Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
J Immunol ; 167(7): 3910-8, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564809

ABSTRACT

A respiratory syncytial virus (RSV) vaccine will need to be administered by 1 mo of age to protect young infants; therefore, it will need to be effective in the presence of maternally acquired RSV Abs. In the present study, the immunogenicity and efficacy of two live attenuated RSV vaccine candidates of different level of attenuation were evaluated in mice passively immunized with varying quantities of RSV Abs. The replication of the RSV vaccines was suppressed in the lower, but not the upper, respiratory tract of the passively immunized mice. Immunization with either vaccine candidate was highly efficacious against challenge with wild-type RSV in both passively immunized and control mice. Nonetheless, a high level of immunity was seen even in passively/actively immunized animals that failed to develop a humoral immune response, suggesting that T cells mediated the immunity. Depletion of CD4+ and CD8+ T cells in passively/actively immunized and control animals at the time of challenge with wild-type RSV demonstrated that CD4+ and CD8+ T cells made significant independent contributions to the restriction of replication of RSV challenge virus in both the upper and lower respiratory tracts. Although passively acquired serum RSV Abs suppressed the primary systemic and mucosal Ab responses of IgM, IgG, and IgA isotypes, B lymphocytes were nevertheless primed for robust secondary Ab responses. Thus, immunity mediated by CD4+ and CD8+ T cells and Abs can be readily induced in mice by live RSV vaccine candidates in the presence of physiologic levels of RSV neutralizing Abs.


Subject(s)
Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes/immunology , Animals , Cell Line , Immunity, Mucosal , Immunization, Passive , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Vaccines, Attenuated/immunology , Virus Replication
2.
J Virol ; 75(20): 9731-40, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559806

ABSTRACT

A recombinant live attenuated dengue virus type 4 (DEN4) vaccine candidate, 2ADelta30, was found previously to be generally well tolerated in humans, but a rash and an elevation of liver enzymes in the serum occurred in some vaccinees. 2ADelta30, a non-temperature-sensitive (non-ts) virus, contains a 30-nucleotide deletion (Delta30) in the 3' untranslated region (UTR) of the viral genome. In the present study, chemical mutagenesis of DEN4 was utilized to generate attenuating mutations which may be useful in further attenuation of the 2ADelta30 candidate vaccine. Wild-type DEN4 2A virus was grown in Vero cells in the presence of 5-fluorouracil, and a panel of 1,248 clones were isolated. Twenty ts mutant viruses were identified that were ts in both simian Vero and human liver HuH-7 cells (n = 13) or only in HuH-7 cells (n = 7). Each of the 20 ts mutant viruses possessed an attenuation phenotype, as indicated by restricted replication in the brains of 7-day-old mice. The complete nucleotide sequence of the 20 ts mutant viruses identified nucleotide substitutions in structural and nonstructural genes as well as in the 5' and 3' UTRs, with more than one change occurring, in general, per mutant virus. A ts mutation in the NS3 protein (nucleotide position 4995) was introduced into a recombinant DEN4 virus possessing the Delta30 deletion, thereby creating rDEN4Delta30-4995, a recombinant virus which is ts and more attenuated than rDEN4Delta30 virus in the brains of mice. We are assembling a menu of attenuating mutations that should be useful in generating satisfactorily attenuated recombinant dengue vaccine viruses and in increasing our understanding of the pathogenesis of dengue virus.


Subject(s)
Dengue Virus/drug effects , Dengue/virology , Fluorouracil/pharmacology , Mutagens/pharmacology , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Animals, Suckling , Brain/virology , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Dengue Virus/genetics , Dengue Virus/physiology , Genes, Viral , Genome, Viral , Humans , Mice , Molecular Sequence Data , RNA Helicases , Recombination, Genetic , Serine Endopeptidases , Temperature , Vaccines, Attenuated/genetics , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Vaccines/genetics , Virus Replication
3.
J Virol ; 73(12): 9773-80, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10559287

ABSTRACT

Human respiratory syncytial virus (RSV) exists as two antigenic subgroups, A and B, both of which should be represented in a vaccine. The F and G glycoproteins are the major neutralization and protective antigens, and the G protein in particular is highly divergent between the subgroups. The existing system for reverse genetics is based on the A2 strain of RSV subgroup A, and most efforts to develop a live attenuated RSV vaccine have focused on strain A2 or other subgroup A viruses. In the present study, the development of a live attenuated subgroup B component was expedited by the replacement of the F and G glycoproteins of recombinant A2 virus with their counterparts from the RSV subgroup B strain B1. This gene replacement was initially done for wild-type (wt) recombinant A2 virus to create a wt AB chimeric virus and then for a series of A2 derivatives which contain various combinations of A2-derived attenuating mutations located in genes other than F and G. The wt AB virus replicated in cell culture with an efficiency which was comparable to that of the wt A2 and B1 parents. AB viruses containing temperature-sensitive mutations in the A2 background exhibited levels of temperature sensitivity in vitro which were similar to those of A2 viruses bearing the same mutations. In chimpanzees, the replication of the wt AB chimera was intermediate between that of the A2 and B1 wt viruses and was accompanied by moderate rhinorrhea, as previously seen in this species. An AB chimeric virus, rABcp248/404/1030, which was constructed to contain a mixture of attenuating mutations derived from two different biologically attenuated A2 viruses, was highly attenuated in both the upper and lower respiratory tracts of chimpanzees. This attenuated AB chimeric virus was immunogenic and conferred a high level of resistance on chimpanzees to challenge with wt AB virus. The rABcp248/404/1030 chimeric virus is a promising vaccine candidate for RSV subgroup B and will be evaluated next in humans. Furthermore, these results suggest that additional attenuating mutations derived from strain A2 can be inserted into the A2 background of the recombinant chimeric AB virus as necessary to modify the attenuation phenotype in a reasonably predictable manner to achieve an optimal balance between attenuation and immunogenicity in a virus bearing the subgroup B antigenic determinants.


Subject(s)
HN Protein , Respiratory Syncytial Virus, Human/genetics , Vaccines, Synthetic/genetics , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Animals , Chlorocebus aethiops , Humans , Pan troglodytes , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Syncytial Virus, Human/physiology , Sigmodontinae , Temperature , Tumor Cells, Cultured , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Vero Cells , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Virus Replication
4.
Vaccine ; 17(11-12): 1416-24, 1999 Mar 17.
Article in English | MEDLINE | ID: mdl-10195777

ABSTRACT

cpts530/1009 is a live-attenuated, temperature-sensitive (ts) RSV vaccine candidate that was shown previously to be attenuated for seronegative humans. It was generated by two rounds of chemical mutagenesis: first, a partially attenuated, cold-passaged (cp), non-ts RSV mutant (cpRSV) was mutagenized to yield the ts derivative cpts530, and then cpts530 was mutagenized to yield cpts530/1009, which is more ts. Previous nucleotide (nt) sequence analysis of cpts530 showed that it has a single nt change compared to cpRSV that results in an amino acid substitution at residue 521 in the L protein. Reverse genetics confirmed that this mutation is responsible for the ts phenotype of cpts530. Here, determination of the complete 15,222-nt sequence of cpts530/ 1009 identified a single change compared to cpts530, namely a point mutation at nt 12002, which results in a methionine-tovaline substitution at amino acid 1169 in the L protein. The contribution of the 1009 mutation to the level of temperature sensitivity and attenuation exhibited by cpts530/1009 was evaluated by its introduction alone or with the 530 and cp mutations into the full-length cDNA clone of wild-type (wt) RSV. Subsequent analysis of infectious viruses recovered from the mutant cDNAs indicated that (i) the 1009 mutation indeed was a ts mutation and the level of temperature sensitivity specified by the 1009 mutation was less than that specified by the 530 mutation, (ii) the 530 and 1009 mutations each contributed to attenuation in the upper respiratory tract of mice and their effects were additive, (iii) viruses bearing the 1009 mutation were more attenuated in the lower respiratory tract of mice than viruses bearing the 530 mutation and (iv) the combination of the 530 and 1009 mutations in the cpRSV background resulted in the same level of temperature sensitivity and attenuation in mice as that observed for the biologically-derived cpts530/1009 mutant. These data show that the genetic basis of the attenuation and temperature sensitivity of the cpts530/1009 candidate vaccine virus is the sum of the contributions of seven identified amino acid substitutions, i.e. the 5 cpRSV mutations, the 530 mutation and the 1009 mutation.


Subject(s)
HN Protein , Vaccines, Attenuated , Viral Proteins/immunology , Viral Vaccines , Amino Acid Substitution , Animals , Chlorocebus aethiops , Humans , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Phenotype , RNA, Viral/chemistry , Respiratory Syncytial Viruses , Sequence Analysis , Structure-Activity Relationship , Temperature , Transfection , Vero Cells , Viral Envelope Proteins , Viral Proteins/chemistry
5.
J Virol ; 73(4): 3438-42, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074199

ABSTRACT

The NS2 and SH genes of respiratory syncytial virus (RSV) have been separately deleted from a recombinant wild-type RSV strain, A2 (M. N. Teng and P. L. Collins, J. Virol. 73:466-473, 1998; A. Bukreyev et al., J. Virol. 71:8973-8982, 1997; and this study). The resulting viruses, designated rA2DeltaNS2 and rA2DeltaSH, were administered to chimpanzees to evaluate their levels of attenuation and immunogenicity. Recombinant virus rA2DeltaNS2 replicated to moderate levels in the upper respiratory tract, was highly attenuated in the lower respiratory tract, and induced significant resistance to challenge with wild-type RSV. The replication of rA2DeltaSH virus was only moderately reduced in the lower, but not the upper, respiratory tract. However, chimpanzees infected with either virus developed significantly less rhinorrhea than those infected with wild-type RSV. These findings demonstrate that a recombinant RSV mutant lacking either the NS2 or SH gene is attenuated and indicate that these deletions may be useful as attenuating mutations in new, live recombinant RSV vaccine candidates for both pediatric and elderly populations. The DeltaSH mutation was incorporated into a recombinant form of the cpts248/404 vaccine candidate, was evaluated for safety in seronegative chimpanzees, and can now be evaluated as a vaccine for humans.


Subject(s)
Genes, Viral , HN Protein , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Animals , Base Sequence , DNA, Recombinant , Gene Deletion , Molecular Sequence Data , Pan troglodytes , Respiratory Syncytial Viruses/pathogenicity , Viral Envelope Proteins , Virulence/genetics
6.
J Virol ; 73(2): 871-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9882287

ABSTRACT

Respiratory syncytial virus (RSV) cpts530/1030 is an attenuated, temperature-sensitive subgroup A vaccine candidate derived previously from cold-passaged RSV (cpRSV) by two sequential rounds of chemical mutagenesis and biological selection. Here, cpts530/1030 was shown to be highly attenuated in the upper and lower respiratory tracts of seronegative chimpanzees. However, evaluation in seropositive children showed that it retains sufficient replicative capacity and virulence to preclude its direct use as a live attenuated vaccine. Nucleotide sequence analysis of the genome of cpts530/1030 showed that it had acquired two nucleotide substitutions (compared to its cpts530 parent), both of which were in the L gene: a silent mutation at nucleotide position 8821 (amino acid 108) and a missense mutation at nucleotide position 12458 resulting in a tyrosine-to-asparagine change at amino acid 1321, herein referred to as the 1030 mutation. It also contained the previously identified 530 missense mutation at nucleotide 10060 in the L gene. The genetic basis of attenuation of cpts530/1030 was defined by the introduction of the 530 and 1030 mutations into a cDNA clone of cpRSV, from which recombinant RSV was derived and analyzed to determine the contribution of each mutation to the temperature sensitivity (ts) and attenuation (att) phenotypes of cpts530/1030. The 530 mutation, derived from cpts530, was previously shown to be responsible for the ts and att phenotypes of that virus. In the present study, the 1030 mutation was shown to be responsible for the increased temperature sensitivity of cpts530/1030. In addition, the 1030 mutation was shown to be responsible for the increased level of attenuation of cpts530/1030 in the upper and lower respiratory tracts of mice. The 530 and 1030 mutations were additive in their effects on the ts and att phenotypes. It was possible to introduce the 1030 mutation, but not the 530 mutation, into an attenuated vaccine candidate with residual reactogenicity in very young infants, namely, cpts248/404, by use of reverse genetics. The inability to introduce the 530 mutation into the cpts248/404 virus was shown to be due to its incompatibility with the 248 missense mutation at the level of L protein function. The resulting rA2cp248/404/1030 mutant virus was more temperature sensitive and more attenuated than the cpts248/404 parent virus, making it a promising new RSV vaccine candidate created by use of reverse genetics to improve upon an existing vaccine virus.


Subject(s)
HN Protein , Mutation, Missense , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Child, Preschool , Chlorocebus aethiops , Double-Blind Method , Humans , Infant , Mice , Mice, Inbred BALB C , Pan troglodytes , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , Sequence Analysis, DNA , Temperature , Tumor Cells, Cultured , Vaccines, Attenuated , Vero Cells , Viral Envelope Proteins , Viral Vaccines/genetics , Virus Replication
7.
Virology ; 247(2): 232-9, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9705916

ABSTRACT

Respiratory syncytial virus (RSV) cpts248/404 is a live-attenuated, temperature-sensitive (ts) vaccine candidate derived from cole-passaged cpRSV by two rounds of chemical mutagenesis and biological selection. Previous sequence analysis showed that these two steps introduced three single nucleotide substitutions into the cpRSV parent. Two of these occurred with the coding sequence for the L protein, and each resulted in a single amino acid substitution: Gin-831-Leu (248 mutation) and Asp-1183-Glu (404-L mutation). The third mutation resulted in a nucleotide substitution at position 9 of the c/s-acting gene start signal of the M2 gene (404-M2 mutation). In the present study, the genetic basis of attenuation of cpts248/404 was defined by the introduction of each of these mutations (singly or in combination) into a full-length cDNA clone of cpRSV. Recombinant RSV derived from each mutant cDNA was analyzed to determine the contribution of each mutation to the ts and attenuation phenotypes of the virus. This analysis showed that the 248 mutation specifies a significant reduction of plaque formation at 38 degrees and is responsible for an intermediate level of attenuation in mice. In contrast, the 404-L mutation did not contribute to the ts or attenuation phenotype alone or in combination with other mutations and is thus an incidental change. unexpectedly, the 404-M2 mutation alone specified complete restriction of plaque formation at 37 degrees C an a high level of attenuation in mice. This indicates that the level of temperature sensitivity and attenuation of cpts248/404 can be attributed primarily to the 404-M2 mutation. Thus the cpts248/404 virus contains a set of ts and non-ts attenuating mutations, which likely accounts for its genetic stability. The recombinant version of this virus, rA2cp248/404, was phenotypically indistinguishable from cpts248/404 and represents a background into which additional mutations can be introduced as needed to obtain the desired level of attenuation for successful immunization of the very young human infant.


Subject(s)
Antigens, Viral/genetics , HN Protein , Point Mutation , Respiratory Syncytial Viruses/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Animals , Antigens, Viral/immunology , Cell Line , Chickens , DNA, Complementary , Humans , Mice , Mice, Inbred BALB C , Phenotype , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/physiology , Temperature , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Envelope Proteins , Viral Vaccines/immunology , Virus Replication
8.
J Virol ; 72(5): 4467-71, 1998 May.
Article in English | MEDLINE | ID: mdl-9557743

ABSTRACT

A set of five missense mutations previously identified by nucleotide sequence analysis of subgroup A cold-passaged (cp) respiratory syncytial virus (RSV) has been introduced into a recombinant wild-type strain of RSV. This recombinant virus, designated rA2cp, appears to replicate less efficiently in the upper and lower respiratory tracts of seronegative chimpanzees than either biologically derived or recombinant wild-type RSV. Infection with rA2cp also resulted in significantly less rhinorrhea and cough than infection with wild-type RSV. These findings confirm the role of the cp mutations in attenuation of RSV and identify their usefulness for inclusion in future live attenuated recombinant RSV vaccine candidates.


Subject(s)
Mutation , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/pathogenicity , Animals , Base Sequence , Cell Line , DNA, Viral , Humans , Molecular Sequence Data , Pan troglodytes , Recombination, Genetic
9.
Virology ; 252(2): 373-5, 1998 Dec 20.
Article in English | MEDLINE | ID: mdl-9878616

ABSTRACT

A recombinant human antibody fragment, designated RSV Fab 19, efficiently neutralizes respiratory syncytial virus (RSV). Here we report the results of our sequence analysis of antibody escape mutants that identified F glycoprotein amino acids critical for binding of human or murine RSV F-neutralizing antibodies.


Subject(s)
Antibodies, Monoclonal , Epitopes/analysis , Immunoglobulin Fab Fragments , Respiratory Syncytial Viruses/immunology , Viral Fusion Proteins/immunology , Amino Acid Substitution , Animals , Cell Line , Humans , Neutralization Tests , Point Mutation , Recombinant Proteins , Respiratory Syncytial Viruses/genetics , Tumor Cells, Cultured , Viral Fusion Proteins/analysis , Viral Fusion Proteins/genetics
10.
Virology ; 225(2): 419-22, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8918930

ABSTRACT

The complete nucleotide sequence of the RSV cpts-248/404 live attenuated vaccine candidate was determined from cloned cDNA and was compared to that of the RSV A2/HEK7 wild-type, cold-passaged cp-RSV, and cpts-248 virus, which constitute the series of progenitor viruses. RSV cpts-248/404 is more attenuated and more temperature sensitive (ts) (shut-off temperature 36 degrees) than its cpts-248 parent virus (shut-off temperature 38 degrees) and is currently being evaluated in phase I clinical trials in humans. Our ultimate goal is to identify the genetic basis for the host range attenuation phenotype exhibited by cp-RSV (i.e., efficient replication in tissue culture but decreased replication in chimpanzees and humans) and for the ts and attenuation phenotypes of its chemically mutagenized derivatives, cpts-248 and cpts-248/404. Compared with its cpts-248 parent, the cpts-248/404 virus possesses an amino acid change in the polymerase (L) protein and a single nucleotide substitution in the M2 gene start sequence. In total, the cpts-248/404 mutant differs from its wild-type RSV A2/HEK7 progenitor in seven amino acids [four in the polymerase (L) protein, two in the fusion (F) glycoprotein, and one in the (N) nucleoprotein] and one nucleotide difference in the M2 gene start sequence. Heterogeneity at nucleotide position 4 (G or C, negative sense, compared to G in the RSV A2/HEK7 progenitor) in the leader region of vRNA developed during passage of the cpts-248/404 in tissue culture. Biologically cloned derivatives of RSV cpts-248/404 virus that differed at position 4 possessed the same level of temperature sensitivity and exhibited the same level of replication in the upper and lower respiratory tract of mice, suggesting that heterogeneity at this position is not clinically relevant. The determination of the nucleotide sequence of the cpts-248/404 virus will allow evaluation of the stability of the eight mutations that are associated with the attenuation phenotype during vaccine production and following replication in humans.


Subject(s)
RNA, Viral/genetics , Respiratory Syncytial Viruses/genetics , Viral Vaccines , Animals , Cold Temperature , Humans , Mice , Molecular Sequence Data , Mutation , Sequence Analysis , Temperature
11.
J Infect Dis ; 173(4): 829-39, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8603960

ABSTRACT

Optimal immunization of neonates against disease caused by respiratory syncytial virus (RSV) probably will require multiple doses of a vaccine containing viruses of both subgroups A and B. Live subgroup B RSV mutants were generated containing multiple attenuating mutations, ts (temperature-sensitive) and non-ts (host range), that were introduced by prolonged passage in cell culture or by chemical mutagenesis. The cold-passaged (cp)-52 mutant was restricted in replication compared to wild type virus in rodents and nonhuman primates. In addition, the attenuation phenotype of cp-52 was stable after prolonged replication in immunosuppressed rodents. One or two ts mutations were then introduced into the cp-52 mutant to generate additional candidate vaccine strains that were more attenuated in vivo than the cp-52 parental virus. Tests in humans are being done to determine if one or more of the RSV B-1 mutants exhibit a satisfactory balance between attenuation and immunogenicity.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Chlorocebus aethiops , Mutagenesis , Respiratory Syncytial Virus Infections/immunology , Serotyping , Sigmodontinae , Virus Replication
12.
Virus Genes ; 13(3): 269-73, 1996.
Article in English | MEDLINE | ID: mdl-9035372

ABSTRACT

A cold-passaged (cp) temperature-sensitive (ts) mutant of human respiratory syncytial virus designated RSV cpts-248 was previously derived by random chemical mutagenesis of the non-ts mutant cp-RSV that possesses one or more host range mutations. We previously demonstrated in rodents and seronegative chimpanzees that the cpts-248 virus is more attenuated than cp-RSV and is more stable genetically than previously isolated RSV ts mutants. In the present study, we determined that the acquisition of the ts phenotype and the increased attenuation of the cpts-248 virus are associated with a single nucleotide substitution at nucleotide 10,989 that results in a change in the coding region (amino acid position 831) of the polymerase gene. The identification of this attenuating ts mutation is important because cpts-248 was used as the parent virus for the generation of a number of further attenuated mutants that are currently being evaluated as candidate vaccine strains in clinical trials in infants. Furthermore, technology now exists to rationally design new vaccine candidates by incorporating multiple attenuating mutations, such as the one identified here, into infectious viruses that are genetically stable and appropriately attenuated.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Respiratory Syncytial Virus, Human/enzymology , Viral Proteins/genetics , Viral Vaccines/genetics , Animals , Base Sequence , Chlorocebus aethiops , Cold Temperature , DNA, Viral , Genes, Viral , Humans , Molecular Sequence Data , Mutagenesis , Phenotype , Respiratory Syncytial Virus, Human/genetics , Vero Cells
13.
Virology ; 208(2): 478-84, 1995 Apr 20.
Article in English | MEDLINE | ID: mdl-7747420

ABSTRACT

In previous studies, a mutant (cp-RSV) of the RSV A2 strain derived from 52 serial cold passages in bovine embryonic tissue culture was highly attenuated in seropositive adults and children but caused upper respiratory tract disease in seronegative infants. We investigated the genetic basis for this attenuation phenotype by comparing the complete genomic RNA sequence of this virus with the published sequence of strain A2 as well as with that of its unattenuated wild-type parent (HEK-7) virus. RNA was extracted from virions grown in tissue culture, reverse transcribed, amplified by the polymerase chain reaction (PCR), cloned, and sequenced. Changes from the published A2 wild-type sequence were confirmed on independently derived cDNA clones and by direct sequencing of PCR fragments. The HEK-7 parent virus was then analyzed at these positions by direct sequencing of PCR fragments. Fifteen nucleotide differences between the published A2 wild-type virus and cp-RSV were found. None appeared to involve cis-acting RNA sequences. Of the 15 nucleotide differences, only 1 occurred outside a translational open reading frame (ORF), and 2 which did occur within ORFs were silent at the amino acid level. The remaining 12 nucleotide differences encoded amino acid changes. All 3 of the mutations which were silent at the amino acid level, and 8 of the 12 which resulted in amino acid differences, were also present in the HEK-7 parent virus and therefore were not changes acquired during the cold passages. Thus, the remaining 4 nucleotide differences and the attendant 4 amino acid changes are associated with the attenuation phenotype of the cp-RSV. Two of the changes occur in the F gene and two in the L gene. These results confirm the previously described RSV genomic sequence, provide the first sequence of a live attenuated RSV vaccine strain, provide the first sequence of an RSV strain which has been evaluated in chimpanzees and humans, and indicate that attenuation in humans of a pneumovirus can be associated with a relatively small number of nucleotide and amino acid changes.


Subject(s)
Genes, Viral/genetics , HN Protein , Mutation/genetics , Respiratory Syncytial Viruses/genetics , Viral Vaccines/genetics , Amino Acids/genetics , Cloning, Molecular , Cold Temperature , Genetic Variation/genetics , Genome, Viral , Humans , Open Reading Frames/genetics , Phenotype , RNA, Viral/genetics , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Sequence Analysis, DNA , Serial Passage , Vaccines, Attenuated/genetics , Viral Envelope Proteins , Viral Proteins/biosynthesis , Viral Proteins/genetics
14.
J Virol ; 68(8): 5321-5, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035532

ABSTRACT

In previous studies, children immunized with a formalin-inactivated respiratory syncytial virus vaccine (FI-RSV) developed severe pulmonary disease with greater frequency than did controls during subsequent natural RSV infection. In earlier efforts to develop an animal model for this phenomenon, extensive pulmonary histopathology developed in FI-RSV-immunized cotton rats and mice subsequently challenged with RSV. In mice, depletion of CD4+ T cells at the time of RSV challenge completely abrogated this histopathology. Furthermore, the predominant cytokine mRNA present in lungs of FI-RSV-immunized mice during subsequent infection with RSV was that characteristically secreted by Th2 T cells, namely interleukin-4 (IL-4). In the present studies, we sought to determine the relative contributions of gamma interferon (IFN-gamma), IL-2, IL-4, and IL-10 to the lymphocytic infiltration into the lungs observed following RSV challenge of mice previously immunized with FI-RSV. Mice previously immunized with FI-RSV or infected with RSV were depleted of IFN-gamma, IL-2, IL-4, or IL-10 immediately before RSV challenge, and the magnitude of inflammatory cell infiltration around bronchioles and pulmonary blood vessels was quantified. The phenomenon of pulmonary-histopathology potentiation by FI-RSV was reproduced in the present study, thereby allowing us to investigate the effect of cytokine depletion on the process. Simultaneous depletion of both IL-4 and IL-10 completely abrogated pulmonary histopathology in FI-RSV-immunized mice. Depletion of IL-4 alone significantly reduced bronchiolar, though not perivascular, histopathology. Depletion of IL-10 alone had no effect. Depletion of IFN-gamma, IL-2, or both together had no effect on the observed histopathology. These data indicate that FI-RSV immunization primes for a Th2-, IL-4-, and IL-10-dependent inflammatory response to subsequent RSV infection. It is possible that this process played a role in enhanced disease observed in infants and children immunized with FI-RSV.


Subject(s)
Interleukin-10/physiology , Interleukin-4/physiology , Lung/pathology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/pathogenicity , Animals , Formaldehyde/pharmacology , Immunization , Interferon-gamma/physiology , Interleukin-2/physiology , Lung/microbiology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/immunology , Viral Vaccines
15.
J Virol ; 67(2): 1044-9, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8419638

ABSTRACT

Previous studies demonstrated that the pulmonary resistance to respiratory syncytial virus (RSV) challenge induced by immunization with a recombinant vaccinia virus expressing the M2 protein of RSV (vac-M2) was significantly greater 9 days after immunization than at 28 days and was mediated predominantly by CD8+ T cells. In this study, we have extended these findings and sought to determine whether the level of CD8+ cytotoxic T-lymphocyte (CTL) activity measured in vitro correlates with the resistance to RSV challenge in vivo. Three lines of evidence documented an association between the presence of pulmonary CTL activity and resistance to RSV challenge. First, vac-M2 immunization induced pulmonary CD8+ CTL activity and pulmonary resistance to RSV infection in BALB/c (H-2d) mice, whereas significant levels of pulmonary CTL activity and resistance to RSV infection were not seen in BALB.K (H-2k) or BALB.B (H-2b) mice. Second, pulmonary CD8+ CTL activity was not induced by infection with other vaccinia virus-RSV recombinants that did not induce resistance to RSV challenge. Third, the peak of pulmonary CTL activity correlated with the peak of resistance to RSV replication (day 6), with little resistance being observed 45 days after immunization. An accelerated clearance of virus was not observed when mice were challenged with RSV 45 days after immunization with vac-M2. The results indicate that resistance to RSV induced by immunization with vac-M2 is mainly mediated by primary pulmonary CTLs and that this resistance decreases to very low levels within 2 months following immunization. The implications for inclusion of CTL epitopes into RSV vaccines are discussed in the context of these observations.


Subject(s)
CD8 Antigens/immunology , HN Protein , Respiratory Syncytial Viruses/immunology , Respirovirus Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/immunology , Animals , Female , H-2 Antigens , Immunity, Innate , Immunotherapy, Active , Major Histocompatibility Complex , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Recombinant Proteins/immunology , Respirovirus Infections/therapy , Vaccinia virus/genetics , Viral Envelope Proteins
16.
J Virol ; 66(12): 7444-51, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1433525

ABSTRACT

In previous studies, it was observed that children immunized with a formalin-inactivated respiratory syncytial virus vaccine (FI-RSV) developed severe pulmonary disease with greater frequency during subsequent natural RSV infection than did controls. During earlier efforts to develop an animal model of this phenomenon, enhanced pulmonary histopathology was observed after intranasal RSV challenge of FI-RSV-immunized cotton rats. Progress in understanding the immunologic basis for these observations has been hampered by the lack of reagents useful in manipulating the immune response of the cotton rat. This problem prompted us to reinvestigate the characteristics of immunity to RSV in the mouse. In the present studies, extensive pulmonary histopathology was observed in FI-RSV-immunized or RSV-infected BALB/c mice upon RSV challenge, and studies to determine the relative contributions of CD4+ or CD8+ T cells to this process were undertaken. Mice previously immunized with FI-RSV or infected with RSV were depleted of CD4+, CD8+, or both T-cell subsets immediately prior to RSV challenge, and the magnitude of inflammatory cell infiltration around bronchioles and pulmonary blood vessels and into alveolar spaces was quantified. The magnitude of infiltration at each anatomic site in previously FI-RSV-immunized or RSV-infected, nondepleted animals was similar, indicating that this is not a relevant model for enhanced disease. However, the effect of T-cell subset depletion on pulmonary histopathology following RSV challenge was very different between the two groups. Depletion of CD4+ T cells completely abrogated pulmonary histopathology in FI-RSV-immunized mice, whereas it had a much smaller effect on mice previously infected with RSV. FI-RSV-immunized or RSV-infected animals depleted of CD8+ T cells had only a modest reduction of pulmonary histopathology. In addition, RSV infection induced high levels of major histocompatibility complex class I-restricted cytotoxic T-cell activity, whereas FI-RSV immunization induced a low level. These data indicate that immunization with FI-RSV induces a cellular immune response different from that induced by RSV infection, which likely played a role in enhanced disease observed in infants and children.


Subject(s)
Antibodies, Viral/analysis , CD4 Antigens/immunology , Lung/pathology , Lymphocyte Depletion , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Animals , Female , Formaldehyde , H-2 Antigens/immunology , Haplotypes , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Respiratory Syncytial Viruses/physiology , Spleen/immunology , Virus Replication
17.
J Virol ; 66(2): 1277-81, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1731105

ABSTRACT

It was previously demonstrated that the vaccinia virus recombinants expressing the respiratory syncytial virus (RSV) F, G, or M2 (also designated as 22K) protein (Vac-F, Vac-G, or Vac-M2, respectively) induced almost complete resistance to RSV challenge in BALB/c mice. In the present study, we sought to identify the humoral and/or cellular mediators of this resistance. Mice were immunized by infection with a single recombinant vaccinia virus and were subsequently given a monoclonal antibody directed against CD4+ or CD8+ T cells or gamma interferon (IFN-gamma) to cause depletion of effector T cells or IFN-gamma, respectively, at the time of RSV challenge (10 days after immunization). Mice immunized with Vac-F or Vac-G were completely or almost completely resistant to RSV challenge after depletion of both CD4+ and CD8+ T cells prior to challenge, indicating that these cells were not required at the time of virus challenge for expression of resistance to RSV infection induced by the recombinants. In contrast, the high level of protection of mice immunized with Vac-M2 was completely abrogated by depletion of CD8+ T cells, whereas depletion of CD4+ T cells or IFN-gamma resulted in intermediate levels of resistance. These results demonstrate that antibodies are sufficient to mediate the resistance to RSV induced by the F and G proteins, whereas the resistance induced by the M2 protein is mediated primarily by CD8+ T cells, with CD4+ T cells and IFN-gamma also contributing to resistance.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , CD8 Antigens/immunology , HN Protein , Immunization , Respiratory Syncytial Viruses/immunology , T-Lymphocyte Subsets/immunology , Vaccinia virus/immunology , Viral Proteins , Viral Vaccines , Animals , Antigens, Viral/genetics , CD4 Antigens/immunology , CD4-CD8 Ratio , Cell Line , Female , Humans , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Recombination, Genetic , Respiratory Syncytial Viruses/genetics , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Vaccinia virus/genetics , Viral Envelope Proteins
18.
Vaccine ; 10(7): 475-84, 1992.
Article in English | MEDLINE | ID: mdl-1609551

ABSTRACT

In studies conducted in the 1960s, children previously immunized with a formalin-inactivated respiratory syncytial virus (RSV) vaccine (FI-RSV) developed a greater incidence and severity of pulmonary disease during subsequent natural RSV infection than did controls. It was previously shown that cotton rats immunized with FI-RSV or immunoaffinity-purified fusion (F) glycoprotein developed enhanced pulmonary histopathology following intranasal challenge with RSV. In the present studies, various forms of immunization, including parenteral inoculation of an immunoaffinity-purified F glycoprotein or a chimeric FG glycoprotein produced in insect cells using a baculovirus vector (Bac-FG), intradermal infection with a vaccinia-F recombinant (Vac-F) or intranasal infection with an adenovirus-F recombinant (Ad-F) or RSV, were compared for immunogenicity, efficacy and ability to alter the host so that enhanced pulmonary histopathology developed during RSV infection 3 months after immunization. Immunization of cotton rats with F glycoprotein, Bac-FG, Vac-F, Ad-F or infection with RSV induced high levels of ELISA-F antibodies, but the antibodies induced by purified F glycoprotein of Bac-FG had low levels of neutralizing activity. Immunization with Vac-F or Ad-F, or infection with RSV induced a high level of resistance to pulmonary RSV replication, whereas animals immunized with Bac-FG or FI-RSV were only partially protected. Following RSV challenge, animals immunized with purified F glycoprotein or Bac-FG developed the highest levels of bronchiolar and alveolar histopathology, those immunized with FI-RSV had intermediate levels, and those immunized with Vac-F or RSV had histopathology scores at control levels. Ad-F immunized animals had elevated scores of bronchiolar but not alveolar histopathology; however, this finding was not reproducible. Passive transfer of pooled immune sera from animals infected with RSV or Vac-F and Vac-G was highly protective, whereas pooled sera from animals immunized with Bac-FG failed to protect the lungs against RSV challenge. Increased pulmonary histopathology was not observed in the passively immunized animals following RSV challenge, suggesting that the histopathology was mediated by RSV-specific T cells. These data indicate that subunit F glycoprotein or chimeric FG vaccines share with FI-RSV the properties of (i) induction of F antibodies with low neutralizing activity and (ii) enhancement of pulmonary histopathology during subsequent RSV infection. These observations confirm the need for caution in studies involving the administration of RSV subunit vaccines to seronegative humans.


Subject(s)
Antigens, Viral/immunology , HN Protein , Lung/pathology , Respiratory Syncytial Viruses/immunology , Respirovirus Infections/pathology , Vaccinia virus/immunology , Viral Fusion Proteins/immunology , Viral Proteins , Viral Vaccines/immunology , Animals , Antibodies/analysis , Antibodies, Viral/analysis , Female , Immunization , Immunization, Passive , Male , Sigmodontinae , Vaccines, Synthetic/immunology , Viral Envelope Proteins
19.
J Virol ; 65(3): 1634-7, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1995956

ABSTRACT

The ability of recombinant vaccinia viruses that separately encoded 9 of the 10 known respiratory syncytial virus (RSV) proteins to induce resistance to RSV challenge was studied in BALB/c mice. Resistance was examined at two intervals following vaccination to examine early (day 9) as well as late (day 28) immunity. BALB/c mice were inoculated simultaneously by the intranasal and intraperitoneal routes with a recombinant vaccinia virus encoding one of the following RSV proteins: F, G, N, P, SH, M, 1B, 1C, or M2 (22K). A parainfluenza virus type 3 HN protein recombinant (Vac-HN) served as a negative control. One half of the mice were challenged with RSV intranasally on day 9, and the remaining animals were challenged on day 28 postvaccination. Mice previously immunized by infection with RSV, Vac-F, or Vac-G were completely or almost completely resistant to RSV challenge on both days. In contrast, immunization with Vac-HN, -P, -SH, -M, -1B, or -1C did not induce detectable resistance to RSV challenge. Mice previously infected with Vac-M2 or Vac-N exhibited significant but not complete resistance on day 9. However, in both cases resistance had largely waned by day 28 and was detectable only in mice immunized with Vac-M2. These results demonstrate that F and G proteins expressed by recombinant vaccinia viruses are the most effective RSV protective antigens. This study also suggests that RSV vaccines need only contain the F and G glycoproteins, because the immunity conferred by the other proteins is less effective and appears to wane rapidly with time.


Subject(s)
Respiratory Syncytial Viruses/immunology , Respirovirus Infections/immunology , Viral Proteins/immunology , Viral Vaccines , Animals , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred BALB C , Neutralization Tests , Recombination, Genetic , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/isolation & purification , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL