immunology , Measles virus/metabolism , Neutralization Tests , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/immunology , Reassortant Viruses , Viral Vaccines/immunology , Virus Replication
11.
J Virol ; 77(1): 270-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477832

ABSTRACT

Members of the Paramyxovirinae subfamily of the Paramyxoviridae family of viruses have the unusual requirement that the nucleotide length of the viral genome must be an even multiple of six in order for efficient RNA replication, and hence virus replication, to occur. Human parainfluenza virus type 2 (HPIV2) is the only member of the genus that has been reported to have a genome length that is not an even multiple of six, and it has also been recovered from a full-length antigenomic-sense cDNA that did not conform to the "rule of six." To reexamine the issue of nucleotide length in natural isolates of HPIV2, a complete consensus genomic sequence was determined for three HPIV2 strains: Greer, Vanderbilt/1994 (V94), and Vanderbilt/1998. Each of these strains was found to have a genome length of 15,654 nucleotides (nt), thus conforming in each case to the rule of six. To directly examine the requirement that the genomic length of HPIV2 be an even multiple of six, we constructed six full-length antigenomic HPIV2/V94 cDNAs that deviated from a polyhexameric length by 0 to 5 nt. Recombinant HPIV2s were readily recovered from all of the cDNAs, including those that did not conform to the rule of six. One recombinant HPIV2 isolate was completely sequenced for each of the nonpolyhexameric antigenomic cDNAs. These were found to contain small nucleotide insertions or deletions that conferred polyhexameric length to the recovered genome. Interestingly, almost all of the length corrections occurred within the hemagglutinin-neuraminidase and large polymerase genes or the intervening intergenic region and thus were proximal to the insert that caused the deviation from the rule of six. These results demonstrate, in the context of complete infectious virus, that HPIV2 has a strong and seemingly absolute requirement for a polyhexameric genome.


Subject(s)
Genome, Viral , Parainfluenza Virus 2, Human/genetics , Amino Acid Sequence , Animals , Base Sequence , Chlorocebus aethiops , Molecular Sequence Data , Mutation , Parainfluenza Virus 2, Human/growth & development , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells
12.
Virus Genes ; 24(1): 77-92, 2002.
Article in English | MEDLINE | ID: mdl-11928991

ABSTRACT

A complete consensus sequence was determined for the genomic RNA of human parainfluenza virus type 1 (HPIV1) strain Washington/20993/1964 (HPIV1 WASH/64), a clinical isolate that previously was shown to be virulent in adults. The sequence exhibited a high degree of relatedness to both Sendai virus, a PIV1 virus recovered from mice, and human PIV3 (HPIV3) with regard to cis-acting regulatory regions and protein-coding sequences. This consensus sequence was used to generate a full-length antigenomic cDNA and to recover a recombinant wild-type HPIV1 (rHPIV1). Interestingly, the rHPIV1 could be rescued from full-length antigenomic rHPIV1 cDNA using HPIV3 support plasmids, HPIV1 support plasmids, or a mixture thereof. The replication of rHPIV1 in vitro and in the respiratory tract of hamsters was similar to that of its biologically derived parent virus. The similar biological properties of rHPIV1 and HPIV1 WASH/64 in vitro and in vivo, together with the previous demonstration of the virulence of this specific isolate in humans, authenticates the rHPIV1 sequence as that of a wild-type virus. This rHPIV1 can now be used to study the biological properties of HPIV1 and as a substrate to introduce attenuating mutations for the generation of live-attenuated HPIV1 vaccine candidates.


Subject(s)
Genome, Viral , Parainfluenza Virus 1, Human/genetics , Paramyxoviridae/genetics , Base Sequence , Cell Line , Humans , Molecular Sequence Data , Parainfluenza Virus 1, Human/classification , Plasmids , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis
13.
Vaccine ; 20(13-14): 1846-52, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11906774

ABSTRACT

A recombinant live-attenuated chimeric human parainfluenza virus type 1 (HPIV1) candidate vaccine was previously generated by replacing the fusion (F) and hemagglutinin-neuraminidase (HN) glycoprotein open reading frames (ORFs) of the HPIV3 candidate vaccine, rHPIV3cp45, with those of wild-type HPIV1. Previously, this recombinant chimeric virus, designated rHPIV3-1cp45, exhibited a greater level of the temperature sensitivity of replication in vitro and a greater level of attenuation of replication in the respiratory tract of immunized hamsters when compared to its HPIV3cp45 parent virus. In the present study, rHPIV3-1cp45 was evaluated for its level of attenuation and efficacy in African green monkeys (Cercopithecus aethiops), a primate in which both HPIV1 and HPIV3 wild-type viruses replicate efficiently. The rHPIV3-1cp45 candidate vaccine was as restricted in replication in the upper and lower respiratory tract as its thoroughly characterized rHPIV3cp45 parent indicating that the attenuating mutations present in the rHPIV3cp45 backbone specified an appropriate level of attenuation of rHPIV3-1cp45 for primates. The level to which rHPIV3-1cp45 replicated in the respiratory tract of African green monkeys was also sufficient to induce a strong immune response to HPIV1 and provided protection against challenge with wild-type HPIV1. These results provide a basis for further evaluation of this HPIV1 candidate vaccine in humans.


Subject(s)
Parainfluenza Vaccines/pharmacology , Parainfluenza Virus 1, Human/immunology , Animals , Antibodies, Viral/biosynthesis , Chimera/genetics , Chimera/immunology , Chlorocebus aethiops , Humans , Mutation , Parainfluenza Vaccines/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Temperature , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/pharmacology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology , Virus Replication
14.
J Virol ; 78(4): 2017-28, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747566

ABSTRACT

Human parainfluenza virus type 1 (HPIV1) is a significant cause of respiratory tract disease in infants and young children for which a vaccine is needed. In the present study, we sought to attenuate HPIV1 by the importation of one or more known attenuating point mutations from heterologous paramyxoviruses into homologous sites in HPIV1. The introduced mutations were derived from three attenuated paramyxoviruses: (i) HPIV3cp45, a live-attenuated HPIV3 vaccine candidate containing multiple attenuating mutations; (ii) the respiratory syncytial virus cpts530 with an attenuating mutation in the L polymerase protein; and (iii) a murine PIV1 (MPIV1) attenuated by a mutation in the accessory C protein. Recombinant HPIV1 (rHPIV1) mutants bearing a single imported mutation in C, any of three different mutations in L, or a pair of mutations in F exhibited a 100-fold or greater reduction in replication in the upper or lower respiratory tract of hamsters. Both temperature-sensitive (ts) (mutations in the L and F proteins) and non-ts (the mutation in the C protein) attenuating mutations were identified. rHPIV1 mutants containing a combination of mutations in L were generated that were more attenuated than viruses bearing the individual mutations, showing that the systematic accretion of mutations can yield progressive increases in attenuation. Hamsters immunized with rHPIV1 mutants bearing one or two mutations developed neutralizing antibodies and were resistant to challenge with wild-type HPIV1. Thus, importation of attenuating mutations from heterologous viruses is an effective means for rapidly identifying mutations that attenuate HPIV1 and for generating live-attenuated HPIV1 vaccine candidates.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Paramyxoviridae/genetics , Respirovirus Infections/prevention & control , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Viral/blood , Base Sequence , Cricetinae , Humans , Mesocricetus , Molecular Sequence Data , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/pathogenicity , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae/immunology , Point Mutation , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Temperature , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Proteins/immunology
15.
J Virol ; 78(4): 2029-36, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747567

ABSTRACT

The Y942H and L992F temperature-sensitive (ts) and attenuating amino acid substitution mutations, previously identified in the L polymerase of the HPIV3cp45 vaccine candidate, were introduced into homologous positions of the L polymerase of recombinant human parainfluenza virus type 1 (rHPIV1). In rHPIV1, the Y942H mutation specified the ts phenotype in vitro and the attenuation (att) phenotype in hamsters, whereas the L992F mutation specified neither phenotype. Each of these codon mutations was generated by a single nucleotide substitution and therefore had the potential to readily revert to a codon specifying the wild-type amino acid residue. We introduced alternative amino acid assignments at codon 942 or 992 as a strategy to increase genetic stability and to generate mutants that exhibit a range of attenuation. Twenty-three recombinants with codon substitutions at position 942 or 992 of the L protein were viable. One highly ts and att mutant, the Y942A virus, which had a difference of three nucleotides from the codon encoding a wild-type tyrosine, also possessed a high level of genetic and phenotypic stability upon serial passage in vitro at restrictive temperatures compared to that of the parent Y942H virus, which possessed a single nucleotide substitution. We obtained mutants with substitutions at position 992 that, in contrast to the L992F virus, possessed the ts and att phenotypes. These findings identify the use of alternative codon substitution mutations as a method that can be used to generate candidate vaccine viruses with increased genetic stability and/or a modified level of attenuation.


Subject(s)
Amino Acid Substitution , Codon/genetics , DNA-Directed DNA Polymerase/genetics , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 1, Human/pathogenicity , Viral Proteins/genetics , Animals , Cell Line , Cricetinae , Humans , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/enzymology , Parainfluenza Virus 1, Human/genetics , Phenotype , Respiratory System/virology , Respirovirus Infections/virology , Temperature , Vaccines, Attenuated/genetics , Virus Replication
16.
J Virol ; 77(2): 1141-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502830

ABSTRACT

The Kansas strain of bovine parainfluenza virus type 3 (BPIV3) is 100- to 1,000-fold restricted in replication in the respiratory tracts of nonhuman primates compared to human PIV3 (HPIV3), an important pathogen of infants and young children. BPIV3 is also restricted in replication in human infants and children, yet it is immunogenic and is currently being evaluated in clinical trials as a vaccine candidate to protect against illness caused by HPIV3. We have examined the genetic basis for the host range attenuation phenotype of BPIV3 by exchanging each open reading frame (ORF) of a recombinant wild-type HPIV3 with the analogous ORF from BPIV3, with the caveats that the multiple ORFs of the P gene were exchanged as a single unit and that the HN and F genes were exchanged as a single unit. Recombinant chimeric bovine-human PIV3s were recovered from cDNA, and the levels of viral replication in vitro and in the respiratory tract of rhesus monkeys were determined. Recombinant chimeric HPIV3s bearing the BPIV3 N or P ORF were highly attenuated in the upper and lower respiratory tracts of monkeys, whereas those bearing the BPIV3 M or L ORF or the F and HN genes were only moderately attenuated. This indicates that the genetic determinants of the host range restriction of replication of BPIV3 for primates are polygenic, with the major determinants being the N and P ORFs. Monkeys immunized with these bovine-human chimeric viruses, including the more highly attenuated ones, developed higher levels of HPIV3 hemagglutination-inhibiting serum antibodies than did monkeys immunized with BPIV3 and were protected from challenge with wild-type HPIV3. Furthermore, host range determinants could be combined with attenuating point mutations to achieve an increased level of attenuation. Thus, chimeric recombinant bovine-human PIV3 viruses that manifest different levels of attenuation in rhesus monkeys are available for evaluation as vaccine candidates to protect infants from the severe lower respiratory tract disease caused by HPIV3.


Subject(s)
Parainfluenza Virus 3, Bovine/physiology , Virus Replication , Animals , Cell Line , Chimera , DNA, Complementary , Humans , Macaca mulatta , Open Reading Frames , Parainfluenza Virus 3, Bovine/genetics , Parainfluenza Virus 3, Bovine/immunology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/immunology , Temperature
17.
J Virol ; 78(13): 6927-37, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15194769

ABSTRACT

The growth properties and antigenic relatedness of the CAN98-75 (CAN75) and the CAN97-83 (CAN83) human metapneumovirus (HMPV) strains, which represent the two distinct HMPV genetic lineages and exhibit 5 and 63% amino acid divergence in the fusion (F) and attachment (G) proteins, respectively, were investigated in vitro and in rodents and nonhuman primates. Both strains replicated to high titers (> or =6.0 log(10)) in the upper respiratory tract of hamsters and to moderate titers (> or =3.6 log(10)) in the lower respiratory tract. The two lineages exhibited 48% antigenic relatedness based on reciprocal cross-neutralization assay with postinfection hamster sera, and infection with each strain provided a high level of resistance to reinfection with the homologous or heterologous strain. Hamsters immunized with a recombinant human parainfluenza virus type 1 expressing the fusion F protein of the CAN83 strain developed a serum antibody response that efficiently neutralized virus from both lineages and were protected from challenge with either HMPV strain. This result indicates that the HMPV F protein is a major antigenic determinant that mediates extensive cross-lineage neutralization and protection. Both HMPV strains replicated to low titers in the upper and lower respiratory tracts of rhesus macaques but induced high levels of HMPV-neutralizing antibodies in serum effective against both lineages. The level of HMPV replication in chimpanzees was moderately higher, and infected animals developed mild colds. HMPV replicated the most efficiently in the respiratory tracts of African green monkeys, and the infected animals developed a high level of HMPV serum-neutralizing antibodies (1:500 to 1:1,000) effective against both lineages. Reciprocal cross-neutralization assays in which postinfection sera from all three primate species were used indicated that CAN75 and CAN83 are 64 to 99% related antigenically. HMPV-infected chimpanzees and African green monkeys were highly protected from challenge with the heterologous HMPV strain. Taken together, the results from hamsters and nonhuman primates support the conclusion that the two HMPV genetic lineages are highly related antigenically and are not distinct antigenic subtypes or subgroups as defined by reciprocal cross-neutralization in vitro.


Subject(s)
Antibodies, Viral/blood , Antigenic Variation , Metapneumovirus/genetics , Viral Fusion Proteins/immunology , Virus Replication , Animals , Antibodies, Viral/immunology , Base Sequence , Cell Line , Cricetinae , Cross Reactions , Disease Models, Animal , Humans , Metapneumovirus/immunology , Metapneumovirus/physiology , Molecular Sequence Data , Neutralization Tests , Pan troglodytes , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Paramyxoviridae Infections/virology , Respiratory System/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics
18.
Virology ; 297(1): 153-60, 2002 May 25.
Article in English | MEDLINE | ID: mdl-12083845

ABSTRACT

Human parainfluenza virus type 1 (HPIV1), a major cause of croup in infants and young children, accounts for 6% of hospitalizations for pediatric respiratory tract disease. The antigenically related Sendai virus, referred to here as murine PIV1 (MPIV1), is being considered for use as a live-attenuated vaccine to protect against HPIV1 (J. L. Hurwitz, K. F. Soike, M. Y., Sangster, A. Portner, R. E. Sealy, D. H. Dawson, and C. Coleclough, 1997, Vaccine 15(5), 533-540) and also as a recombinant vaccine vector expressing antigens to protect against viral disease in humans. However, in the 1950s MPIV1 was reported to have been isolated from humans, suggesting that zoonotic transmission might have occurred. It is therefore important to examine the ability of MPIV1 to replicate in nonhuman primates, i.e., surrogate hosts for humans. In the present study the level of replication of MPIV1 and HPIV1 was compared in African green monkeys and chimpanzees. Surprisingly, MPIV1 replicated as efficiently as HPIV1 in the upper and lower respiratory tract of African green monkeys at doses of 10(4) and 10(6) and replicated only slightly less efficiently at both sites in chimpanzees. African green monkeys immunized with MPIV1 were highly resistant to subsequent challenge with HPIV1 even though MPIV1 did not induce a detectable HPIV1-neutralizing antibody response. The high level of replication of MPIV1 observed in the upper and lower respiratory tract of these primates suggests that MPIV1 likely would require significant attenuation before it could be given to humans as a vaccine against HPIV1 or as a vaccine vector. Its ability to efficiently replicate in nonhuman primates suggests that MPIV1 lacks a significant host range restriction in primates and could theoretically cause zoonotic disease in humans.


Subject(s)
Chlorocebus aethiops/virology , Disease Models, Animal , Pan troglodytes/virology , Parainfluenza Virus 1, Human/physiology , Respiratory System/virology , Sendai virus/physiology , Animals , Antibodies, Viral/analysis , Neutralization Tests , Parainfluenza Virus 1, Human/immunology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Species Specificity , Vaccines, Attenuated , Viral Vaccines/administration & dosage , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL